1
|
Jasim AH, Abu-Raghif AR, Hussein ZA. Protective Effects of Niclosamide Ethanolamine Against Testosterone-Induced Benign Prostatic Hyperplasia in Rats. Drug Res (Stuttg) 2025. [PMID: 40294597 DOI: 10.1055/a-2576-4153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Benign prostatic hyperplasia is a common urological condition in aging men. The anthelmintic agent niclosamide ethanolamide exhibits a wide range of pharmacological activities. This study aimed to evaluate the protective effect of niclosamide ethanolamide in testosterone propionate-induced benign prostatic hyperplasia in rats along with elucidating the probable mechanism of action by investigating the influence on PPAR-γ and Wnt/β-catenin. 40 male Wistar rats were divided randomly into 4 groups. The healthy (control) group, received daily oral and subcutaneous administration of the vehicle. The Induced (TP) group, received only a daily dose of testosterone propionate 3 mg/kg, SC for 28 days. The treated groups (TP+FIN) and (TP+NE), received a concomitant administration of a daily dose of testosterone propionate along with finasteride 5 mg/kg/day and niclosamide ethanolamide 50 mg/kg/day respectively through oral gavage. Animals were euthanized on day 30 of the experiment and prostate tissue samples were collected to evaluate prostate index, prostate hyperplastic markers by ELISA, and gene expression by RT-qPCR. Results revealed that niclosamide ethanolamide significantly reduced prostate index compared to the induced (TP) group (P<0.0001). The agent nearly normalized BPH markers including 5α-reductase type-2 enzyme, dihydrotestosterone, and PCNA compared to the induced (TP) group (P<0.0001). The agent reduced the tissue level of β-catenin while elevating PPAR-γ to control levels (P<0.05). The current study revealed that NE can help prevent BPH in rats by upregulating the PPAR-γ receptor and inhibiting the Wnt pathway.
Collapse
Affiliation(s)
- Ali Hussein Jasim
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | | - Zeena Ayad Hussein
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
2
|
Elfar G, Aning O, Ngai T, Yeo P, Chan J, Sim S, Goh L, Yuan J, Phua C, Yeo J, Mak S, Goh B, Chow PH, Tam W, Ho Y, Cheok C. p53-dependent crosstalk between DNA replication integrity and redox metabolism mediated through a NRF2-PARP1 axis. Nucleic Acids Res 2024; 52:12351-12377. [PMID: 39315696 PMCID: PMC11551750 DOI: 10.1093/nar/gkae811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Mechanisms underlying p53-mediated protection of the replicating genome remain elusive, despite the quintessential role of p53 in maintaining genomic stability. Here, we uncover an unexpected function of p53 in curbing replication stress by limiting PARP1 activity and preventing the unscheduled degradation of deprotected stalled forks. We searched for p53-dependent factors and elucidated RRM2B as a prime factor. Deficiency in p53/RRM2B results in the activation of an NRF2 antioxidant transcriptional program, with a concomitant elevation in basal PARylation in cells. Dissecting the consequences of p53/RRM2B loss revealed a crosstalk between redox metabolism and genome integrity that is negotiated through a hitherto undescribed NRF2-PARP1 axis, and pinpoint G6PD as a primary oxidative stress-induced NRF2 target and activator of basal PARylation. This study elucidates how loss of p53 could be destabilizing for the replicating genome and, importantly, describes an unanticipated crosstalk between redox metabolism, PARP1 and p53 tumor suppressor pathway that is broadly relevant in cancers and can be leveraged therapeutically.
Collapse
Affiliation(s)
- Gamal Ahmed Elfar
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Obed Aning
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Tsz Wai Ngai
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Pearlyn Yeo
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Joel Wai Kit Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shang Hong Sim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Leonard Goh
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Ju Yuan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Cheryl Zi Jin Phua
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Joanna Zhen Zhen Yeo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Brian Kim Poh Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
- Surgery Academic ClinicalProgramme, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chit Fang Cheok
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| |
Collapse
|
3
|
Wiggins R, Woo J, Mito S. Optimizing Niclosamide for Cancer Therapy: Improving Bioavailability via Structural Modification and Nanotechnology. Cancers (Basel) 2024; 16:3548. [PMID: 39456642 PMCID: PMC11506536 DOI: 10.3390/cancers16203548] [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: 09/15/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Inhibition of multiple cancer-related pathways has made niclosamide a promising candidate for the treatment of various cancers. However, its clinical application has been significantly limited by poor bioavailability. This review will discuss current findings on improving niclosamide bioavailability through modification of its chemical structure and utilization of novel nanotechnologies, like electrospraying and supercritical fluids, to improve drug delivery. For example, niclosamide derivatives, such as o-alkylamino-tethered niclosamide derivates, niclosamide ethanolamine salt, and niclosamide piperazine salt, have demonstrated increased water solubility without compromising anticancer activity in vitro. Additionally, this review briefly discusses recent findings on the first pass metabolism of niclosamide in vivo, the role of cytochrome P450-mediated hydroxylation, UDP-glucuronosyltransferase mediated glucuronidation, and how enzymatic inhibition could enhance niclosamide bioavailability. Ultimately, there is a need for researchers to synthesize, evaluate, and improve upon niclosamide derivatives while experimenting with the employment of nanotechnologies, such as targeted delivery and nanoparticle modification, as a way to improve drug administration. Researchers should strive to improve drug-target accuracy, its therapeutic index, and increase the drug's efficacy as an anti-neoplastic agent.
Collapse
Affiliation(s)
| | | | - Shizue Mito
- Department of Medical Education, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA; (R.W.); (J.W.)
| |
Collapse
|
4
|
Maslah N, Rety S, Bonnamy M, Aguinaga L, Huynh T, Parietti V, Giraudier S, Fenaux P, Cassinat B. Niclosamide combined to Azacitidine to target TP53-mutated MDS/AML cells. Leukemia 2024; 38:1630-1633. [PMID: 38777834 PMCID: PMC11216995 DOI: 10.1038/s41375-024-02281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Nabih Maslah
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France.
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France.
| | - Salome Rety
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
| | - Melina Bonnamy
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
| | - Lorea Aguinaga
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France
| | - Tony Huynh
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France
| | - Veronique Parietti
- Université Paris Cité, INSERM/CNRS, US53/UAR2030, Institut de Recherche Saint-Louis, Paris, France
| | - Stephane Giraudier
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France
| | - Pierre Fenaux
- APHP, Hopital Saint-Louis, Service d'Hematologie Seniors, Universite Paris Cite, Paris, France.
| | - Bruno Cassinat
- INSERM UMR 1131, Universite Paris Cite, IRSL, Paris, France
- APHP, Hopital Saint-Louis, Laboratoire de Biologie Cellulaire, Universite Paris Cite, Paris, France
| |
Collapse
|
5
|
Kaur R, Suresh PK. Chemoresistance Mechanisms in Non-Small Cell Lung Cancer-Opportunities for Drug Repurposing. Appl Biochem Biotechnol 2024; 196:4382-4438. [PMID: 37721630 DOI: 10.1007/s12010-023-04595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 09/19/2023]
Abstract
Globally, lung cancer contributes significantly to the public health burden-associated mortality. As this form of cancer is insidious in nature, there is an inevitable diagnostic delay leading to chronic tumor development. Non-small cell lung cancer (NSCLC) constitutes 80-85% of all lung cancer cases, making this neoplasia form a prevalent subset of lung carcinoma. One of the most vital aspects for proper diagnosis, prognosis, and adequate therapy is the precise classification of non-small cell lung cancer based on biomarker expression profiling. This form of biomarker profiling has provided opportunities for improvements in patient stratification, mechanistic insights, and probable druggable targets. However, numerous patients have exhibited numerous toxic side effects, tumor relapse, and development of therapy-based chemoresistance. As a result of these exacting situations, there is a dire need for efficient and effective new cancer therapeutics. De novo drug development approach is a costly and tedious endeavor, with an increased attrition rate, attributed, in part, to toxicity-related issues. Drug repurposing, on the other hand, when combined with computer-assisted systems biology approach, provides alternatives to the discovery of new, efficacious, and safe drugs. Therefore, in this review, we focus on a comparison of the conventional therapy-based chemoresistance mechanisms with the repurposed anti-cancer drugs from three different classes-anti-parasitic, anti-depressants, and anti-psychotics for cancer treatment with a primary focus on NSCLC therapeutics. Certainly, amalgamating these novel therapeutic approaches with that of the conventional drug regimen in NSCLC-affected patients will possibly complement/synergize the existing therapeutic modalities. This approach has tremendous translational significance, since it can combat drug resistance and cytotoxicity-based side effects and provides a relatively new strategy for possible application in therapy of individuals with NSCLC.
Collapse
Affiliation(s)
- Rajdeep Kaur
- Department of Bio-Medical Sciences, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India
| | - P K Suresh
- Department of Bio-Medical Sciences, School of Biosciences and Technology, VIT University, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
6
|
Žuna K, Tyschuk T, Beikbaghban T, Sternberg F, Kreiter J, Pohl EE. The 2-oxoglutarate/malate carrier extends the family of mitochondrial carriers capable of fatty acid and 2,4-dinitrophenol-activated proton transport. Acta Physiol (Oxf) 2024; 240:e14143. [PMID: 38577966 PMCID: PMC11475482 DOI: 10.1111/apha.14143] [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: 10/19/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
AIMS Metabolic reprogramming in cancer cells has been linked to mitochondrial dysfunction. The mitochondrial 2-oxoglutarate/malate carrier (OGC) has been suggested as a potential target for preventing cancer progression. Although OGC is involved in the malate/aspartate shuttle, its exact role in cancer metabolism remains unclear. We aimed to investigate whether OGC may contribute to the alteration of mitochondrial inner membrane potential by transporting protons. METHODS The expression of OGC in mouse tissues and cancer cells was investigated by PCR and Western blot analysis. The proton transport function of recombinant murine OGC was evaluated by measuring the membrane conductance (Gm) of planar lipid bilayers. OGC-mediated substrate transport was measured in proteoliposomes using 14C-malate. RESULTS OGC increases proton Gm only in the presence of natural (long-chain fatty acids, FA) or chemical (2,4-dinitrophenol) protonophores. The increase in OGC activity directly correlates with the increase in the number of unsaturated bonds of the FA. OGC substrates and inhibitors compete with FA for the same protein binding site. Arginine 90 was identified as a critical amino acid for the binding of FA, ATP, 2-oxoglutarate, and malate, which is a first step towards understanding the OGC-mediated proton transport mechanism. CONCLUSION OGC extends the family of mitochondrial transporters with dual function: (i) metabolite transport and (ii) proton transport facilitated in the presence of protonophores. Elucidating the contribution of OGC to uncoupling may be essential for the design of targeted drugs for the treatment of cancer and other metabolic diseases.
