1
|
Arias-Betancur A, Fontova P, Alonso-Carrillo D, Carreira-Barral I, Duis J, García-Valverde M, Soto-Cerrato V, Quesada R, Pérez-Tomás R. Deregulation of lactate permeability using a small-molecule transporter (Lactrans-1) disturbs intracellular pH and triggers cancer cell death. Biochem Pharmacol 2024; 229:116469. [PMID: 39117009 DOI: 10.1016/j.bcp.2024.116469] [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: 02/14/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Due to the relevance of lactic acidosis in cancer, several therapeutic strategies have been developed targeting its production and/or regulation. In this matter, inhibition approaches of key proteins such as lactate dehydrogenase or monocarboxylate transporters have showed promising results, however, metabolic plasticity and tumor heterogeneity limits their efficacy. In this study, we explored the anticancer potential of a new strategy based on disturbing lactate permeability independently of monocarboxylate transporters activity using a small molecule ionophore named Lactrans-1. Derived from click-tambjamines, Lactrans-1 facilitates transmembrane lactate transportation in liposome models and reduces cancer cell viability. The results showed that Lactrans-1 triggered both apoptosis and necrosis depending on the cell line tested, displaying a synergistic effect in combination with first-line standard chemotherapeutic cisplatin. The ability of this compound to transport outward lactate anions was confirmed in A549 and HeLa cells, two cancer cell lines having distinct rates of lactate production. In addition, through cell viability reversion experiments it was possible to establish a correlation between the amount of lactate transported and the cytotoxic effect exhibited. The movement of lactate anions was accompanied with intracellular pH disturbances that included basification of lysosomes and acidification of the cytosol and mitochondria. We also observed mitochondrial swelling, increased ROS production and activation of oxidative stress signaling pathways p38-MAPK and JNK/SAPK. Our findings provide evidence that enhancement of lactate permeability is critical for cellular pH homeostasis and effective to trigger cancer cell death, suggesting that Lactrans-1 may be a promising anticancer therapy.
Collapse
Affiliation(s)
- Alain Arias-Betancur
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Integral Adult Dentistry, Research Centre for Dental Sciences (CICO), Dental School, Universidad de La Frontera, 4811230 Temuco, Chile
| | - Pere Fontova
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Daniel Alonso-Carrillo
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Israel Carreira-Barral
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Janneke Duis
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Avans University of Applied Science, 4818 AJ Breda, the Netherlands
| | - María García-Valverde
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Roberto Quesada
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001 Burgos, Spain.
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain; Molecular Signalling, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
2
|
Hassan SM, Farid A, Panda SS, Bekheit MS, Dinkins H, Fayad W, Girgis AS. Indole Compounds in Oncology: Therapeutic Potential and Mechanistic Insights. Pharmaceuticals (Basel) 2024; 17:922. [PMID: 39065774 PMCID: PMC11280311 DOI: 10.3390/ph17070922] [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: 06/18/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer remains a formidable global health challenge, with current treatment modalities such as chemotherapy, radiotherapy, surgery, and targeted therapy often hindered by low efficacy and adverse side effects. The indole scaffold, a prominent heterocyclic structure, has emerged as a promising candidate in the fight against cancer. This review consolidates recent advancements in developing natural and synthetic indolyl analogs, highlighting their antiproliferative activities against various cancer types over the past five years. These analogs are categorized based on their efficacy against common cancer types, supported by biochemical assays demonstrating their antiproliferative properties. In this review, emphasis is placed on elucidating the mechanisms of action of these compounds. Given the limitations of conventional cancer therapies, developing targeted therapeutics with enhanced selectivity and reduced side effects remains a critical focus in oncological research.
Collapse
Affiliation(s)
- Sara M. Hassan
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Alyaa Farid
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Siva S. Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt;
| | - Holden Dinkins
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt;
| |
Collapse
|
3
|
Hossain MM, Khalid A, Akhter Z, Parveen S, Ayaz MO, Bhat AQ, Badesra N, Showket F, Dar MS, Ahmed F, Dhiman S, Kumar M, Singh U, Hussain R, Keshari P, Mustafa G, Nargorta A, Taneja N, Gupta S, Mir RA, Kshatri AS, Nandi U, Khan N, Ramajayan P, Yadav G, Ahmed Z, Singh PP, Dar MJ. Discovery of a novel and highly selective JAK3 inhibitor as a potent hair growth promoter. J Transl Med 2024; 22:370. [PMID: 38637842 PMCID: PMC11025159 DOI: 10.1186/s12967-024-05144-4] [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: 12/06/2023] [Accepted: 03/23/2024] [Indexed: 04/20/2024] Open
Abstract
JAK-STAT signalling pathway inhibitors have emerged as promising therapeutic agents for the treatment of hair loss. Among different JAK isoforms, JAK3 has become an ideal target for drug discovery because it only regulates a narrow spectrum of γc cytokines. Here, we report the discovery of MJ04, a novel and highly selective 3-pyrimidinylazaindole based JAK3 inhibitor, as a potential hair growth promoter with an IC50 of 2.03 nM. During in vivo efficacy assays, topical application of MJ04 on DHT-challenged AGA and athymic nude mice resulted in early onset of hair regrowth. Furthermore, MJ04 significantly promoted the growth of human hair follicles under ex-vivo conditions. MJ04 exhibited a reasonably good pharmacokinetic profile and demonstrated a favourable safety profile under in vivo and in vitro conditions. Taken together, we report MJ04 as a highly potent and selective JAK3 inhibitor that exhibits overall properties suitable for topical drug development and advancement to human clinical trials.
