151
|
Sarbadhikary P, George BP. A Review on Traditionally Used African Medicinal Plant Annickia chlorantha, Its Phytochemistry, and Anticancer Potential. PLANTS 2022; 11:plants11172293. [PMID: 36079675 PMCID: PMC9460708 DOI: 10.3390/plants11172293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022]
Abstract
Annickia chlorantha Setten & P.J.Maas belongs to the Annonaceae family and is a multi-purpose medicinal plant, which has been extensively used for the traditional treatment option for malaria in western and central Africa. Its phytochemical composition is dominated particularly by various biologically active protoberberines and acetogenins. This review aims to provide a comprehensive review on the traditional uses, phytochemical profiles, and the toxicology of this plant from a myriad of available publications. Even after its tremendous applications against several different human ailments, this plant has been underestimated for its anticancer potential. Herein, based on the phytochemical composition, we discuss the probable mode of mechanism for its antiproliferative activity, which highlights its importance for cytotoxicity screenings against cancer cells. Additionally, this article discusses several research questions and suggests the future directions of its applications in medicinal plant-based anticancer research.
Collapse
|
152
|
Li JJ, Xia XP, Wu LM, Zhu Z, Shi YN, Zhang XC, Xia YS, Lu GR. Cancer suppression by ferroptosis and its role in digestive system tumors. Shijie Huaren Xiaohua Zazhi 2022; 30:718-728. [DOI: 10.11569/wcjd.v30.i16.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Jia-Jia Li
- Department of Gastroenterology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Xuan-Ping Xia
- Department of Gastroenterology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Li-Min Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Zheng Zhu
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Yu-Ning Shi
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Xu-Chao Zhang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Yu-Shan Xia
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Guang-Rong Lu
- Department of Gastroenterology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
153
|
Oh J, An H, Yeo HJ, Choi S, Oh J, Kim S, Kim JM, Choi J, Lee S. Colchicine as a novel drug for the treatment of osteosarcoma through drug repositioning based on an FDA drug library. Front Oncol 2022; 12:893951. [PMID: 36059694 PMCID: PMC9433722 DOI: 10.3389/fonc.2022.893951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundColchicine is a traditional medication that is currently approved to treat gout and familial Mediterranean fever (FMF). However, colchicine has a wide range of anti-inflammatory activities, and several studies have indicated that it may be useful in a variety of other conditions, such as rheumatic disease, cardiac disease, and cancer. Osteosarcoma, the most common type of bone sarcoma, is derived from primitive bone-forming mesenchymal cells. In this study, we investigated whether colchicine could be used to treat osteosarcoma through the regulation of cell cycle signaling.MethodsTwo human osteosarcoma cell lines, U2OS and Saos-2, were used. A clonogenic assay was used to determine the antiproliferative effects of colchicine on osteosarcoma cells. Reactive oxygen species (ROS) production and apoptosis were measured by flow cytometry. Migration and invasion assays were performed to investigate the inhibitory effects of colchicine. The signaling pathways related to colchicine treatment were verified by GO biological process (GOBP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses.ResultsColchicine was selected as the lead compound based on the results of initial screening and cell viability assays conducted in Saos-2 and U2Os cells. Colchicine reduced the viability of Saos-2 and U2OS cells in a concentration-dependent manner. It also significantly inhibited colony-forming ability and induced ROS production and apoptosis. It also inhibited the migration and invasion of both Saos-2 and U2OS cells. GOBP and KEGG enrichment analyses indicated the involvement of microtubule-based processes and cancer-related pathways.ConclusionsThese findings suggest that colchicine has therapeutic potential in osteosarcoma.