Collapse
Affiliation(s)
- Kristina Žuna
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| | - Tatyana Tyschuk
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
- Present address:
Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVAViennaAustria
| | - Taraneh Beikbaghban
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| | - Jürgen Kreiter
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
- Present address:
Institute of Molecular and Cellular PhysiologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Elena E. Pohl
- Physiology and Biophysics, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| |
Collapse
|
7
|
Yu Y, Liu H, Yuan L, Pan M, Bei Z, Ye T, Qian Z. Niclosamide - encapsulated lipid nanoparticles for the reversal of pulmonary fibrosis. Mater Today Bio 2024; 25:100980. [PMID: 38434573 PMCID: PMC10907778 DOI: 10.1016/j.mtbio.2024.100980] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 03/05/2024] Open
Abstract
Pulmonary fibrosis (PF) is a serious and progressive fibrotic interstitial lung disease that is possibly life-threatening and that is characterized by fibroblast accumulation and collagen deposition. Nintedanib and pirfenidone are currently the only two FDA-approved oral medicines for PF. Some drugs such as antihelminthic drug niclosamide (Ncl) have shown promising therapeutic potentials for PF treatment. Unfortunately, poor aqueous solubility problems obstruct clinical application of these drugs. Herein, we prepared Ncl-encapsulated lipid nanoparticles (Ncl-Lips) for pulmonary fibrosis therapy. A mouse model of pulmonary fibrosis induced by bleomycin (BLM) was generated to assess the effects of Ncl-Lips and the mechanisms of reversing fibrosis in vivo. Moreover, cell models treated with transforming growth factor β1 (TGFβ1) were used to investigate the mechanism through which Ncl-Lips inhibit fibrosis in vitro. These findings demonstrated that Ncl-Lips could alleviate fibrosis, consequently reversing the changes in the levels of the associated marker. Moreover, the results of the tissue distribution experiment showed that Ncl-Lips had aggregated in the lung. Additionally, Ncl-Lips improved the immune microenvironment in pulmonary fibrosis induced by BLM. Furthermore, Ncl-Lips suppressed the TGFβ1-induced activation of fibroblasts and epithelial-mesenchymal transition (EMT) in epithelial cells. Based on these results, we demonstrated that Ncl-Lips is an efficient strategy for reversing pulmonary fibrosis via drug-delivery.
Collapse
Affiliation(s)
- Yan Yu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hongyao Liu
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer and Frontiers Science Center for Disease-Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liping Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhongwu Bei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tinghong Ye
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer and Frontiers Science Center for Disease-Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
8
|
Peuget S, Zhou X, Selivanova G. Translating p53-based therapies for cancer into the clinic. Nat Rev Cancer 2024; 24:192-215. [PMID: 38287107 DOI: 10.1038/s41568-023-00658-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/31/2024]
Abstract
Inactivation of the most important tumour suppressor gene TP53 occurs in most, if not all, human cancers. Loss of functional wild-type p53 is achieved via two main mechanisms: mutation of the gene leading to an absence of tumour suppressor activity and, in some cases, gain-of-oncogenic function; or inhibition of the wild-type p53 protein mediated by overexpression of its negative regulators MDM2 and MDMX. Because of its high potency as a tumour suppressor and the dependence of at least some established tumours on its inactivation, p53 appears to be a highly attractive target for the development of new anticancer drugs. However, p53 is a transcription factor and therefore has long been considered undruggable. Nevertheless, several innovative strategies have been pursued for targeting dysfunctional p53 for cancer treatment. In mutant p53-expressing tumours, the predominant strategy is to restore tumour suppressor function with compounds acting either in a generic manner or otherwise selective for one or a few specific p53 mutations. In addition, approaches to deplete mutant p53 or to target vulnerabilities created by mutant p53 expression are currently under development. In wild-type p53 tumours, the major approach is to protect p53 from the actions of MDM2 and MDMX by targeting these negative regulators with inhibitors. Although the results of at least some clinical trials of MDM2 inhibitors and mutant p53-restoring compounds are promising, none of the agents has yet been approved by the FDA. Alternative strategies, based on a better understanding of p53 biology, the mechanisms of action of compounds and treatment regimens as well as the development of new technologies are gaining interest, such as proteolysis-targeting chimeras for MDM2 degradation. Other approaches are taking advantage of the progress made in immune-based therapies for cancer. In this Review, we present these ongoing clinical trials and emerging approaches to re-evaluate the current state of knowledge of p53-based therapies for cancer.
Collapse
Affiliation(s)
- Sylvain Peuget
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaolei Zhou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Materials Science and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Galina Selivanova
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Lawrence MJ, Grayson P, Jeffrey JD, Docker MF, Garroway CJ, Wilson JM, Manzon RG, Wilkie MP, Jeffries KM. Differences in the transcriptome response in the gills of sea lamprey acutely exposed to 3-trifluoromethyl-4-nitrophenol (TFM), niclosamide or a TFM:niclosamide mixture. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 48:101122. [PMID: 37659214 DOI: 10.1016/j.cbd.2023.101122] [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: 06/08/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023]
Abstract
Sea lamprey (Petromyzon marinus) control in the Laurentian Great Lakes of North America makes use of two pesticides: 3-trifluoromethyl-4-nitrophenol (TFM) and niclosamide, which are often co-applied. Sea lamprey appear to be vulnerable to these agents resulting from a lack of detoxification responses with evidence suggesting that lampricide mixtures produce a synergistic effect. However, there is a lack of information pertaining to the physiological responses of sea lamprey to niclosamide and TFM:niclosamide mixtures. Here, we characterized the transcriptomic responses of the sea lamprey to TFM, niclosamide, and a TFM:niclosamide (1.5 %) mixture in the gill. Along with a control, larval sea lamprey were exposed to each treatment for 6 h, after which gill tissues were extracted for measuring whole-transcriptome responses using RNA sequencing. Differential gene expression patterns were summarized, which included identifying the broad roles of genes and common expression patterns among the treatments. While niclosamide treatment resulted in no differentially expressed genes, TFM- and mixture-treated fish had several differentially expressed genes that were associated with the cell cycle, DNA damage, metabolism, immune function, and detoxification. However, there was no common differential expression among treatments. For the first time, we characterized the transcriptomic response of sea lamprey to niclosamide and a TFM:niclosamide mixture and identified that these agents impact mRNA transcript abundance of genes associated with the cell cycle and cellular death, and immune function, which are likely mediated through mitochondrial dysregulation. These results may help to inform the production of more targeted and effective lampricides in sea lamprey control efforts.
Collapse
Affiliation(s)
- M J Lawrence
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - P Grayson
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - J D Jeffrey
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - M F Docker
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - C J Garroway
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - J M Wilson
- Department of Biology, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - R G Manzon
- Department of Biology, University of Regina, Regina, SK S4S 0A2, Canada
| | - M P Wilkie
- Department of Biology, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - K M Jeffries
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
10
|
Melnik BC. Acne Transcriptomics: Fundamentals of Acne Pathogenesis and Isotretinoin Treatment. Cells 2023; 12:2600. [PMID: 37998335 PMCID: PMC10670572 DOI: 10.3390/cells12222600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
This review on acne transcriptomics allows for deeper insights into the pathogenesis of acne and isotretinoin's mode of action. Puberty-induced insulin-like growth factor 1 (IGF-1), insulin and androgen signaling activate the kinase AKT and mechanistic target of rapamycin complex 1 (mTORC1). A Western diet (hyperglycemic carbohydrates and milk/dairy products) also co-stimulates AKT/mTORC1 signaling. The AKT-mediated phosphorylation of nuclear FoxO1 and FoxO3 results in their extrusion into the cytoplasm, a critical switch which enhances the transactivation of lipogenic and proinflammatory transcription factors, including androgen receptor (AR), sterol regulatory element-binding transcription factor 1 (SREBF1), peroxisome proliferator-activated receptor γ (PPARγ) and signal transducer and activator of transcription 3 (STAT3), but reduces the FoxO1-dependent expression of GATA binding protein 6 (GATA6), the key transcription factor for infundibular keratinocyte homeostasis. The AKT-mediated phosphorylation of the p53-binding protein MDM2 promotes the degradation of p53. In contrast, isotretinoin enhances the expression of p53, FoxO1 and FoxO3 in the sebaceous glands of acne patients. The overexpression of these proapoptotic transcription factors explains isotretinoin's desirable sebum-suppressive effect via the induction of sebocyte apoptosis and the depletion of BLIMP1(+) sebocyte progenitor cells; it also explains its adverse effects, including teratogenicity (neural crest cell apoptosis), a reduced ovarian reserve (granulosa cell apoptosis), the risk of depression (the apoptosis of hypothalamic neurons), VLDL hyperlipidemia, intracranial hypertension and dry skin.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, 49069 Osnabrück, Germany
| |
Collapse
|
11
|
Huang Z, Chen S, Yu L. Predicting new drug indications based on double variational autoencoders. Comput Biol Med 2023; 164:107261. [PMID: 37487382 DOI: 10.1016/j.compbiomed.2023.107261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/29/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
Experimental drug development is costly, complex, and time-consuming, and the number of drugs that have been put into application treatment is small. The identification of drug-disease correlations can provide important information for drug discovery and drug repurposing. Computational drug repurposing is an important and effective method that can be used to determine novel treatments for diseases. In recent years, an increasing number of large databases have been utilized for biological data research, particularly in the fields of drugs and diseases. Consequently, researchers have begun to explore the application of deep neural networks in biological data development. One particularly promising method for unsupervised learning is the deep generative model, with the variational autoencoder (VAE) being among the mainstream models. Here, we propose a drug indication prediction algorithm called DIDVAE (predicting new drug indications based on double variational autoencoders), which generates new data by learning the latent variable distribution of known data to achieve the goal of predicting drug-disease associations. In the experiment, we compared the DIDVAE algorithm with the BBNR, DrugNet, MBiRW and DRRS algorithms on a unified dataset. The comprehensive experimental results show that, compared with these prediction algorithms, the DIDVAE algorithm provides an overall improved prediction. In addition, further analysis and verification of the predicted unknown drug-disease association also proved the practicality of the method.
Collapse
Affiliation(s)
- Zhaoyang Huang
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Shengjian Chen
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Liang Yu
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, Shaanxi, China.
| |
Collapse
|
12
|
Yang L, Fan X, Zhou C, Wang Z, Cui Z, Wu X, Xu Z, Yang J, Zhang X. Construction and validation of a novel ferroptosis-related prognostic signature for lung adenocarcinoma. Transl Lung Cancer Res 2023; 12:1766-1781. [PMID: 37691861 PMCID: PMC10483076 DOI: 10.21037/tlcr-23-351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023]
Abstract
Background Lung cancer has the highest prevalence and mortality of all cancers, and lung adenocarcinoma (LUAD) occupies the largest proportion of lung cancers. Herein, this study is aimed at constructing a ferroptosis-related prognostic signature for LUAD and conducting functional analysis based on the signature, highlighting the importance of ferroptosis in LUAD. Methods We employed RNA-sequencing (RNA-seq) and clinical data from The Cancer Genome Atlas (TCGA) database. Univariate Cox regression, Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis were conducted to build the ferroptosis-related genes (FRGs) prognostic signature. The efficacy of this FRG signature was further analyzed with Kaplan-Meier (KM) plot, multivariate Cox regression, and the receiver operating characteristic (ROC) curves. Enrichment analysis was used to evaluate key pathways. The expression of immunomodulators, immune infiltration status, and drug sensitivity correlation were explored to predict the response to various therapies. The expression of FRGs was validated in LUAD samples with western blot (WB) and immunohistochemistry (IHC) staining. Cell viability assay and lipid peroxidation detection were measured after small interfering RNA (siRNA) knockdown of two FRGs in lung cancer cell lines. Results A seven-gene signature was constructed and used to divide LUAD patients into high- and low-risk groups. High-risk patients were notably related to shorter overall survival (OS), and multivariate Cox regression demonstrated that our signature was an independent predictor of OS. ROC curve analysis presented a maximum area under the curve (AUC) value of 0.740 for the experimental cohort and 0.705 for the validation cohort. The low-risk group showed higher levels of plasma cell infiltration and higher expression of programmed cell death protein 1 (PDCD1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). Ferroptosis inducers such as talazoparib or cisplatin had lower IC50 values in the high-risk group, while navitoclax (BCL-2 gene family inhibition and apoptosis inducer) had higher IC50 values in the high-risk group. Additionally, peroxiredoxin-6 (PRDX6) and acyl-CoA synthetase long chain family member 3 (ACSL3) were upregulated in LUAD tissues. Lipid peroxide assay showed that silencing PRDX6 or ACSL3 promoted lipid peroxidation and ferroptosis in lung cancer cells. Conclusions Our novel ferroptosis-related signature shows potential clinical and functional importance in LUAD patients, and further research on ferroptosis as a therapeutic target in LUAD is warranted.