Collapse
Affiliation(s)
- Md Mehedi Hossain
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Arfan Khalid
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Zaheen Akhter
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Sabra Parveen
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Neetu Badesra
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Farheen Showket
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Mohmmad Saleem Dar
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Farhan Ahmed
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Sumit Dhiman
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Mukesh Kumar
- Medicinal Product Chemistry, Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Umed Singh
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Razak Hussain
- Department of Entomology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Pankaj Keshari
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ghulam Mustafa
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Amit Nargorta
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Neha Taneja
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Somesh Gupta
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Riyaz A Mir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Aravind Singh Kshatri
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Utpal Nandi
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Nooruddin Khan
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - P Ramajayan
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Govind Yadav
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Zabeer Ahmed
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India
| | - Parvinder Pal Singh
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India.
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India.
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, 180001, India.
- Academy of Scientific & Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
4
|
Pachimatla AG, Fenstermaker R, Ciesielski M, Yendamuri S. Survivin in lung cancer: a potential target for therapy and prevention-a narrative review. Transl Lung Cancer Res 2024; 13:362-374. [PMID: 38496694 PMCID: PMC10938099 DOI: 10.21037/tlcr-23-621] [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: 09/26/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024]
Abstract
Background and Objective A versatile biomarker, survivin, is highly expressed in proliferating cells of multiple cancers in humans and animals. It is an apoptosis-regulating protein, engaging in a cascade of reactions that involve several other genes and protein interactions. Currently, researchers are investigating its therapeutic potential due to the evidence linking its overexpression to advanced-stage lung cancer. This review is centered around examining survivin-related molecular mechanisms and its therapeutic role specifically in lung cancer. Our objective is to discuss the role of survivin in prognosis and treatment response, shedding light on immune-targeted therapies, as well as outlining future directions for survivin-based vaccines in lung cancer. Methods The PubMed database and the United States National Library of Medicine search engine at the National Institutes of Health were searched on 24 August 2023 to identify published research studies. Searching "((((((airway [Title/Abstract]) OR (lung [Title/Abstract])) OR (pulm[Title/Abstract])) OR (bronch[Title/Abstract])) OR (nslc[Title/Abstract])) AND (((cancer[Title/Abstract]) OR (carcino[Title/Abstract])) OR (oncol[Title/Abstract]))) AND (survivin[Title/Abstract])" gave 728 results. After screening the title and abstracts and excluding the review articles 168 titles were shortlisted and full text studied. The discussions are added to relevant sections. Key Content and Findings Survivin is a cell cycle-dependent, inhibitor of apoptosis protein that contributes to carcinogenesis, tumor vascularization, metastasis, and treatment resistance. Several treatments that impact survivin either directly or indirectly have been reported as effective in treating lung cancer. Immunity-based therapy, a novel approach known for its targeted nature and minimal side effects, is currently under investigation for lung cancer treatment. Emerging survivin-centered vaccines exhibit promising attributes in terms of safety, effectiveness, and ability to stimulate an immune response. These factors point towards a significant potential for advancing the future of lung cancer prevention and enhancing overall survival rates. Conclusions Nuclear survivin is a potential biomarker for advanced non-small cell lung cancer. It plays a role in determining drug responsiveness and is found to be significantly elevated in cases of resistance to chemotherapy. Multiple compounds and immunization strategies have been identified to impact lung cancer cells; however, they are currently in the early stages of phase I or phase II clinical trials. The substantial promise of survivin-based immunogenicity-focused treatments warrants in-depth investigation and exploration.