Collapse
Affiliation(s)
- Li Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xinxin Fan
- Department of Hematology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Chao Zhou
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Ziqi Wang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Zelong Cui
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuan Wu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Zhiwei Xu
- Clinical Research Service Centre, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jia Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
13
|
Zhou Y, Cui G, Xu H, Chun J, Yang D, Zhang Z, Yang L, Wang J, Wan M, Calvisi DF, Lin S, Chen X, Wang H. Loss of TP53 cooperates with c-MET overexpression to drive hepatocarcinogenesis. Cell Death Dis 2023; 14:476. [PMID: 37500626 PMCID: PMC10374654 DOI: 10.1038/s41419-023-05958-y] [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: 10/17/2022] [Revised: 06/13/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is a deadly malignancy with high genetic heterogeneity. TP53 mutation and c-MET activation are frequent events in human HCCs. Here, we discovered that the simultaneous mutations in TP53 and activation of c-MET occur in ~20% of human HCCs, and these patients show a poor prognosis. Importantly, we found that concomitant deletion of Trp53 and overexpression of c-MET (c-MET/sgp53) in the mouse liver led to HCC formation in vivo. Consistent with human HCCs, RNAseq showed that c-MET/sgp53 mouse HCCs were characterized by activated c-MET and Ras/MAPK cascades and increased tumor cell proliferation. Subsequently, a stably passaged cell line derived from a c-MET/sgp53 HCC and corresponding subcutaneous xenografts were generated. Also, in silico analysis suggested that the MEK inhibitor trametinib has a higher inhibition score in TP53 null human HCC cell lines, which was validated experimentally. We consistently found that trametinib effectively inhibited the growth of c-MET/sgp53 HCC cells and xenografts, supporting the possible usefulness of this drug for treating human HCCs with TP53-null mutations. Altogether, our study demonstrates that loss of TP53 cooperates with c-MET to drive hepatocarcinogenesis in vivo. The c-MET/sgp53 mouse model and derived HCC cell lines represent novel and useful preclinical tools to study hepatocarcinogenesis in the TP53 null background.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
| | - Guofei Cui
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Hongwei Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Joanne Chun
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Doris Yang
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Zheng Zhang
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Yang
- Beijing University of Chinese Medicine, Beijing, China
| | - Jingxiao Wang
- School of Life Sciences, Beijing, University of Chinese Medicine, Beijing, China
| | - Meijuan Wan
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, 93053, Germany
| | - Shumei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, San Francisco, CA, USA.
- Liver Cancer Laboratory, University of Hawaii Cancer Center, Honolulu, HI, USA.
| | - Haichuan Wang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Wanas H, Elbadawy HM, Almikhlafi MA, Hamoud AE, Ali EN, Galal AM. Combination of Niclosamide and Pirfenidone Alleviates Pulmonary Fibrosis by Inhibiting Oxidative Stress and MAPK/Nf-κB and STATs Regulated Genes. Pharmaceuticals (Basel) 2023; 16:ph16050697. [PMID: 37242480 DOI: 10.3390/ph16050697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
The pathogenesis of pulmonary fibrosis (PF) is extremely complex and involves numerous intersecting pathways. The successful management of PF may require combining multiple agents. There is a growing body of evidence that suggests the potential benefits of niclosamide (NCL), an FDA-approved anthelminthic drug, in targeting different fibrogenesis molecules. This study aimed at investigating the anti-fibrotic potential of NCL alone and in combination with pirfenidone (PRF), an approved drug for PF, in a bleomycin (BLM) induced PF experimental model. PF was induced in rats by intratracheal BLM administration. The effect of NCL and PRF individually and in combination on different histological and biochemical parameters of fibrosis was investigated. Results revealed that NCL and PRF individually and in combination alleviated the histopathological changes, extracellular matrix deposition and myofibroblastic activation induced by BLM. NCL and PRF either individually or in combination inhibited the oxidative stress and subsequent pathways. They modulated the process of fibrogenesis by inhibiting MAPK/NF-κB and downstream cytokines. They inhibited STATs and downstream survival-related genes including BCL-2, VEGF, HIF-α and IL-6. Combining both drugs showed significant improvement in the tested markers in comparison to the monotherapy. NCL, therefore, has a potential synergistic effect with PRF in reducing the severity of PF.
Collapse
Affiliation(s)
- Hanaa Wanas
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah 41477, Saudi Arabia
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11956, Egypt
| | - Hossein M Elbadawy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah 41477, Saudi Arabia
| | - Mohannad A Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah 41477, Saudi Arabia
| | - Amany E Hamoud
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11956, Egypt
| | - Eid N Ali
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11956, Egypt
- Department of Anatomy, Faculty of Medicine, Taibah University, Madinah 41477, Saudi Arabia
| | - Amr M Galal
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11956, Egypt
| |
Collapse
|
15
|
Rahmé R, Braun T, Manfredi JJ, Fenaux P. TP53 Alterations in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Biomedicines 2023; 11:biomedicines11041152. [PMID: 37189770 DOI: 10.3390/biomedicines11041152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
TP53 mutations are less frequent in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) than in solid tumors, except in secondary and therapy-related MDS/AMLs, and in cases with complex monosomal karyotype. As in solid tumors, missense mutations predominate, with the same hotspot mutated codons (particularly codons 175, 248, 273). As TP53-mutated MDS/AMLs are generally associated with complex chromosomal abnormalities, it is not always clear when TP53 mutations occur in the pathophysiological process. It is also uncertain in these MDS/AML cases, which often have inactivation of both TP53 alleles, if the missense mutation is only deleterious through the absence of a functional p53 protein, or through a potential dominant-negative effect, or finally a gain-of-function effect of mutant p53, as demonstrated in some solid tumors. Understanding when TP53 mutations occur in the disease course and how they are deleterious would help to design new treatments for those patients who generally show poor response to all therapeutic approaches.
Collapse
Affiliation(s)
- Ramy Rahmé
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institut de Recherche Saint Louis (IRSL), INSERM U1131, Université Paris Cité, 75010 Paris, France
- Ecole Doctorale Hématologie-Oncogenèse-Biothérapies, Université Paris Cité, 75010 Paris, France
- Clinical Hematology Department, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Sorbonne Paris Nord, 93000 Bobigny, France
| | - Thorsten Braun
- Clinical Hematology Department, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Sorbonne Paris Nord, 93000 Bobigny, France
| | - James J Manfredi
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pierre Fenaux
- Senior Hematology Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, 75010 Paris, France
| |
Collapse
|
16
|
Shi W, Sethi G. Long noncoding RNAs induced control of ferroptosis: Implications in cancer progression and treatment. J Cell Physiol 2023; 238:880-895. [PMID: 36924057 DOI: 10.1002/jcp.30992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
A novel kind of nonapoptotic, iron-dependent cell death brought on by lipid peroxidation is known as ferroptosis. Numerous pathological processes, including neurotoxicity, neurological disorders, ischemia-reperfusion damage, and particularly cancer, have been demonstrated to be influenced by changes in the ferroptosis-regulating network. Recent studies have established the critical roles that ferroptosis can play in cancer development and the evolution of resistance to standard chemoradiotherapy, thus suggesting that ferroptosis may be a feasible therapeutic strategy for cancer management. Gene expression may be regulated at the transcriptional and posttranscriptional levels by long noncoding RNAs (lncRNAs). They have been implicated in tumorigenesis. Some lncRNAs participate in the biological process of ferroptosis, which represents an exciting alternative to regulate ferroptosis as a means of cancer therapy. Even though there is evidence that lncRNAs have a mechanistic role in the ferroptosis of cancer cells, research on the mechanism and potential treatments for these lncRNAs is still lacking. We elucidate the potential mechanisms by which lncRNAs modulate ferroptosis in cancer and examine the promise and challenges of employing lncRNAs as novel therapeutic targets in cancer.
Collapse
Affiliation(s)
- Wei Shi
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Jiang H, Greathouse RL, Tiche SJ, Zhao M, He B, Li Y, Li AM, Forgo B, Yip M, Li A, Shih M, Banuelos S, Zhou MN, Gruber JJ, Rankin EB, Hu Z, Shimada H, Chiu B, Ye J. Mitochondrial Uncoupling Induces Epigenome Remodeling and Promotes Differentiation in Neuroblastoma. Cancer Res 2023; 83:181-194. [PMID: 36318118 PMCID: PMC9851961 DOI: 10.1158/0008-5472.can-22-1029] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/23/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022]
Abstract
The Warburg effect is the major metabolic hallmark of cancer. According to Warburg himself, the consequence of the Warburg effect is cell dedifferentiation. Therefore, reversing the Warburg effect might be an approach to restore cell differentiation in cancer. In this study, we used a mitochondrial uncoupler, niclosamide ethanolamine (NEN), to activate mitochondrial respiration, which induced neural differentiation in neuroblastoma cells. NEN treatment increased the NAD+/NADH and pyruvate/lactate ratios and also the α-ketoglutarate/2-hydroxyglutarate (2-HG) ratio. Consequently, NEN treatment induced promoter CpG island demethylation and epigenetic landscape remodeling, activating the neural differentiation program. In addition, NEN treatment upregulated p53 but downregulated N-Myc and β-catenin signaling in neuroblastoma cells. Importantly, even under hypoxia, NEN treatment remained effective in inhibiting 2-HG generation, promoting DNA demethylation, and suppressing hypoxia-inducible factor signaling. Dietary NEN intervention reduced tumor growth rate, 2-HG levels, and expression of N-Myc and β-catenin in tumors in an orthotopic neuroblastoma mouse model. Integrative analysis indicated that NEN treatment upregulated favorable prognosis genes and downregulated unfavorable prognosis genes, which were defined using multiple neuroblastoma patient datasets. Altogether, these results suggest that mitochondrial uncoupling is an effective metabolic and epigenetic therapy for reversing the Warburg effect and inducing differentiation in neuroblastoma. SIGNIFICANCE Targeting cancer metabolism using the mitochondrial uncoupler niclosamide ethanolamine leads to methylome reprogramming and differentiation in neuroblastoma, providing a therapeutic opportunity to reverse the Warburg effect and suppress tumor growth. See related commentary by Byrne and Bell, p.167.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Rachel L. Greathouse
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sarah Jane Tiche
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Man Zhao
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Bo He
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Yang Li
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA,Cancer Biology Program, Stanford University School of Medicine
| | - Balint Forgo
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Michaela Yip
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Allison Li
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Moriah Shih
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Selene Banuelos
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Meng-Ning Zhou
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Joshua J. Gruber
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center. Dallas, TX 75235, USA
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA
| | - Zhen Hu
- Olivia Consulting Service, Redwood City, CA 94063, USA
| | - Hiroyuki Shimada
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Bill Chiu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA,Stanford Cancer Institute, Stanford University School of Medicine. Stanford, CA 94305, US
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA 94305, USA,Cancer Biology Program, Stanford University School of Medicine,Stanford Cancer Institute, Stanford University School of Medicine. Stanford, CA 94305, US,Correspondence to: Jiangbin Ye (), CCSR-S, Rm.1245, 269 Campus Drive, Stanford, CA 94305, Tel: 650-724-7459
| |
Collapse
|
18
|
Byrne FL, Bell JL. Neuroblastoma Differentiation: The Untapped Potential of Mitochondrial Uncouplers. Cancer Res 2023; 83:167-169. [PMID: 36651076 DOI: 10.1158/0008-5472.can-22-3350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023]
Abstract
While the goal of most anticancer treatments is to kill cancer cells, some therapies halt cancer progression by inducing cancer cell differentiation. For example, retinoic acid induces neuroblastoma cell differentiation in vitro and is used as maintenance therapy for children with high-risk neuroblastoma. A new study by Jiang and colleagues has revealed the mitochondrial uncoupler niclosamide ethanolamine (NEN) induces neuroblastoma cell differentiation in vitro and slows neuroblastoma tumor growth in vivo. Mitochondrial uncoupler molecules alter cell metabolism by forcing cells to "burn" more nutrients, resulting in a switch from anabolic to catabolic metabolism. NEN-induced neuroblastoma cell differentiation was associated with disruption of Warburg metabolism, epigenetic remodeling, and downregulation of key oncogenic drivers of neuroblastoma development, including MYCN. NEN is currently used as an antiparasitic worm treatment and is safe to use in children but has poor pharmacokinetic properties. However, derivatives of NEN and structurally distinct uncouplers that have improved pharmacokinetic properties are in development. Results of this study ignite the idea that mitochondrial uncouplers could be used as differentiating agents and expand the pharmacotherapy toolkit to treat cancer, including neuroblastoma. See related article by Jiang et al., p. 181.
Collapse
Affiliation(s)
- F L Byrne
- School of Biotechnology & Biomolecular Sciences, Faculty of Science, University of New South Wales (UNSW) Sydney, Kensington, Australia
| | - J L Bell
- Children's Cancer Institute Australia, Randwick, Australia.,School of Women's & Children's Health, UNSW Sydney, Kensington, Australia
| |
Collapse
|
19
|
Kauerová T, Pérez-Pérez MJ, Kollar P. Salicylanilides and Their Anticancer Properties. Int J Mol Sci 2023; 24:ijms24021728. [PMID: 36675241 PMCID: PMC9861143 DOI: 10.3390/ijms24021728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Salicylanilides are pharmacologically active compounds with a wide spectrum of biological effects. Halogenated salicylanilides, which have been used for decades in human and veterinary medicine as anthelmintics, have recently emerged as candidates for drug repurposing in oncology. The most prominent example of salicylanilide anthelmintic, that is intensively studied for its potential anticancer properties, is niclosamide. Nevertheless, recent studies have discovered extensive anticancer potential in a number of other salicylanilides. This potential of their anticancer action is mediated most likely by diverse mechanisms of action such as uncoupling of oxidative phosphorylation, inhibition of protein tyrosine kinase epidermal growth factor receptor, modulation of different signaling pathways as Wnt/β-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways or induction of B-Raf V600E inhibition. Here we provide a comprehensive overview of the current knowledge about the proposed mechanisms of action of anticancer activity of salicylanilides based on preclinical in vitro and in vivo studies, or structural requirements for such an activity.
Collapse
Affiliation(s)
- Tereza Kauerová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | | | - Peter Kollar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
- Correspondence: ; Tel.: +420-541-562-892
| |
Collapse
|
20
|
Jiang H, Li AM, Ye J. The magic bullet: Niclosamide. Front Oncol 2022; 12:1004978. [PMID: 36479072 PMCID: PMC9720275 DOI: 10.3389/fonc.2022.1004978] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/12/2022] [Indexed: 08/27/2023] Open
Abstract
The term 'magic bullet' is a scientific concept proposed by the German Nobel laureate Paul Ehrlich in 1907, describing a medicine that could specifically and efficiently target a disease without harming the body. Oncologists have been looking for a magic bullet for cancer therapy ever since. However, the current therapies for cancers-including chemotherapy, radiation therapy, hormone therapy, and targeted therapy-pose either pan-cytotoxicity or only single-target efficacy, precluding their ability to function as a magic bullet. Intriguingly, niclosamide, an FDA-approved drug for treating tapeworm infections with an excellent safety profile, displays broad anti-cancer activity in a variety of contexts. In particular, niclosamide inhibits multiple oncogenic pathways such as Wnt/β-catenin, Ras, Stat3, Notch, E2F-Myc, NF-κB, and mTOR and activates tumor suppressor signaling pathways such as p53, PP2A, and AMPK. Moreover, niclosamide potentially improves immunotherapy by modulating pathways such as PD-1/PDL-1. We recently discovered that niclosamide ethanolamine (NEN) reprograms cellular metabolism through its uncoupler function, consequently remodeling the cellular epigenetic landscape to promote differentiation. Inspired by the promising results from the pre-clinical studies, several clinical trials are ongoing to assess the therapeutic effect of niclosamide in cancer patients. This current review summarizes the functions, mechanism of action, and potential applications of niclosamide in cancer therapy as a magic bullet.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
21
|
Wotring JW, McCarty SM, Shafiq K, Zhang CJ, Nguyen T, Meyer SR, Fursmidt R, Mirabelli C, Clasby MC, Wobus CE, O’Meara MJ, Sexton JZ. In Vitro Evaluation and Mitigation of Niclosamide's Liabilities as a COVID-19 Treatment. Vaccines (Basel) 2022; 10:1284. [PMID: 36016172 PMCID: PMC9412300 DOI: 10.3390/vaccines10081284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Niclosamide, an FDA-approved oral anthelmintic drug, has broad biological activity including anticancer, antibacterial, and antiviral properties. Niclosamide has also been identified as a potent inhibitor of SARS-CoV-2 infection in vitro, generating interest in its use for the treatment or prevention of COVID-19. Unfortunately, there are several potential issues with using niclosamide for COVID-19, including low bioavailability, significant polypharmacology, high cellular toxicity, and unknown efficacy against emerging SARS-CoV-2 variants of concern. In this study, we used high-content imaging-based immunofluorescence assays in two different cell models to assess these limitations and evaluate the potential for using niclosamide as a COVID-19 antiviral. We show that despite promising preliminary reports, the antiviral efficacy of niclosamide overlaps with its cytotoxicity giving it a poor in vitro selectivity index for anti-SARS-CoV-2 inhibition. We also show that niclosamide has significantly variable potency against the different SARS-CoV-2 variants of concern and is most potent against variants with enhanced cell-to-cell spread including the B.1.1.7 (alpha) variant. Finally, we report the activity of 33 niclosamide analogs, several of which have reduced cytotoxicity and increased potency relative to niclosamide. A preliminary structure-activity relationship analysis reveals dependence on a protonophore for antiviral efficacy, which implicates nonspecific endolysosomal neutralization as a dominant mechanism of action. Further single-cell morphological profiling suggests niclosamide also inhibits viral entry and cell-to-cell spread by syncytia. Altogether, our results suggest that niclosamide is not an ideal candidate for the treatment of COVID-19, but that there is potential for developing improved analogs with higher clinical translational potential in the future.
Collapse
Affiliation(s)
- Jesse W. Wotring
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sean M. McCarty
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Khadija Shafiq
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Theophilus Nguyen
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109, USA
| | - Sophia R. Meyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Reid Fursmidt
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109, USA
| | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Martin C. Clasby
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109, USA
- U-M Center for Drug Repurposing, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
22
|
Mito S, Cheng B, Garcia BA, Gonzalez D, Ooi XY, Ruiz TC, Elisarraras FX, Tsin A, Chew SA, Arriaga MA. SAR study of niclosamide derivatives in the human glioblastoma U-87 MG cells. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02907-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
23
|
Weiss A, Delavenne E, Matias C, Lagler H, Simon D, Li P, Hansen JU, Dos Santos TP, Jana B, Priemel P, Bangert C, Bauer M, Eberl S, Nussbaumer-Pröll A, Anne Österreicher Z, Matzneller P, Quint T, Weber M, Nielsen HM, Rades T, Johansen HK, Westh H, Kim W, Mylonakis E, Friis C, Guardabassi L, Pace J, Lundberg CV, M'Zali F, Butty P, Sørensen N, Nielsen HB, Toft-Kehler R, Guttman-Yassky E, Stingl G, Zeitlinger M, Sommer M. Topical niclosamide (ATx201) reduces Staphylococcus aureus colonization and increases Shannon diversity of the skin microbiome in atopic dermatitis patients in a randomized, double-blind, placebo-controlled Phase 2 trial. Clin Transl Med 2022; 12:e790. [PMID: 35522900 PMCID: PMC9076020 DOI: 10.1002/ctm2.790] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
Background In patients with atopic dermatitis (AD), Staphylococcus aureus frequently colonizes lesions and is hypothesized to be linked to disease severity and progression. Treatments that reduce S. aureus colonization without significantly affecting the skin commensal microbiota are needed. Methods and findings In this study, we tested ATx201 (niclosamide), a small molecule, on its efficacy to reduce S. aureus and propensity to evolve resistance in vitro. Various cutaneous formulations were then tested in a superficial skin infection model. Finally, a Phase 2 randomized, double‐blind and placebo‐controlled trial was performed to investigate the impact of ATx201 OINTMENT 2% on S. aureus colonization and skin microbiome composition in patients with mild‐to‐severe AD (EudraCT:2016‐003501‐33). ATx201 has a narrow minimal inhibitory concentration distribution (.125–.5 μg/ml) consistent with its mode of action – targeting the proton motive force effectively stopping cell growth. In murine models, ATx201 can effectively treat superficial skin infections of methicillin‐resistant S. aureus. In a Phase 2 trial in patients with mild‐to‐severe AD (N = 36), twice‐daily treatment with ATx201 OINTMENT 2% effectively reduces S. aureus colonization in quantitative colony forming unit (CFU) analysis (primary endpoint: 94.4% active vs. 38.9% vehicle success rate, p = .0016) and increases the Shannon diversity of the skin microbiome at day 7 significantly compared to vehicle. Conclusion These results suggest that ATx201 could become a new treatment modality as a decolonizing agent.
Collapse
Affiliation(s)
- Anne Weiss
- UNION Therapeutics, Hellerup, Denmark.,Novo Nordisk Foundation for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | | | | | - Heimo Lagler
- Department of Medicine 1, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Wien, Austria
| | | | - Ping Li
- UNION Therapeutics, Hellerup, Denmark
| | - Jon U Hansen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Teresa Pires Dos Santos
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Bimal Jana
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Petra Priemel
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | - Sabine Eberl
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | | | | | - Peter Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | - Tamara Quint
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | | | - Thomas Rades
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Helle Krogh Johansen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Wooseong Kim
- Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Christian Friis
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Luca Guardabassi
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - John Pace
- UNION Therapeutics, Hellerup, Denmark
| | | | | | | | | | | | | | | | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | - Morten Sommer
- UNION Therapeutics, Hellerup, Denmark.,Novo Nordisk Foundation for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
24
|
Mitochondrial Dysfunction Pathway Alterations Offer Potential Biomarkers and Therapeutic Targets for Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5634724. [PMID: 35498135 PMCID: PMC9045977 DOI: 10.1155/2022/5634724] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 04/02/2022] [Indexed: 11/29/2022]
Abstract
The mitochondrion is a very versatile organelle that participates in some important cancer-associated biological processes, including energy metabolism, oxidative stress, mitochondrial DNA (mtDNA) mutation, cell apoptosis, mitochondria-nuclear communication, dynamics, autophagy, calcium overload, immunity, and drug resistance in ovarian cancer. Multiomics studies have found that mitochondrial dysfunction, oxidative stress, and apoptosis signaling pathways act in human ovarian cancer, which demonstrates that mitochondria play critical roles in ovarian cancer. Many molecular targeted drugs have been developed against mitochondrial dysfunction pathways in ovarian cancer, including olive leaf extract, nilotinib, salinomycin, Sambucus nigra agglutinin, tigecycline, and eupatilin. This review article focuses on the underlying biological roles of mitochondrial dysfunction in ovarian cancer progression based on omics data, potential molecular relationship between mitochondrial dysfunction and oxidative stress, and future perspectives of promising biomarkers and therapeutic targets based on the mitochondrial dysfunction pathway for ovarian cancer.