Collapse
Affiliation(s)
- Akhil Goud Pachimatla
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Robert Fenstermaker
- Department of Neurosurgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael Ciesielski
- Department of Neurosurgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| |
Collapse
|
5
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Yin SS, Chen C, Liu Z, Liu SL, Guo JH, Zhang C, Zhang QW, Gao FH. Isoalantolactone mediates the degradation of BCR-ABL protein in imatinib-resistant CML cells by down-regulating survivin. Cell Cycle 2023; 22:1407-1420. [PMID: 37202916 PMCID: PMC10281474 DOI: 10.1080/15384101.2023.2209963] [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: 02/09/2022] [Revised: 11/02/2022] [Accepted: 02/10/2023] [Indexed: 05/20/2023] Open
Abstract
Isoalantolactone (Iso) is a bioactive lactone isolated from the root of Inula helenium L, which has been reported to have many pharmacological effects. To investigate the role and mechanism of isoalantolactone in chronic myeloid leukemia (CML), we first investigated isoalantolactone's anti-proliferative effects on imatinib-sensitive and imatinib-resistant CML cells by CCK8. Flow cytometry was used to detect isoalantolactone-induced cell apoptosis. Survivin was overexpressed in KBM5 and KBM5T315I cells using the lentivirus vector pSIN-3×flag-PURO. In KBM5 and KBM5T315I cells, shRNA was used to knockdown survivin. Cellular Thermal Shift Assay (CETSA) was used to detect the interaction between isoalantolactone and survivin. The ubiquitin of survivin induced by isoalantolactone was detected through immunoprecipitation. Quantitative polymerase-chain reaction (Q-PCR) and western blotting were used to detect the levels of mRNA and protein. Isoalantolactone inhibits the proliferation and promotes apoptosis of imatinib-resistant CML cells. Although isoalantolactone inhibits the proteins of BCR-ABL and survivin, it cannot inhibit survivin and BCR-ABL mRNA levels. Simultaneously, it was shown that isoalantolactone can degrade survivin protein by increasing ubiquitination. It was demonstrated that isoalantolactone-induced survivin mediated downregulation of BCR-ABL protein. It was also revealed that isoalantolactone triggered BCR-ABL protein degradation via caspase-3. Altogether, isoalantolactone inhibits survivin through the ubiquitin proteasome pathway, and mediates BCR-ABL downregulation in a caspase-3 dependent manner. These data suggest that isoalantolactone is a natural compound, which can be used as a potential drug to treat TKI-resistant CML.
Collapse
MESH Headings
- Humans
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Survivin
- Caspase 3
- Drug Resistance, Neoplasm
- Cell Proliferation
- Fusion Proteins, bcr-abl
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Apoptosis
- RNA, Messenger
- Ubiquitins/pharmacology
- Ubiquitins/therapeutic use
- Cell Line, Tumor
Collapse
Affiliation(s)
- Shan-Shan Yin
- Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing, Dongcheng District, Beijing, China
| | - Zhen Liu
- Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan-Ling Liu
- Department of Clinical Laboratory, The First Hospital of Changsha City, Changsha, China
| | - Jia-Hui Guo
- Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Zhang
- Department of Geriatrics, Shanghai ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quan-Wu Zhang
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Li X, Zhou L, Wang R, Zhang Y, Li W. Dihydromyricetin suppresses tumor growth via downregulation of the EGFR/Akt/survivin signaling pathway. J Biochem Mol Toxicol 2023:e23328. [PMID: 36807944 DOI: 10.1002/jbt.23328] [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: 06/17/2022] [Revised: 11/23/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Deregulation of epidermal growth factor receptor (EGFR) signaling is frequently observed in non-small cell lung cancer (NSCLC). The present study aimed to determine the impact of dihydromyricetin (DHM) on NSCLC, a natural compound extracted from Ampelopsis grossedentata with various pharmacological activities. Results of the present study demonstrated that DHM may act as a promising antitumor agent for NSCLC therapy, inhibiting the growth of cancer cells in vitro and in vivo. Mechanistically, results of the present study demonstrated that exposure to DHM downregulated the activity of wild-type (WT) and mutant EGFRs (mutations, exon 19 deletion, and L858R/T790M mutation). Moreover, western blot analysis indicated that DHM induced cell apoptosis via suppression of the antiapoptotic protein, survivin. Results of the present study further demonstrated that depletion or activation of EGFR/Akt signaling may regulate survivin expression though modulating ubiquitination. Collectively, these results suggested that DHM may act as a potential EGFR inhibitor, and may provide a novel choice of treatment strategy for patients with NSCLC.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ruike Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yangnan Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Li C, Qiu Y, Zhang Y. Research Progress on Therapeutic Targeting of Cancer-Associated Fibroblasts to Tackle Treatment-Resistant NSCLC. Pharmaceuticals (Basel) 2022; 15:1411. [PMID: 36422541 PMCID: PMC9696940 DOI: 10.3390/ph15111411] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 08/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for most lung cancer cases and is the leading cause of cancer-related deaths worldwide. Treatment options for lung cancer are no longer limited to surgery, radiotherapy, and chemotherapy, as targeted therapy and immunotherapy offer a new hope for patients. However, drug resistance in chemotherapy and targeted therapy, and the low response rates to immunotherapy remain important challenges. Similar to tumor development, drug resistance occurs because of significant effects exerted by the tumor microenvironment (TME) along with cancer cell mutations. Cancer-associated fibroblasts (CAFs) are a key component of the TME and possess multiple functions, including cross-talking with cancer cells, remodeling of the extracellular matrix (ECM), secretion of various cytokines, and promotion of epithelial-mesenchymal transition, which in turn provide support for the growth, invasion, metastasis, and drug resistance of cancer cells. Therefore, CAFs represent valuable therapeutic targets for lung cancer. Herein, we review the latest progress in the use of CAFs as potential targets and mediators of drug resistance for NSCLC treatment. We explored the role of CAFs on the regulation of the TME and surrounding ECM, with particular emphasis on treatment strategies involving combined CAF targeting within the current framework of cancer treatment.