Collapse
|
25
|
The role of ALOX15B in heat stress-induced apoptosis of porcine sertoli cells. Theriogenology 2022; 185:6-15. [DOI: 10.1016/j.theriogenology.2022.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 01/03/2023]
|
26
|
Ashrafizadeh M, Zarrabi A, Mostafavi E, Aref AR, Sethi G, Wang L, Tergaonkar V. Non-coding RNA-based regulation of inflammation. Semin Immunol 2022; 59:101606. [PMID: 35691882 DOI: 10.1016/j.smim.2022.101606] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/01/2022] [Accepted: 05/25/2022] [Indexed: 01/15/2023]
Abstract
Inflammation is a multifactorial process and various biological mechanisms and pathways participate in its development. The presence of inflammation is involved in pathogenesis of different diseases such as diabetes mellitus, cardiovascular diseases and even, cancer. Non-coding RNAs (ncRNAs) comprise large part of transcribed genome and their critical function in physiological and pathological conditions has been confirmed. The present review focuses on miRNAs, lncRNAs and circRNAs as ncRNAs and their potential functions in inflammation regulation and resolution. Pro-inflammatory and anti-inflammatory factors are regulated by miRNAs via binding to 3'-UTR or indirectly via affecting other pathways such as SIRT1 and NF-κB. LncRNAs display a similar function and they can also affect miRNAs via sponging in regulating levels of cytokines. CircRNAs mainly affect miRNAs and reduce their expression in regulating cytokine levels. Notably, exosomal ncRNAs have shown capacity in inflammation resolution. In addition to pre-clinical studies, clinical trials have examined role of ncRNAs in inflammation-mediated disease pathogenesis and cytokine regulation. The therapeutic targeting of ncRNAs using drugs and nucleic acids have been analyzed to reduce inflammation in disease therapy. Therefore, ncRNAs can serve as diagnostic, prognostic and therapeutic targets in inflammation-related diseases in pre-clinical and clinical backgrounds.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396 Istanbul, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
27
|
Beltukova DM, Belik VP, Semak BV, Semenova IV, Smolin AG, Vasyutinskii OS. Relaxation dynamics of alkyl derivatives of fluorescein MitoFluo and C 8-Fl in solutions with liposomes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 263:120145. [PMID: 34274636 DOI: 10.1016/j.saa.2021.120145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
We present results of experimental and theoretical studies of excited state dynamics in two alkyl derivatives of fluorescein, MitoFluo and C8-Fl in solutions with liposomes. The liposomes DOPC and soybeanPC + 20% Cardiolipin (Azo-Cl), modelling cellular and inner mitochondrial membranes, respectively, were used in experiments. Both types of liposomes were shown to reduce significantly the fluorescence quantum yield as compared to that of pure fluorescein derivatives in solutions, while DOPC liposomes also caused a noticeable (ca 10 nm) red shift of fluorescence maximum. The study of fluorescence polarization decay has been carried out where important fluorescence parameters: polarization anisotropy, fluorescence lifetimes, and rotational diffusion times have been determined. It was shown that the isotropic fluorescence decay of C8-Fl in liposome containing solutions was single-exponential and the anisotropic decay was double-exponential for both types of lyposomes. In the case of MitoFluo both isotropic and anisotropic fluorescence decays were fitted satisfactory only with double-exponential functions. The interpretation of the experimental data obtained was supported by ab initio calculations of the structure and excitation properties of MitoFluo and C8-Fl in aqueous solution. The analysis of anisotropic fluorescence decay allowed for isolation of the contributions of fluorescein derivatives free in solution from those embedded in liposomes. Also, the experimental data suggest that MitoFluo interacts with liposomes more effectively than C8-Fl. Basing on the experimental and theoretical results obtained we conclude that free C8-Fl and MitoFluo molecules in solution were mostly in their dimer forms.
Collapse
Affiliation(s)
- Dina M Beltukova
- Ioffe Institute, 26 Polytekhnicheskaya, St.Petersburg 194021, Russia
| | - Victor P Belik
- Ioffe Institute, 26 Polytekhnicheskaya, St.Petersburg 194021, Russia
| | - Bogdan V Semak
- Ioffe Institute, 26 Polytekhnicheskaya, St.Petersburg 194021, Russia
| | - Irina V Semenova
- Ioffe Institute, 26 Polytekhnicheskaya, St.Petersburg 194021, Russia.
| | - Andrey G Smolin
- Ioffe Institute, 26 Polytekhnicheskaya, St.Petersburg 194021, Russia
| | | |
Collapse
|
28
|
Egolf S, Zou J, Anderson A, Simpson CL, Aubert Y, Prouty S, Ge K, Seykora JT, Capell BC. MLL4 mediates differentiation and tumor suppression through ferroptosis. SCIENCE ADVANCES 2021; 7:eabj9141. [PMID: 34890228 PMCID: PMC8664260 DOI: 10.1126/sciadv.abj9141] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The epigenetic regulator, MLL4 (KMT2D), has been described as an essential gene in both humans and mice. In addition, it is one of the most commonly mutated genes in all of cancer biology. Here, we identify a critical role for Mll4 in the promotion of epidermal differentiation and ferroptosis, a key mechanism of tumor suppression. Mice lacking epidermal Mll4, but not the related enzyme Mll3 (Kmt2c), display features of impaired differentiation and human precancerous neoplasms, all of which progress with age. Mll4 deficiency profoundly alters epidermal gene expression and uniquely rewires the expression of key genes and markers of ferroptosis (Alox12, Alox12b, and Aloxe3). Beyond revealing a new mechanistic basis for Mll4-mediated tumor suppression, our data uncover a potentially much broader and general role for ferroptosis in the process of differentiation and skin homeostasis.
Collapse
Affiliation(s)
- Shaun Egolf
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jonathan Zou
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Amy Anderson
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Cory L. Simpson
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
| | - Yann Aubert
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Stephen Prouty
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
| | - Kai Ge
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John T. Seykora
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Brian C. Capell
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Penn Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Corresponding author.
| |
Collapse
|
29
|
Single-cell RNA-seq highlights a specific carcinoembryonic cluster in ovarian cancer. Cell Death Dis 2021; 12:1082. [PMID: 34775482 PMCID: PMC8590695 DOI: 10.1038/s41419-021-04358-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022]
Abstract
Expounding the heterogeneity for ovarian cancer (OC) with the cognition in developmental biology might be helpful to search for robust prognostic markers and effective treatments. In the present study, we employed single-cell RNA-seq with ovarian cancers, normal ovary, and embryo tissue to explore their heterogeneity. Then the differentiation process of clusters was explored; the pivotal cluster and markers were identified. Furthermore, the consensus clustering algorithm was used to explore the different clinical phenotypes in OC. At last, a prognostic model was construct and used to assess the prognosis for OCs. As a result, eight diverse clusters were identified, and the similarity existed in some clusters between embryo and tumours based on their gene expression. Meaningfully, a subtype of malignant epithelial cluster, PEG10+ EME, was associated with poor survival and was an intermediate stage of embryo to tumour. PEG10 was a CSC marker and might influence CSC self-renewal and promote cisplatin resistance via NOTCH pathway. Utilising specific gene profiles of PEG10+ EME based on public data sets, four phenotypes with different survival and clinical response to anti-PD-1/PD-L1 immunotherapy were identified. These insights allowed for the investigation of single-cell transcriptome of OCs and embryo, which advanced our current understanding of OC pathogenesis and resulted in promising therapeutic strategies.
Collapse
|
30
|
Ngai TW, Elfar GA, Yeo P, Phua N, Hor JH, Chen S, Ho YS, Cheok CF. Nitro-Deficient Niclosamide Confers Reduced Genotoxicity and Retains Mitochondrial Uncoupling Activity for Cancer Therapy. Int J Mol Sci 2021; 22:10420. [PMID: 34638761 PMCID: PMC8508655 DOI: 10.3390/ijms221910420] [Citation(s) in RCA: 2] [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: 07/16/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 11/16/2022] Open
Abstract
Niclosamide is an oral anthelmintic drug, approved for use against tapeworm infections. Recent studies suggest however that niclosamide may have broader clinical applications in cancers, spurring increased interest in the functions and mechanisms of niclosamide. Previously, we reported that niclosamide targets a metabolic vulnerability in p53-deficient tumours, providing a basis for patient stratification and personalised treatment strategies. In the present study, we functionally characterised the contribution of the aniline 4'-NO2 group on niclosamide to its cellular activities. We demonstrated that niclosamide induces genome-wide DNA damage that is mechanistically uncoupled from its antitumour effects mediated through mitochondrial uncoupling. Elimination of the nitro group in ND-Nic analogue significantly reduced γH2AX signals and DNA breaks while preserving its antitumour mechanism mediated through a calcium signalling pathway and arachidonic acid metabolism. Lipidomics profiling further revealed that ND-Nic-treated cells retained a metabolite profile characteristic of niclosamide-treated cells. Notably, quantitative scoring of drug sensitivity suggests that elimination of its nitro group enhanced the target selectivity of niclosamide against p53 deficiency. Importantly, the results also raise concern that niclosamide may impose a pleiotropic genotoxic effect, which limits its clinical efficacy and warrants further investigation into alternative drug analogues that may ameliorate any potential unwanted side effects.
Collapse
Affiliation(s)
- Tsz Wai Ngai
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
| | - Gamal Ahmed Elfar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Pearlyn Yeo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Nicholas Phua
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
| | - Jin Hui Hor
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
| | - Shuwen Chen
- Analytical Science and Technology (Metabolomics), Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore; (S.C.); (Y.S.H.)
| | - Ying Swan Ho
- Analytical Science and Technology (Metabolomics), Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore; (S.C.); (Y.S.H.)
| | - Chit Fang Cheok
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (T.W.N.); (G.A.E.); (P.Y.); (N.P.); (J.H.H.)
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| |
Collapse
|
31
|
Niclosamide Is Active In Vitro against Mycetoma Pathogens. Molecules 2021; 26:molecules26134005. [PMID: 34209118 PMCID: PMC8271592 DOI: 10.3390/molecules26134005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 11/16/2022] Open
Abstract
Redox-active drugs are the mainstay of parasite chemotherapy. To assess their repurposing potential for eumycetoma, we have tested a set of nitroheterocycles and peroxides in vitro against two isolates of Madurella mycetomatis, the main causative agent of eumycetoma in Sudan. All the tested compounds were inactive except for niclosamide, which had minimal inhibitory concentrations of around 1 µg/mL. Further tests with niclosamide and niclosamide ethanolamine demonstrated in vitro activity not only against M. mycetomatis but also against Actinomadura spp., causative agents of actinomycetoma, with minimal inhibitory concentrations below 1 µg/mL. The experimental compound MMV665807, a related salicylanilide without a nitro group, was as active as niclosamide, indicating that the antimycetomal action of niclosamide is independent of its redox chemistry (which is in agreement with the complete lack of activity in all other nitroheterocyclic drugs tested). Based on these results, we propose to further evaluate the salicylanilides, niclosamidein particular, as drug repurposing candidates for mycetoma.