Collapse
|
9
|
Yang J, Wang L, Guan X, Qin JJ. Inhibiting STAT3 signaling pathway by natural products for cancer prevention and therapy: In vitro and in vivo activity and mechanisms of action. Pharmacol Res 2022; 182:106357. [PMID: 35868477 DOI: 10.1016/j.phrs.2022.106357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/17/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in signal transmission from the plasma membrane to the nucleus, regulating the expression of genes involved in essential cell functions and controlling the processes of cell cycle progression and apoptosis. Thus, STAT3 has been elucidated as a promising target for developing anticancer drugs. Many natural products have been reported to inhibit the STAT3 signaling pathway during the past two decades and have exhibited significant anticancer activities in vitro and in vivo. However, there is no FDA-approved STAT3 inhibitor yet. The major mechanisms of these natural product inhibitors of the STAT3 signaling pathway include targeting the upstream regulators of STAT3, directly binding to the STAT3 SH2 domain and inhibiting its activation, inhibiting STAT3 phosphorylation and/or dimerization, and others. In the present review, we have systematically discussed the development of these natural product inhibitors of STAT3 signaling pathway as well as their in vitro and in vivo anticancer activity and mechanisms of action. Outlooks and perspectives on the associated challenges are provided as well.
Collapse
Affiliation(s)
- Jing Yang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Lingling Wang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
10
|
Batbold U, Liu JJ. Artemisia santolinifolia-Mediated Chemosensitization via Activation of Distinct Cell Death Modes and Suppression of STAT3/Survivin-Signaling Pathways in NSCLC. Molecules 2021; 26:molecules26237200. [PMID: 34885780 PMCID: PMC8658962 DOI: 10.3390/molecules26237200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/27/2023] Open
Abstract
Conventional chemotherapy remains an integral part of lung cancer therapy, regardless of its toxicity and drug resistance. Consequently, the discovery of an alternative to conventional chemotherapy is critical. Artemisia santolinifolia ethanol extract (AS) was assessed for its chemosensitizer ability when combined with the conventional anticancer drug, docetaxel (DTX), against non-small cell lung cancer (NSCLC). SRB assay was used to determine cell viability for A549 and H23 cell lines. The potential for this combination was examined by the combination index (CI). Further cell death, analyses with Annexin V/7AAD double staining, and corresponding protein expressions were analyzed. Surprisingly, AS synergistically enhanced the cytotoxic effect of DTX by inducing apoptosis in H23 cells through the caspase-dependent pathway, whereas selectively increased necrotic cell population in A549 cells, following the decline in GPX4 level and reactive oxygen species (ROS) activation with the highest rate in the combination treatment group. Furthermore, our results highlight the chemosensitization ability of AS when combined with DTX. It was closely associated with synergistic inhibition of oncogenesis signaling molecule STAT3 in both cell lines and concurrently downregulating prosurvival protein Survivin. Conclusively, AS could enhance DTX-induced cancer cells apoptosis by abrogating substantial prosurvival proteins' expressions and triggering two distinct cell death pathways. Our data also highlight that AS might serve as an adjunctive therapeutic option along with a conventional chemotherapeutic agent in the management of NSCLC patients.
Collapse
Affiliation(s)
- Uyanga Batbold
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Jun-Jen Liu
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Correspondence:
| |
Collapse
|
11
|
Mitra S, Prova SR, Sultana SA, Das R, Nainu F, Emran TB, Tareq AM, Uddin MS, Alqahtani AM, Dhama K, Simal-Gandara J. Therapeutic potential of indole alkaloids in respiratory diseases: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153649. [PMID: 34325978 DOI: 10.1016/j.phymed.2021.153649] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Indole alkaloids are very promising for potential therapeutic purposes and appear to be particularly effective against respiratory diseases. Several experimental studies have been performed, both in vivo and in vitro, to evaluate the effectiveness of indole alkaloids for the management of respiratory disorders, including asthma, emphysema, tuberculosis, cancer, and pulmonary fibrosis. PURPOSE The fundamental objective of this review was to summarize the in-depth therapeutic potential of indole alkaloids against various respiratory disorders. STUDY DESIGN In addition to describing the therapeutic potential, this review also evaluates the toxicity of these alkaloids, which have been utilized for therapeutic benefits but have demonstrated toxic consequences. Some indole alkaloids, including scholaricine, 19-epischolaricine, vallesamine, and picrinine, which are derived from the plant Alstonia scholaris, have shown toxic effects in non-rodent models. METHODS This review also discusses clinical studies exploring the therapeutic efficacy of indole alkaloids, which have confirmed the promising benefits observed in vivo and in vitro. RESULTS The indole alkaloidal compounds have shown efficacy in subjects with respiratory diseases. CONCLUSION The available data established both preclinical and clinical studies confirm the potential of indole alkaloids to treat the respiratory disorders.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Shajuthi Rahman Prova
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Sifat Ara Sultana
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi 90245, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh.
| | - Abu Montakim Tareq
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Ali M Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E32004 Ourense, Spain.
| |
Collapse
|
12
|
Li Y, Lu W, Yang J, Edwards M, Jiang S. Survivin as a biological biomarker for diagnosis and therapy. Expert Opin Biol Ther 2021; 21:1429-1441. [PMID: 33877952 DOI: 10.1080/14712598.2021.1918672] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Survivin (SVN) is a member of the inhibitor of apoptosis (IAP) protein family that promotes cellular proliferation and inhibits apoptosis. Overexpression of SVN is associated with autoimmune disease, hyperplasia, and tumors and can be used as a biomarker in these diseases. SVN is widely recognized as a tumor-associated antigen (TAA) and has become an important target for cancer diagnosis and treatment.Areas covered: We reviewed SVN research progress from the PubMed and clinical trials focused on SVN from https://clinicaltrials.gov since 2000 and anticipate future developments in the field. The trials reviewed cover various modalities including diagnostics for early detection and disease progression, small molecule inhibitors of the SVN pathway and immunotherapy targeting SVN epitopes.Expert opinion: The most promising developments involve anti-SVN immunotherapy, with several therapeutic SVN vaccines under evaluation in phase I/II trials. SVN is an important new immune-oncology target that expands the repertoire of individualized combination treatments for cancer.
Collapse
Affiliation(s)
- Yuming Li
- Department of Oncology, University of Oxford, Oxford, UK.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenshu Lu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Jiarun Yang
- Department of Oncology, University of Oxford, Oxford, UK
| | - Mark Edwards
- Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| | - Shisong Jiang
- Department of Oncology, University of Oxford, Oxford, UK.,Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| |
Collapse
|
13
|
Abstract
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
Collapse
|
14
|
Multi-Smart and Scalable Bioligands-Free Nanomedical Platform for Intratumorally Targeted Tambjamine Delivery, a Difficult to Administrate Highly Cytotoxic Drug. Biomedicines 2021; 9:biomedicines9050508. [PMID: 34064518 PMCID: PMC8147975 DOI: 10.3390/biomedicines9050508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/18/2021] [Accepted: 04/26/2021] [Indexed: 01/05/2023] Open
Abstract
Cancer is one of the leading causes of mortality worldwide due, in part, to limited success of some current therapeutic approaches. The clinical potential of many promising drugs is restricted by their systemic toxicity and lack of selectivity towards cancer cells, leading to insufficient drug concentration at the tumor site. To overcome these hurdles, we developed a novel drug delivery system based on polyurea/polyurethane nanocapsules (NCs) showing pH-synchronized amphoteric properties that facilitate their accumulation and selectivity into acidic tissues, such as tumor microenvironment. We have demonstrated that the anticancer drug used in this study, a hydrophobic anionophore named T21, increases its cytotoxic activity in acidic conditions when nanoencapsulated, which correlates with a more efficient cellular internalization. A biodistribution assay performed in mice has shown that the NCs are able to reach the tumor and the observed systemic toxicity of the free drug is significantly reduced in vivo when nanoencapsulated. Additionally, T21 antitumor activity is preserved, accompanied by tumor mass reduction compared to control mice. Altogether, this work shows these NCs as a potential drug delivery system able to reach the tumor microenvironment, reducing the undesired systemic toxic effects. Moreover, these nanosystems are prepared under scalable methodologies and straightforward process, and provide tumor selectivity through a smart mechanism independent of targeting ligands.
Collapse
|
15
|
Hassan M, Abuhamdah S, Abdel-Bary M, Wahman M, Abd-Elhamid T, Beshay M, Mosallam K, Elsadek B. Circulating and local nuclear expression of survivin and fibulin-3 genes in discriminating benign from malignant respiratory diseases: correlation analysis. Biosci Rep 2021; 41:BSR20203097. [PMID: 33226065 PMCID: PMC7789905 DOI: 10.1042/bsr20203097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/10/2020] [Accepted: 11/18/2020] [Indexed: 01/05/2023] Open
Abstract
Survivin is an inhibitor of apoptosis as well as a promoter of cell proliferation. Fibulin-3 is a matrix glycoprotein that displays potential for tumor suppression or propagation. The present study aimed to validate the expression levels of survivin and fibulin-3 in benign and malignant respiratory diseases. This case-control study included 219 patients categorized into five groups. Group A included 63 patients with lung cancer, group B included 63 patients with various benign lung diseases, group D included 45 patients with malignant pleural mesothelioma (MPM), and group E included 48 patients with various benign pleural diseases. Group C included 60 healthy individuals (control group). Serum survivin and fibulin-3 levels were measured by ELISA, whereas their nuclear expressions in the lung and pleura were assessed via Western blot analysis. The results showed significantly higher survivin serum levels and significantly lower fibulin-3 levels in group A compared with in group B and controls (P<0.001). There were significantly higher serum levels of survivin and fibulin-3 in group D compared with in group E and controls (P<0.001), consistent with observed nuclear survivin and fibulin-3 expression levels. Fibulin-3 was determined to have higher value than survivin in discriminating lung cancer from MPM (P<0.05). Survivin and fibulin-3 could be useful diagnostic markers for lung and pleural cancers, and fibulin-3 expression was particularly useful in differentiating lung cancer from MPM.