Collapse
|
32
|
Huang H, He Q, Guo B, Xu X, Wu Y, Li X. Progress in Redirecting Antiparasitic Drugs for Cancer Treatment. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2747-2767. [PMID: 34188451 PMCID: PMC8235938 DOI: 10.2147/dddt.s308973] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
Drug repurposing is a feasible strategy in developing novel medications. Regarding the cancer field, scientists are continuously making efforts to redirect conventional drugs into cancer treatment. This approach aims at exploring new applications in the existing agents. Antiparasitic medications, including artemisinin derivatives (ARTs), quinine-related compounds, niclosamide, ivermectin, albendazole derivatives, nitazoxanide and pyrimethamine, have been deeply investigated and widely applied in treating various parasitic diseases for a long time. Generally, their pharmacokinetic and pharmacodynamic properties are well understood, while the side effects are roughly acceptable. Scientists noticed that some of these agents have anticancer potentials and explored the underlying mechanisms to achieve drug repurposing. Recent studies show that these agents inhibit cancer progression via multiple interesting ways, inducing ferroptosis induction, autophagy regulation, mitochondrial disturbance, immunoregulation, and metabolic disruption. In this review, we summarize the recent advancement in uncovering antiparasitic drugs' anticancer properties from the perspective of their pharmacological targets. Instead of paying attention to the previously discovered mechanisms, we focus more on newly emerging ones that are worth noticing. While most investigations are focusing on the mechanisms of their antiparasitic effect, more in vivo exploration in clinical trials in the future is necessary. Moreover, we also paid attention to what limits the clinical application of these agents. For some of these agents like ARTs and niclosamide, drug modification, novel delivery system invention, or drug combination are strongly recommended for future exploration.
Collapse
Affiliation(s)
- Haoyang Huang
- Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Qing He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| | - Binghua Guo
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Xudong Xu
- Department of Clinical Medicine, School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yinjuan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| |
Collapse
|
33
|
Wang G, Gaikwad H, McCarthy MK, Gonzalez-Juarrero M, Li Y, Armstrong M, Reisdorph N, Morrison TE, Simberg D. Lipid nanoparticle formulation of niclosamide (nano NCM) effectively inhibits SARS-CoV-2 replication in vitro. PRECISION NANOMEDICINE 2021; 4:724-737. [PMID: 34676370 PMCID: PMC8528232 DOI: 10.33218/001c.18813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As exemplified by the COVID-19 pandemic, highly infective respiratory viruses can spread rapidly in the population because of lack of effective approaches to control viral replication and spread. Niclosamide (NCM) is an old anthelminthic drug (World Health Organization essential medicine list) with pleiotropic pharmacological activities. Several recent publications demonstrated that NCM has broad antiviral activities and potently inhibits viral replication, including replication of SARS-CoV-2, SARS-CoV, and dengue viruses. Unfortunately, NCM is almost completely insoluble in water, which limits its clinical use. We developed a cost-effective lipid nanoparticle formulation of NCM (nano NCM) using only FDA-approved excipient and demonstrated potency against SARS-CoV-2 infection in cells (Vero E6 and ACE2-expressing lung epithelium cells).
Collapse
Affiliation(s)
- Guankui Wang
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mary K McCarthy
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mercedes Gonzalez-Juarrero
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80521
| | - Yue Li
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| |
Collapse
|
34
|
Duong Phu M, Bross S, Burkhalter MD, Philipp M. Limitations and opportunities in the pharmacotherapy of ciliopathies. Pharmacol Ther 2021; 225:107841. [PMID: 33771583 DOI: 10.1016/j.pharmthera.2021.107841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023]
Abstract
Ciliopathies are a family of rather diverse conditions, which have been grouped based on the finding of altered or dysfunctional cilia, potentially motile, small cellular antennae extending from the surface of postmitotic cells. Cilia-related disorders include embryonically arising conditions such as Joubert, Usher or Kartagener syndrome, but also afflictions with a postnatal or even adult onset phenotype, i.e. autosomal dominant polycystic kidney disease. The majority of ciliopathies are syndromic rather than affecting only a single organ due to cilia being found on almost any cell in the human body. Overall ciliopathies are considered rare diseases. Despite that, pharmacological research and the strive to help these patients has led to enormous therapeutic advances in the last decade. In this review we discuss new treatment options for certain ciliopathies, give an outlook on promising future therapeutic strategies, but also highlight the limitations in the development of therapeutic approaches of ciliopathies.
Collapse
Affiliation(s)
- Max Duong Phu
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Stefan Bross
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany.
| |
Collapse
|
35
|
Dzijak R, Galeta J, Vázquez A, Kozák J, Matoušová M, Fulka H, Dračínský M, Vrabel M. Structurally Redesigned Bioorthogonal Reagents for Mitochondria-Specific Prodrug Activation. JACS AU 2021; 1:23-30. [PMID: 33554213 PMCID: PMC7851953 DOI: 10.1021/jacsau.0c00053] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Indexed: 06/05/2023]
Abstract
The development of abiotic chemical reactions that can be performed in an organelle-specific manner can provide new opportunities in drug delivery and cell and chemical biology. However, due to the complexity of the cellular environment, this remains a significant challenge. Here, we introduce structurally redesigned bioorthogonal tetrazine reagents that spontaneously accumulate in mitochondria of live mammalian cells. The attributes leading to their efficient accumulation in the organelle were optimized to include the right combination of lipophilicity and positive delocalized charge. The best performing mitochondriotropic tetrazines enable subcellular chemical release of TCO-caged compounds as we show using fluorogenic substrates and mitochondrial uncoupler niclosamide. Our work demonstrates that a shrewd redesign of common bioorthogonal reagents can lead to their transformation into organelle-specific probes, opening the possibility to activate prodrugs and manipulate biological processes at the subcellular level by using purely chemical tools.
Collapse
Affiliation(s)
- Rastislav Dzijak
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Juraj Galeta
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Arcadio Vázquez
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Jaroslav Kozák
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Marika Matoušová
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Helena Fulka
- Department
of Cell Nucleus Plasticity, Institute of
Experimental Medicine of the Czech Academy of Sciences, Víden̆ská 1083, 14220 Prague, Czech Republic
| | - Martin Dračínský
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Milan Vrabel
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| |
Collapse
|
36
|
Therapeutic potential of cannabinoids in combination cancer therapy. Adv Biol Regul 2021; 79:100774. [PMID: 33422460 DOI: 10.1016/j.jbior.2020.100774] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
Derivatives of the plant Cannabis sativa have been used for centuries for both medical and recreational purposes, as well as industrial. The first proof of its medicinal use comes from ancient China, although there is evidence of its earlier utilization in Europe and Asia. In the 19th century, European practitioners started to employ cannabis extracts to treat tetanus, convulsions, and mental diseases and, in 1851, cannabis made its appearance in the Pharmacopoeia of the United States as an analgesic, hypnotic and anticonvulsant. It was only in 1937 that the Marijuana Tax Act prohibited the use of this drug in the USA. The general term Cannabis is commonly used by the scientific and scholar community to indicate derivatives of the plant Cannabis sativa. The word cannabinoid is a term describing chemical compounds that are either derivate of Cannabis (phytocannabinoids) or artificial analogues (synthetic) or are produced endogenously by the body (endocannabinoids). A more casual term "marijuana" or "weed", a compound derived from dried Cannabis flower tops and leaves, has progressively superseded the term cannabis when referred to its recreational use. The 2018 World health organisation (WHO) data suggest that nearly 2.5% of the global population (147 million) uses marijuana and some countries, such as Canada and Uruguay, have already legalised it. Due to its controversial history, the medicinal use of cannabinoids has always been a centre of debate. The isolation and characterisation of Δ9 tetrahydrocannabinol (THC), the major psychoactive component of cannabis and the detection of two human cannabinoid receptor (CBRs) molecules renewed interest in the medical use of cannabinoids, boosting research and commercial heed in this sector. Some cannabinoid-based drugs have been approved as medications, mainly as antiemetic, antianorexic, anti-seizure remedies and in cancer and multiple sclerosis patients' palliative care. Nevertheless, due to the stigma commonly associated with these compounds, cannabinoids' potential in the treatment of conditions such as cancer is still largely unknown and therefore underestimated.
Collapse
|
37
|
STAT3 and p53: Dual Target for Cancer Therapy. Biomedicines 2020; 8:biomedicines8120637. [PMID: 33371351 PMCID: PMC7767392 DOI: 10.3390/biomedicines8120637] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor p53 is considered the "guardian of the genome" that can protect cells against cancer by inducing cell cycle arrest followed by cell death. However, STAT3 is constitutively activated in several human cancers and plays crucial roles in promoting cancer cell proliferation and survival. Hence, STAT3 and p53 have opposing roles in cellular pathway regulation, as activation of STAT3 upregulates the survival pathway, whereas p53 triggers the apoptotic pathway. Constitutive activation of STAT3 and gain or loss of p53 function due to mutations are the most frequent events in numerous cancer types. Several studies have reported the association of STAT3 and/or p53 mutations with drug resistance in cancer treatment. This review discusses the relationship between STAT3 and p53 status in cancer, the molecular mechanism underlying the negative regulation of p53 by STAT3, and vice versa. Moreover, it underlines prospective therapies targeting both STAT3 and p53 to enhance chemotherapeutic outcomes.
Collapse
|
38
|
Zhao H, Gao Y, Chen Q, Li J, Ren M, Zhao X, Yue W. RAD51AP1 promotes progression of ovarian cancer via TGF-β/Smad signalling pathway. J Cell Mol Med 2020; 25:1927-1938. [PMID: 33314567 PMCID: PMC7882964 DOI: 10.1111/jcmm.15877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 01/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the leading causes of female deaths. However, the molecular pathogenesis of OC has still remained elusive. This study aimed to explore the potential genes associated with the progression of OC. In the current study, 3 data sets of OC were downloaded from the GEO database to identify hub gene. Somatic mutation data obtained from TCGA were used to analyse the mutation. Immune cells were used to estimate effect of the hub gene to the tumour microenvironment. RNA‐seq and clinical data of OC patients retrieved from TCGA were used to investigate the diagnostic and prognostic values of hub gene. A series of in vitro assays were performed to indicate the function of hub gene and its possible mechanisms in OC. As a result, RAD51AP1 was found as a hub gene, which expression higher was mainly associated with poor survival in OC patients. Up‐regulation of RAD51AP1 was closely associated with mutations. RAD51AP1 up‐regulation accompanied by accumulated Th2 cells, but reduced CD4 + T cells and CD8 + T cells. Nomogram demonstrated RAD51AP1 increased the accuracy of the model. Down‐regulation of RAD51AP1 suppressed proliferation, migration and invasion capabilities of OC cells in vitro. Additionally, scatter plots showed that RAD51AP1 was positively correlated with genes in TGF‐β/Smad pathway. The above‐mentioned results were validated by RT‐qPCR and Western blotting. In conclusion, up‐regulation of RAD51AP1 was closely associated with mutations in OC. RAD51AP1 might represent an indicator for predicting OS of OC patients. Besides, RAD51AP1 might accelerate progression of OC by TGF‐β/Smad signalling pathway.
Collapse
Affiliation(s)
- Hongyu Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Yan Gao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Qi Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Jie Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Meng Ren
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Xiaoting Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital Capital Medical University, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
De Vitto H, Ryu J, Calderon-Aparicio A, Monts J, Dey R, Chakraborty A, Lee MH, Bode AM, Dong Z. Estrogen-related receptor alpha directly binds to p53 and cooperatively controls colon cancer growth through the regulation of mitochondrial biogenesis and function. Cancer Metab 2020; 8:28. [PMID: 33303020 PMCID: PMC7731476 DOI: 10.1186/s40170-020-00234-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 11/30/2020] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Of the genes that control mitochondrial biogenesis and function, ERRα emerges as a druggable metabolic target to be exploited for cancer therapy. Of the genes mutated in cancer, TP53 remains the most elusive to target. A clear understanding of how mitochondrial druggable targets can be accessed to exploit the underlying mechanism(s) explaining how p53-deficient tumors promote cell survival remains elusive. METHODS We performed protein-protein interaction studies to demonstrate that ERRα binds to p53. Moreover, we used gene silencing and pharmacological approaches in tandem with quantitative proteomics analysis by SWATH-MS to investigate the role of the ERRα/p53 complex in mitochondrial biogenesis and function in colon cancer. Finally, we designed in vitro and in vivo studies to investigate the possibility of targeting colon cancers that exhibit defects in p53. RESULTS Here, we are the first to identify a direct protein-protein interaction between the ligand-binding domain (LBD) of ERRα and the C-terminal domain (CTD) of p53. ERRα binds to p53 regardless of p53 mutational status. Furthermore, we show that the ERRα and p53 complex cooperatively control mitochondrial biogenesis and function. Targeting ERRα creates mitochondrial metabolic stresses, such as production of reactive oxygen species (ROS) and mitochondrial membrane permeabilization (MMP), leading to a greater cytotoxic effect that is dependent on the presence of p53. Pharmacological inhibition of ERRα impairs the growth of p53-deficient cells and of p53 mutant patient-derived colon xenografts (PDX). CONCLUSIONS Therefore, our data suggest that by using the status of the p53 protein as a selection criterion, the ERRα/p53 transcriptional axis can be exploited as a metabolic vulnerability.
Collapse
Affiliation(s)
- Humberto De Vitto
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Ali Calderon-Aparicio
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Josh Monts
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Raja Dey
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Abhijit Chakraborty
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Mee-Hyun Lee
- Department of Pathophysiology, Zhengzhou University School of Medicine, 40 North Road, 27 District University, Zhengzhou, 450052, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA.
| | - Zigang Dong
- Department of Pathophysiology, Zhengzhou University School of Medicine, 40 North Road, 27 District University, Zhengzhou, 450052, China.
| |
Collapse
|
40
|
The Undervalued Avenue to Reinstate Tumor Suppressor Functionality of the p53 Protein Family for Improved Cancer Therapy-Drug Repurposing. Cancers (Basel) 2020; 12:cancers12092717. [PMID: 32971841 PMCID: PMC7563196 DOI: 10.3390/cancers12092717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
p53 and p73 are critical tumor suppressors that are often inactivated in human cancers through various mechanisms. Owing to their high structural homology, the proteins have many joined functions and recognize the same set of genes involved in apoptosis and cell cycle regulation. p53 is known as the 'guardian of the genome' and together with p73 forms a barrier against cancer development and progression. The TP53 is mutated in more than 50% of all human cancers and the germline mutations in TP53 predispose to the early onset of multiple tumors in Li-Fraumeni syndrome (LFS), the inherited cancer predisposition. In cancers where TP53 gene is intact, p53 is degraded. Despite the ongoing efforts, the treatment of cancers remains challenging. This is due to late diagnoses, the toxicity of the current standard of care and marginal benefit of newly approved therapies. Presently, the endeavors focus on reactivating p53 exclusively, neglecting the potential of the restoration of p73 protein for cancer eradication. Taken that several small molecules reactivating p53 failed in clinical trials, there is a need to develop new treatments targeting p53 proteins in cancer. This review outlines the most advanced strategies to reactivate p53 and p73 and describes drug repurposing approaches for the efficient reinstatement of the p53 proteins for cancer therapy.
Collapse
|
41
|
Li N, Zhan X. MASS SPECTROMETRY-BASED MITOCHONDRIAL PROTEOMICS IN HUMAN OVARIAN CANCERS. MASS SPECTROMETRY REVIEWS 2020; 39:471-498. [PMID: 32020673 DOI: 10.1002/mas.21618] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/22/2020] [Indexed: 06/10/2023]
Abstract
The prominent characteristics of mitochondria are highly dynamic and regulatory, which have crucial roles in cell metabolism, biosynthetic, senescence, apoptosis, and signaling pathways. Mitochondrial dysfunction might lead to multiple serious diseases, including cancer. Therefore, identification of mitochondrial proteins in cancer could provide a global view of tumorigenesis and progression. Mass spectrometry-based quantitative mitochondrial proteomics fulfils this task by enabling systems-wide, accurate, and quantitative analysis of mitochondrial protein abundance, and mitochondrial protein posttranslational modifications (PTMs). Multiple quantitative proteomics techniques, including isotope-coded affinity tag, stable isotope labeling with amino acids in cell culture, isobaric tags for relative and absolute quantification, tandem mass tags, and label-free quantification, in combination with different PTM-peptide enrichment methods such as TiO2 enrichment of tryptic phosphopeptides and antibody enrichment of other PTM-peptides, increase flexibility for researchers to study mitochondrial proteomes. This article reviews isolation and purification of mitochondria, quantitative mitochondrial proteomics, quantitative mitochondrial phosphoproteomics, mitochondrial protein-involved signaling pathway networks, mitochondrial phosphoprotein-involved signaling pathway networks, integration of mitochondrial proteomic and phosphoproteomic data with whole tissue proteomic and transcriptomic data and clinical information in ovarian cancers (OC) to in-depth understand its molecular mechanisms, and discover effective mitochondrial biomarkers and therapeutic targets for predictive, preventive, and personalized treatment of OC. This proof-of-principle model about OC mitochondrial proteomics is easily implementable to other cancer types. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Na Li
- University Creative Research Initiatives Center, Shandong First Medical University, Shandong, 250062, P. R. China
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
| | - Xianquan Zhan
- University Creative Research Initiatives Center, Shandong First Medical University, Shandong, 250062, P. R. China
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- Department of Oncology, Xiangya Hospital, Central South University, 88 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 88 Xiangya Road, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
42
|
Salamoun JM, Garcia CJ, Hargett SR, Murray JH, Chen SY, Beretta M, Alexopoulos SJ, Shah DP, Olzomer EM, Tucker SP, Hoehn KL, Santos WL. 6-Amino[1,2,5]oxadiazolo[3,4- b]pyrazin-5-ol Derivatives as Efficacious Mitochondrial Uncouplers in STAM Mouse Model of Nonalcoholic Steatohepatitis. J Med Chem 2020; 63:6203-6224. [PMID: 32392051 PMCID: PMC11042500 DOI: 10.1021/acs.jmedchem.0c00542] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Small molecule mitochondrial uncouplers have recently garnered great interest for their potential in treating nonalcoholic steatohepatitis (NASH). In this study, we report the structure-activity relationship profiling of a 6-amino[1,2,5]oxadiazolo[3,4-b]pyrazin-5-ol core, which utilizes the hydroxy moiety as the proton transporter across the mitochondrial inner membrane. We demonstrate that a wide array of substituents is tolerated with this novel scaffold that increased cellular metabolic rates in vitro using changes in oxygen consumption rate as a readout. In particular, compound SHS4121705 (12i) displayed an EC50 of 4.3 μM in L6 myoblast cells and excellent oral bioavailability and liver exposure in mice. In the STAM mouse model of NASH, administration of 12i at 25 mg kg-1 day-1 lowered liver triglyceride levels and improved liver markers such as alanine aminotransferase, NAFLD activity score, and fibrosis. Importantly, no changes in body temperature or food intake were observed. As potential treatment of NASH, mitochondrial uncouplers show promise for future development.
Collapse
Affiliation(s)
- Joseph M Salamoun
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Christopher J Garcia
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stefan R Hargett
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Jacob H Murray
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Sing-Young Chen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Martina Beretta
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Stephanie J Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Divya P Shah
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Ellen M Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Simon P Tucker
- Continuum Biosciences, Pty Ltd., Sydney 2035, Australia
- Continuum Biosciences Inc., Boston, Massachusetts 02116, United States
| | - Kyle L Hoehn
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
43
|
Zhang X, Wang T, Song J, Deng J, Sun Z. Study on follicular fluid metabolomics components at different ages based on lipid metabolism. Reprod Biol Endocrinol 2020; 18:42. [PMID: 32398082 PMCID: PMC7216654 DOI: 10.1186/s12958-020-00599-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/24/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Follicular fluid is an important external environment for the growth and development of oocytes. A thorough identification of specific components in follicular fluid can better the existing understand of intracellular signal transduction and reveal potential biomarkers of oocyte health in women undergoing assisted reproductive therapy. To study on follicular fluid metabolomics components at different ages based on lipid metabolism, we have adopted a new method of SWATH to MRM(the sequential window acquisition of all theoretical fragment-ion spectra to multiple reaction monitor)metabolomics to provide extensive coverage and excellent quantitative data. This was done to investigate the differences in follicular fluid of patients undergoing in vitro fertilization (IVF) and embryo transfer in different age groups and to further explore the relationship between follicular fluid, age and reproductive function. METHOD A combination of Ultra-high-performance liquid chromatography and high resolution mass spectrometry techniques were used to analyze the follicular fluid of 230 patients enrolled for the IVF cycle. The patients were of different ages grouped into two groups:the younger and older patients.The obtained multidimensional chromatographic data were processed by principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA). The charge ratios and mass numbers enabled for the identification of different fragments in the samples. Matching information obtained through database search and the fragment information obtained by fragment ion scan structurally identified substances in the samples. This was used to determine the differential compounds. RESULTS The quality of oocytes decline with age,and the lipid composition in follicular fluid also changes,The lipid metabolism that changes with age may be related to the quality of oocytes.The main differences were in lipid metabolites. Some were up-regulated: Arachidonate, LysoPC(16:1), LysoPC(20:4) and LysoPC(20:3) while others were down-regulated: LysoPC(18:3) and LysoPC(18:1). CONCLUSIONS Metabolomic analysis of follicular fluid revealed that with the increase in age, several differential metabolites are at play. Among these metabolites, lipid metabolism undergoes significant changes that affect the development of oocytes thus causing reduced fertility in older women. These differential metabolites related to follicular development may provide possible detection and treatment targets for promoting oocyte health, and provide scientific basis for understanding the environment of oocyte development.
Collapse
Affiliation(s)
- Xingxing Zhang
- Maternity and Child Health Care of ZaoZhuang, ZaoZhuang, 277100, Shandong, China
| | - Tianqi Wang
- Traditional Chinese Medicine History and Literature, Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jingyan Song
- Department of Gynecology and Obstetrics of Traditional Chinese Medicine, The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jifeng Deng
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou, 510640, China
| | - Zhengao Sun
- Reproductive and Genetic Center of Integrated Traditional and Western Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| |
Collapse
|
44
|
Childress ES, Salamoun JM, Hargett SR, Alexopoulos SJ, Chen SY, Shah DP, Santiago-Rivera J, Garcia CJ, Dai Y, Tucker SP, Hoehn KL, Santos WL. [1,2,5]Oxadiazolo[3,4- b]pyrazine-5,6-diamine Derivatives as Mitochondrial Uncouplers for the Potential Treatment of Nonalcoholic Steatohepatitis. J Med Chem 2020; 63:2511-2526. [PMID: 32017849 DOI: 10.1021/acs.jmedchem.9b01440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Small molecule mitochondrial uncouplers are emerging as a new class of molecules for the treatment of nonalcoholic steatohepatitis. We utilized BAM15, a potent protonophore that uncouples the mitochondria without depolarizing the plasma membrane, as a lead compound for structure-activity profiling. Using oxygen consumption rate as an assay for determining uncoupling activity, changes on the 5- and 6-position of the oxadiazolopyrazine core were introduced. Our studies suggest that unsymmetrical aniline derivatives bearing electron withdrawing groups are preferred compared to the symmetrical counterparts. In addition, alkyl substituents are not tolerated, and the N-H proton of the aniline ring is responsible for the protonophore activity. In particular, compound 10b had an EC50 value of 190 nM in L6 myoblast cells. In an in vivo model of NASH, 10b decreased liver triglyceride levels and showed improvement in fibrosis, inflammation, and plasma ALT. Taken together, our studies indicate that mitochondrial uncouplers have potential for the treatment of NASH.