Collapse
Affiliation(s)
- Mohammed H. Hassan
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Sawsan Abuhamdah
- College of Pharmacy, Al Ain University, Abu Dhabi, UAE
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Mohamed Abdel-Bary
- Department of Cardio-Thoracic Surgery, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Mohammed Wahman
- Department of Oncology and Nuclear Medicine, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Tarek Hamdy Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Morris Beshay
- Department of General Thoracic Surgery, Protestant Hospital of Bethel Foundation, Burgsteig 13, Bielefeld, Germany
| | - Karam Mosallam
- Department of Cardio-Thoracic Surgery, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Bakheet E.M. Elsadek
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, 71524 Assiut, Egypt
| |
Collapse
|
16
|
Wang Q, Zhu Q, Ye Q, Wang J, Dong Q, Chen Y, Wang M, Fu Y, Wu R, Wu T. STAT3 Suppresses Cardiomyocytes Apoptosis in CVB3-Induced Myocarditis Via Survivin. Front Pharmacol 2021; 11:613883. [PMID: 33658937 PMCID: PMC7919905 DOI: 10.3389/fphar.2020.613883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Viral myocarditis (VMC) is a common inflammatory cardiovascular disease with unclear mechanisms, which mainly affects children and adolescents. Apoptosis is the key to CVB3-induced myocarditis, and blocking this process may be beneficial to the therapy of VMC. Hence, this study aimed to explore the protective function of STAT3 on cardiomyocyte apoptosis of VMC and its underlying mechanisms. Methods and Results: In this research, we confirmed that STAT3 was significantly activated in both animal and cell models of VMC. To further clarify what role did STAT3 play in VMC, AG490, an inhibitor of STAT3, was used to suppress p-STAT3. Our results demonstrated that decreased expression of p-STAT3 caused by AG490 significantly aggravated severity of VMC with elevated myocardial inflammation, deteriorative ventricular systolic function and increased mortality. It suggested that STAT3 plays a protective role in VMC. To further identify the anti-apoptosis impact that activated STAT3 made, we constructed lentivirus to regulate the expression of STAT3 in NMCs. We found that up-regulated activated STAT3 attenuated cardiomyocyte apoptosis, but down-regulated one aggravated that, which verified activated STAT3 played an anti-apoptosis role in VMC. Following that, we explored what elements are involved in the anti-apoptotic mechanism of activated STAT3 by using survivin inhibitor YM155. The result showed the anti-apoptotic effect of activated STAT3 does not work in the case of survivin inhibition. Conclusion: Our findings demonstrated STAT3 by targeting survivin alleviated cardiomyocyte apoptosis in CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Qiaoyu Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiongjun Zhu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiaofang Ye
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiajun Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qianqian Dong
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Youran Chen
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Minna Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Fu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rongzhou Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Sun T, Zhang J, Deng B, Fan X, Long T, Jin H, Tao S, Kang P, Tan Q. FOXO1 and FOXO3a sensitize non-small-cell lung cancer cells to cisplatin-induced apoptosis independent of Bim. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1348-1359. [PMID: 33167006 DOI: 10.1093/abbs/gmaa129] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Indexed: 12/12/2022] Open
Abstract
Low sensitivity to chemotherapy has been a major challenge in the treatment of non-small-cell lung cancer (NSCLC). It is of great clinical significance to discover its mechanisms to improve cell sensitivity to chemotherapeutic drugs. The forkhead box subfamily O (FOXO) transcriptional factors are downstream factors of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway and are reported to play pro-apoptotic roles in a variety of cells including NSCLC cells. But their roles and mechanisms in mediating cell response to chemotherapy remain to be discovered. We proposed that FOXO1 and FOXO3a may increase the sensitivity of NSCLC cells to cisplatin. Moreover, we presumed that LY294002, an inhibitor of the PI3K/AKT pathway, may enhance the cytotoxic effects of cisplatin through upregulating FOXO1 and FOXO3a. In the present study, we found that cisplatin initially increased the expressions and nuclear accumulation of FOXO1 and FOXO3a in NSCLC. Knockdown of FOXO1 and FOXO3a significantly decreased the cell sensitivity to cisplatin in vitro and in vivo. Moreover, inhibition of FOXO1 and FOXO3a attenuated cisplatin-induced cell apoptosis independent of Bim, a pro-apoptotic protein downstream of the FOXOs. Moreover, LY294002 synergistically increased the cytotoxic effects of cisplatin. Mechanistically, LY294002 increased the expressions and nuclear accumulation of FOXO1 and FOXO3a. Knockdown of FOXO1 and FOXO3a abrogated the enhancing effect of LY294002 on cisplatin. Taken together, our results suggested that FOXO1 and FOXO3a sensitize NSCLC cells to cisplatin and mediate the enhancing effects of LY294002 on cisplatin.