Collapse
Affiliation(s)
- Elizabeth S Childress
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Joseph M Salamoun
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stefan R Hargett
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Stephanie J Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Sing-Young Chen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Divya P Shah
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - José Santiago-Rivera
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Christopher J Garcia
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yumin Dai
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Simon P Tucker
- Continuum Biosciences, Pty Ltd., 2035 Sydney, Australia.,Continuum Biosciences Inc., Boston, Massachusetts 02116, United States
| | - Kyle L Hoehn
- Departments of Pharmacology and Medicine, University of Virginia, Charlottesville, Virginia 22908, United States.,School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
45
|
Nazarov PA, Kirsanov RS, Denisov SS, Khailova LS, Karakozova MV, Lyamzaev KG, Korshunova GA, Lukyanov KA, Kotova EA, Antonenko YN. Fluorescein Derivatives as Antibacterial Agents Acting via Membrane Depolarization. Biomolecules 2020; 10:biom10020309. [PMID: 32075319 PMCID: PMC7072581 DOI: 10.3390/biom10020309] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
Appending a lipophylic alkyl chain by ester bond to fluorescein has been previously shown to convert this popular dye into an effective protonophoric uncoupler of oxidative phosphorylation in mitochondria, exhibiting neuro- and nephroprotective effects in murine models. In line with this finding, we here report data on the pronounced depolarizing effect of a series of fluorescein decyl esters on bacterial cells. The binding of the fluorescein derivatives to Bacillus subtilis cells was monitored by fluorescence microscopy and fluorescence correlation spectroscopy (FCS). FCS revealed the energy-dependent accumulation of the fluorescein esters with decyl(triphenyl)- and decyl(tri-p-tolyl)phosphonium cations in the bacterial cells. The latter compound proved to be the most potent in suppressing B. subtilis growth.
Collapse
Affiliation(s)
- Pavel A. Nazarov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
- Correspondence: (P.A.N.); (E.A.K.)
| | - Roman S. Kirsanov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
| | - Stepan S. Denisov
- Department of Biochemistry, University of Maastricht, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands;
| | - Ljudmila S. Khailova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
| | - Marina V. Karakozova
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (M.V.K.); (K.A.L.)
| | - Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
| | - Galina A. Korshunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
| | - Konstantin A. Lukyanov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (M.V.K.); (K.A.L.)
| | - Elena A. Kotova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
- Correspondence: (P.A.N.); (E.A.K.)
| | - Yuri N. Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (R.S.K.); (L.S.K.); (K.G.L.); (G.A.K.); (Y.N.A.)
| |
Collapse
|
46
|
Voelkel NF, Peters-Golden M. A new treatment for severe pulmonary arterial hypertension based on an old idea: inhibition of 5-lipoxygenase. Pulm Circ 2020; 10:2045894019882635. [PMID: 32257113 PMCID: PMC7103594 DOI: 10.1177/2045894019882635] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
It has been generally accepted that severe forms of pulmonary arterial hypertension are associated with inflammation. Plasma levels in patients with severe pulmonary arterial hypertension show elevated levels of interleukins and mediators of inflammation and histologically the diseased small pulmonary arterioles show infiltrates of inflammatory and immune cells. Here, we review the literature that connects pulmonary hypertension with the arachidonic acid/5-lipoxygenase-derived leukotriens. This mostly preclinical background data together with the availability of 5-lipoxygenase inhibitors and leukotriene receptor blockers provide the rationale for testing the hypothesis that 5-lipoxygenase products contribute to the pathobiology of severe pulmonary arterial hypertension in a subgroup of patients.
Collapse
Affiliation(s)
- Norbert F. Voelkel
- Department of Pulmonary Medicine,
University of Amsterdam Medical Centers, Amsterdam, the Netherlands
| | - Marc Peters-Golden
- Pulmonary and Critical Care Medicine
Division,
University
of Michigan Medical School, Ann Arbor, MI,
USA
| |
Collapse
|
47
|
Klaus S, Ost M. Mitochondrial uncoupling and longevity - A role for mitokines? Exp Gerontol 2019; 130:110796. [PMID: 31786315 DOI: 10.1016/j.exger.2019.110796] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Aging has been viewed both as a random process due to accumulation of molecular and cellular damage over time and as a programmed process linked to cellular pathway important for growth and maturation. These views converge on mitochondria as both the major producer of damaging reactive oxidant species (ROS) and as signaling organelles. A finite proton leak across the inner mitochondrial membrane leading to a slight uncoupling of oxidative phosphorylation and respiration is an intrinsic property of all mitochondria and according to the "uncoupling to survive" hypothesis it has evolved to protect against ROS production to minimize oxidative damage. This hypothesis is supported by evidence linking an increased endogenous, uncoupling protein (UCP1) mediated, as well as experimentally induced mitochondrial uncoupling to an increased lifespan in rodents. This is possibly due to the synergistic activation of molecular pathways linked to life extending effects of caloric restriction as well as a mitohormetic response. Mitohormesis is an adaptive stress response through mitonuclear signaling which increases stress resistance resulting in health promoting effects. Part of this response is the induction of fibroblast growth factor 21 (FGF21) and growth and differentiation factor 15 (GDF15), two stress-induced mitokines which elicit beneficial systemic metabolic effects via endocrine action.
Collapse
Affiliation(s)
- Susanne Klaus
- German Institute of Human Nutrition in Potsdam Rehbrücke, Nuthetal, Germany; University of Potsdam, Institute of Nutritional Science, Potsdam, Germany.
| | - Mario Ost
- German Institute of Human Nutrition in Potsdam Rehbrücke, Nuthetal, Germany
| |
Collapse
|
48
|
MiR-532-3p suppresses colorectal cancer progression by disrupting the ETS1/TGM2 axis-mediated Wnt/β-catenin signaling. Cell Death Dis 2019; 10:739. [PMID: 31570702 PMCID: PMC6768886 DOI: 10.1038/s41419-019-1962-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/04/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
Abstract
The expression panel of plasma microRNA defined miR-532-3p as a valuable biomarker for colorectal adenoma (CRA). However, its expression pattern and function in colorectal cancer (CRC) have remained unclear. The present study investigated the expression levels of miR-532-3p and found that it was in situ downregulated both in CRA and CRC. Moreover, it functioned as a sensitizer for chemotherapy in CRC by inducing cell cycle arrest and early apoptosis via its activating effects on p53 and apoptotic signaling pathways. In addition, miR-532-3p was found to restrain cell growth, metastasis, and epithelial–mesenchymal transition (EMT) phenotype of CRC. A study on the mechanism behind these effects revealed that miR-532-3p directly binds to 3′UTR regions of ETS1 and TGM2, ultimately repressing the canonical Wnt/β-catenin signaling. Further investigation showed that TGM2 was transcriptionally regulated by ETS1 and ETS1/TGM2 axis served as a vital functional target of miR-532-3p in suppressing CRC progression. To conclude, miR-532-3p mimics could act as potential candidate for molecular therapy in CRC through inactivation of the canonical Wnt/β-catenin signaling and enhancement of chemosensitivity.
Collapse
|
49
|
Computer-aided drug repurposing for cancer therapy: Approaches and opportunities to challenge anticancer targets. Semin Cancer Biol 2019; 68:59-74. [PMID: 31562957 DOI: 10.1016/j.semcancer.2019.09.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Despite huge efforts made in academic and pharmaceutical worldwide research, current anticancer therapies achieve effective treatment in a limited number of neoplasia cases only. Oncology terms such as big killers - to identify tumours with yet a high mortality rate - or undruggable cancer targets, and chemoresistance, represent the current therapeutic debacle of cancer treatments. In addition, metastases, tumour microenvironments, tumour heterogeneity, metabolic adaptations, and immunotherapy resistance are essential features controlling tumour response to therapies, but still, lack effective therapeutics or modulators. In this scenario, where the pharmaceutical productivity and drug efficacy in oncology seem to have reached a plateau, the so-called drug repurposing - i.e. the use of old drugs, already in clinical use, for a different therapeutic indication - is an appealing strategy to improve cancer therapy. Opportunities for drug repurposing are often based on occasional observations or on time-consuming pre-clinical drug screenings that are often not hypothesis-driven. In contrast, in-silico drug repurposing is an emerging, hypothesis-driven approach that takes advantage of the use of big-data. Indeed, the extensive use of -omics technologies, improved data storage, data meaning, machine learning algorithms, and computational modeling all offer unprecedented knowledge of the biological mechanisms of cancers and drugs' modes of action, providing extensive availability for both disease-related data and drugs-related data. This offers the opportunity to generate, with time and cost-effective approaches, computational drug networks to predict, in-silico, the efficacy of approved drugs against relevant cancer targets, as well as to select better responder patients or disease' biomarkers. Here, we will review selected disease-related data together with computational tools to be exploited for the in-silico repurposing of drugs against validated targets in cancer therapies, focusing on the oncogenic signaling pathways activation in cancer. We will discuss how in-silico drug repurposing has the promise to shortly improve our arsenal of anticancer drugs and, likely, overcome certain limitations of modern cancer therapies against old and new therapeutic targets in oncology.
Collapse
|
50
|
Luo F, Luo M, Rong QX, Zhang H, Chen Z, Wang F, Zhao HY, Fu LW. Niclosamide, an antihelmintic drug, enhances efficacy of PD-1/PD-L1 immune checkpoint blockade in non-small cell lung cancer. J Immunother Cancer 2019; 7:245. [PMID: 31511071 PMCID: PMC6739982 DOI: 10.1186/s40425-019-0733-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022] Open
Abstract
Background PD-1/PD-L1 blockade has received approval for clinical application due to its encouraging benefit with improving prognosis in selected populations. Unfortunately, the response to immunotherapy for many patients remains unsatisfactory. It remains a great challenge to generate potential combinations that will outperform single agents alone with regard to anti-tumor activity. Methods Using NSCLC cell lines and mouse models, we explored the effects of combined niclosamide and PD-L1 blockade on tumor growth and T cell function. Furthermore, we investigated the relationship between PD-L1 and p-STAT3 expression in tumor samples from patients with NSCLC using IHC, as well as their relationship to patient survival. Results In vitro, niclosamide, an antihelmintic drug, enhanced the cancer cell lysis mediated by T cells in the presence of PD-L1 blockade. Accordingly, mice treated with niclosamide and PD-L1 antibody showed significant delay in tumor growth and increased survival which were associated with the increase of tumor infiltrating T cells and granzyme B release. Importantly, we found niclosamide could decrease the expression of PD-L1 in both a concentration- and time-dependent manner in NSCLC cells, which was linked to the blockage of p-STAT3 binding to the promoter of PD-L1. Conclusions An enhancement of PD-L1 antibody by niclosamide was observed in inhibition of NSCLC growth in vitro and in vivo, which was involved in blockage of p-STAT3 binding to promoter of PD-L1 and finally downregulation of PD-L1 expression. These encourage the combination therapy of niclosamide and PD-1/PD-L1 blockade to be further studied in clinic. Supplementary information Supplementary information accompanies this paper at 10.1186/s40425-019-0733-7.
Collapse
Affiliation(s)
- Fan Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Min Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qi-Xiang Rong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhen Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hong-Yun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Li-Wu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|