Collapse
Affiliation(s)
- Tianyu Sun
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jingge Zhang
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Bo Deng
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaoqing Fan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Tan Long
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hua Jin
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shaolin Tao
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Poming Kang
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
18
|
Novel Natural-based Biomolecules Discovery for Tackling Chronic Diseases. Biomolecules 2020; 10:biom10121674. [PMID: 33333911 PMCID: PMC7765416 DOI: 10.3390/biom10121674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/19/2020] [Indexed: 11/17/2022] Open
Abstract
In the last decade, natural-derived/-based biomolecules have continuously played an important role in novel drug discovery (as a prototype drug template) for potential chronic disease treatment [...]
Collapse
|
19
|
Gao F, Li M, Yu X, Liu W, Zhou L, Li W. Licochalcone A inhibits EGFR signalling and translationally suppresses survivin expression in human cancer cells. J Cell Mol Med 2020; 25:813-826. [PMID: 33247550 PMCID: PMC7812290 DOI: 10.1111/jcmm.16135] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022] Open
Abstract
Dysfunction of epidermal growth factor receptor (EGFR) signalling plays a critical role in the oncogenesis of non–small‐cell lung cancer (NSCLC). Here, we reported the natural product, licochalcone A, exhibited a profound anti‐tumour efficacy through directly targeting EGFR signalling. Licochalcone A inhibited in vitro cell growth, colony formation and in vivo tumour growth of either wild‐type (WT) or activating mutation EGFR‐expressed NSCLC cells. Licochalcone A bound with L858R single‐site mutation, exon 19 deletion, L858R/T790M mutation and WT EGFR ex vivo, and impaired EGFR kinase activity both in vitro and in NSCLC cells. The in silico docking study further indicated that licochalcone A interacted with both WT and mutant EGFRs. Moreover, licochalcone A induced apoptosis and decreased survivin protein robustly in NSCLC cells. Mechanistically, we found that treatment with licochalcone A translationally suppressed survivin through inhibiting EGFR downstream kinases ERK1/2 and Akt. Depletion of the translation initiation complex by eIF4E knockdown effectively inhibited survivin expression. In contrast, knockdown of 4E‐BP1 showed the opposite effect and dramatically enhanced survivin protein level. Overall, our data indicate that targeting survivin might be an alternative strategy to sensitize EGFR‐targeted therapy.
Collapse
Affiliation(s)
- Feng Gao
- Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The 3rd Xiangya Hospital of Central South University, Changsha, China
| | - Ming Li
- Cell Transplantation and Gene Therapy Institute, The 3rd Xiangya Hospital of Central South University, Changsha, China.,Changsha Stomatological Hospital, Changsha, China
| | - Xinfang Yu
- Cell Transplantation and Gene Therapy Institute, The 3rd Xiangya Hospital of Central South University, Changsha, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Wenbin Liu
- Department of Pathology, Hunan Cancer Hospital, Changsha, China
| | - Li Zhou
- Department of Pathology, Xiangya Hospital of Central South University, Changsha, China
| | - Wei Li
- Cell Transplantation and Gene Therapy Institute, The 3rd Xiangya Hospital of Central South University, Changsha, China.,Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
20
|
Barros-Nepomuceno FWA, de Araújo Viana D, Pinheiro DP, de Cássia Evangelista de Oliveira F, Magalhães Ferreira J, R de Queiroz MG, Ma X, Cavalcanti BC, Pessoa C, Banwell MG. The Effects of the Alkaloid Tambjamine J on Mice Implanted with Sarcoma 180 Tumor Cells. ChemMedChem 2020; 16:420-428. [PMID: 32886437 DOI: 10.1002/cmdc.202000387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/18/2020] [Indexed: 12/12/2022]
Abstract
The tambjamines are a small group of bipyrrolic alkaloids that, collectively, display a significant range of biological activities including antitumor, antimicrobial and immunosuppressive properties. The key objective of the present study was to undertake preclinical assessments of tambjamine J (T-J) so as to determine its in vivo antitumor effects. To that end, sarcoma 180 cells were transplanted in mice and the impacts of the title compound then evaluated using a range of protocols including hematological, biochemical, histopathological, genotoxic and clastogenic assays. As a result it was established that this alkaloid has a significant therapeutic window and effectively reduces tumor growth (by 40 % and 79 % at doses of 10 and 20 mg/kg/day, respectively). In this regard it displays similar antitumor activity to the anticancer agent cyclophosphamide and alters animal weight in an analogous manner.
Collapse
Affiliation(s)
- Francisco Washington A Barros-Nepomuceno
- Institute of Health Sciences, University for International Integration of the Afro-Brazilian Lusophony, Acarape, 62.785-000, CE, Brazil.,Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Daniel de Araújo Viana
- PATHOVET Laboratory, Pathological Anatomy and Veterinary Clinic, Fortaleza, 60.020.001, CE, Brazil
| | - Daniel Pascoalino Pinheiro
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | | | - Jamile Magalhães Ferreira
- Institute of Health Sciences, University for International Integration of the Afro-Brazilian Lusophony, Acarape, 62.785-000, CE, Brazil.,Clinical and Toxicological Analysis Department, Faculty of Pharmacy, Odontology and Nursing, Federal University of Ceará, Fortaleza, 60.714.903, CE, Brazil
| | - Maria Goretti R de Queiroz
- Clinical and Toxicological Analysis Department, Faculty of Pharmacy, Odontology and Nursing, Federal University of Ceará, Fortaleza, 60.714.903, CE, Brazil
| | - Xinghua Ma
- Research School of Chemistry, Institute of Advanced Studies, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bruno Coêlho Cavalcanti
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Claudia Pessoa
- Center for Research and Drug Development, Federal University of Ceará, Fortaleza, 60.430.275, CE, Brazil
| | - Martin G Banwell
- Research School of Chemistry, Institute of Advanced Studies, The Australian National University, Canberra, ACT, 2601, Australia.,Institute for Advanced and Applied Chemical Synthesis, Jinan University, Zhuhai, 519070, Guangdong, China
| |
Collapse
|
21
|
The importance of indole and azaindole scaffold in the development of antitumor agents. Eur J Med Chem 2020; 203:112506. [PMID: 32688198 DOI: 10.1016/j.ejmech.2020.112506] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022]
Abstract
With some indoles and azaindoles being successfully developed as anticancer drugs, the design and synthesis of indole and azaindole derivatives with remarkable antitumor activity has received increasing attention and significant progress has been made. This paper reviews the recent progress in the study of tumorigenesis, mechanism of actions and structure activity relationships about anticancer indole and azindole derivatives. Combining structure activity relationships and molecular targets-related knowledge, this review will help researchers design more effective, safe and cost-effective anticancer indoles and azindoles agents.
Collapse
|
22
|
Caparica R, Júlio A, Araújo MEM, Baby AR, Fonte P, Costa JG, Santos de Almeida T. Anticancer Activity of Rutin and Its Combination with Ionic Liquids on Renal Cells. Biomolecules 2020; 10:biom10020233. [PMID: 32033222 PMCID: PMC7072522 DOI: 10.3390/biom10020233] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/24/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
The renal cell carcinoma (RCC) is the most common type of kidney cancer. Identifying novel and more effective therapies, while minimizing toxicity, continues to be fundamental in curtailing RCC. Rutin, a bioflavonoid widely found in nature, has shown promising anticancer properties, but with limited applicability due to its poor water solubility and pharmacokinetics. Thus, the potential anticancer effects of rutin toward a human renal cancer cell line (786-O), while considering its safety in Vero kidney cells, was assessed, as well as the applicability of ionic liquids (ILs) to improve drug delivery. Rutin (up to 50 µM) did not show relevant cytotoxic effects in Vero cells. However, in 786-O cells, a significant decrease in cell viability was already observed at 50 µM. Moreover, exposure to rutin caused a significant increase in the sub-G1 population of 786-O cells, reinforcing the possible anticancer activity of this biomolecule. Two choline-amino acid ILs, at non-toxic concentrations, enhanced rutin’s solubility/loading while allowing the maintenance of rutin’s anticancer effects. Globally, our findings suggest that rutin may have a beneficial impact against RCC and that its combination with ILs ensures that this poorly soluble drug is successfully incorporated into ILs–nanoparticles hybrid systems, allowing controlled drug delivery.
Collapse
Affiliation(s)
- Rita Caparica
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (R.C.); (A.J.); (P.F.)
- Department of Biomedical Sciences, University of Alcalá, Ctra. Madrid-Barcelona Km. 33.600, Alcalá de Henares, 28871 Madrid, Spain
| | - Ana Júlio
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (R.C.); (A.J.); (P.F.)
- Department of Biomedical Sciences, University of Alcalá, Ctra. Madrid-Barcelona Km. 33.600, Alcalá de Henares, 28871 Madrid, Spain
| | - Maria Eduarda Machado Araújo
- CQE, and Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Campo Grande 1749-016 Lisboa, Portugal;
| | - André Rolim Baby
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, 580 Prof. Lineu Prestes Av., Bl. 15, São Paulo, SP 05508-900, Brazil;
| | - Pedro Fonte
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (R.C.); (A.J.); (P.F.)
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Center for Marine Sciences (CCMar), University of Algarve and Department of Chemistry and Pharmacy, 8005-139 Faro, Portugal
| | - João Guilherme Costa
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (R.C.); (A.J.); (P.F.)
- Correspondence: (J.G.C.); (T.S.d.A.); Tel.: +351-217515500 (T.S.d.A.)
| | - Tânia Santos de Almeida
- CBIOS—Universidade Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal; (R.C.); (A.J.); (P.F.)
- CQE, and Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Campo Grande 1749-016 Lisboa, Portugal;
- Correspondence: (J.G.C.); (T.S.d.A.); Tel.: +351-217515500 (T.S.d.A.)
| |
Collapse
|