Collapse
Affiliation(s)
- Jisun Oh
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Hyun−Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Hyun Jeong Yeo
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Sujin Choi
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Jisu Oh
- Division of Hemato-Oncology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, South Korea
| | - Segi Kim
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
| | - Jin Man Kim
- Department of Oral Microbiology and Immunology School of Dentistry, Seoul National University, Seoul, South Korea
| | - Junwon Choi
- Department of Molecular Science and Technology, Ajou University, Suwon-si, South Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, South Korea
- *Correspondence: Soonchul Lee,
| |
Collapse
|
154
|
Cheng C, Sun Q, Wang X, He B, Jiang T. Enzyme-manipulated hydrogelation of small molecules for biomedical applications. Acta Biomater 2022; 151:88-105. [PMID: 35970483 DOI: 10.1016/j.actbio.2022.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
Enzyme-manipulated hydrogelation based on self-assembly of small molecules is an attractive methodology for development of functional biomaterials. Upon the catalysis of enzymes, small-molecular precursors are converted into assemblable building blocks, which arrange into high-ordered nanofibers via non-covalent interactions at the molecular level, and further trap water to form hydrogels at the macroscopic level. Such approach has numerous advantages of region- and enantioselectivity, and mild reaction conditions for encapsulation of biomedications or cells that are fragile to environmental change. In addition to the common applications as drug reservoirs or cell scaffolds, the utilization of endogenous enzymes as stimuli to initiate self-assembly in the living cells and tissue is considered as an intelligent spatiotemporally controllable hydrogelation strategy for biomedical applications. The enzyme-instructed in situ self-assembly and hydrogelation can modulate the cell behavior, and even present therapeutic bioactivities, which provides a new perspective in the field of disease treatment. In this review, we categorize distinct enzymatic stimuli and elaborate substrate design, catalytic characteristics, and mechanisms of self-assembly and hydrogelation. The biomedical applications in drug delivery, tissue engineering, bioimaging, and in situ gelation-produced bioactivity are outlined. Advantages and limitations regarding the state-of-the-art enzyme-driven hydrogelation technologies and future perspectives are also discussed. STATEMENT OF SIGNIFICANCE: Hydrogel is a semi-solid soft material containing a large amount of water. Due to the features of adjustable flexibility, extremely porous architecture, and the high similarity of structure to natural extracellular matrices, the hydrogel has broad application prospects in biomedicine. In recent 20 years, enzyme-manipulated hydrogelation based on self-assembly of small molecules has developed rapidly as an attractive methodology for the construction of functional biomaterials. Upon the catalysis of enzymes, small-molecular precursors are converted into assemblable building blocks, which arrange into high-ordered nanofibers via non-covalent interactions at the molecular level, and further trap water to form hydrogels at the macroscopic level. This review summarized the characteristics of enzymatic hydrogel, as well as the traditional application and emerging prospect of enzyme-instructed self-assembly and hydrogelation.
Collapse
Affiliation(s)
- Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Qingyun Sun
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xiuping Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
155
|
Rashidi M, Mahmoudian E, Mirzaei S, Mazloomi SN, Bazi A, Azadeh H, Mozaffari M. Harmaline downregulates angiogenesis markers and suppresses the growth of 4T1 breast cancer cells in vivo and in vitro. Chem Biol Interact 2022; 365:110087. [PMID: 35963316 DOI: 10.1016/j.cbi.2022.110087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 11/29/2022]
Abstract
The anti-angiogenic effects of harmaline, an alkaloid with emerging anti-tumor properties, are under investigation. In the present study, the effects of different doses of harmaline, either alone or in combination with doxorubicin (DOX), were assessed in mice models of breast tumor. Breast tumors were created by the subcutaneous injection of 4T1 cells into Balb/c mice. The mice received either normal saline, harmaline alone (10, 20, or 30 mg/kg), or harmaline (20 mg/kg) + DOX (10 mg/kg). Immunohistochemistry, ELISA, and real-time PCR were conducted to measure target parameters. Harmaline significantly increased tumor cells' sensitivity to DOX as confirmed by a significantly reduced tumor volume in the harmaline + DOX group after 24 days (P < 0.05). Also, the levels of Ki-67 (P < 0.001), MMP-2 (P < 0.001), and VEGF (P < 0.001) significantly decreased while the level of E-cadherin increased (P < 0.001) in the tumor tissues of the mice treated with 20 or 30 mg/kg harmaline or harmaline (20 mg/kg) + DOX (10 mg/kg) compared to the control group. There was a significant reduction in the serum level of IL-4 in tumor-bearing mice treated with harmaline (P < 0.05), and IFN-γ serum level was significantly augmented in all experimental groups compared to the control group (P < 0.05). The genes encoding VEGF, VEGF receptor 2, CD105, and COX2 were significantly down-regulated (P < 0.05 for all) in harmaline-treated (either alone or in combination with DOX) mice. In conclusion, harmaline seems to have the potential to be used as an anticancer agent for treating breast cancer.
Collapse
Affiliation(s)
- Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center,Mazandaran University of Medical Sciences, Sari, Iran.
| | - Elham Mahmoudian
- Cellular & Molecular Medicine Department, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Rd., Roger Guindon Hall, Ottawa, ON, K1H 8M5, Canada
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Seyadeh Narges Mazloomi
- The Health of Plant and LivestockProducts Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Food and Drug Administration, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Bazi
- Department of Hematology, Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Hossein Azadeh
- Department of Internal Medicine, Rheumatology Division, Orthopedic Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mobina Mozaffari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
156
|
Chifiriuc MC, Filip R, Constantin M, Pircalabioru GG, Bleotu C, Burlibasa L, Ionica E, Corcionivoschi N, Mihaescu G. Common themes in antimicrobial and anticancer drug resistance. Front Microbiol 2022; 13:960693. [PMID: 36003940 PMCID: PMC9393787 DOI: 10.3389/fmicb.2022.960693] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial and anticancer drug resistance represent two of the main global challenges for the public health, requiring immediate practical solutions. In line with this, we need a better understanding of the origins of drug resistance in prokaryotic and eukaryotic cells and the evolutionary processes leading to the occurrence of adaptive phenotypes in response to the selective pressure of therapeutic agents. The purpose of this paper is to present some of the analogies between the antimicrobial and anticancer drug resistance. Antimicrobial and anticancer drugs share common targets and mechanisms of action as well as similar mechanisms of resistance (e.g., increased drug efflux, drug inactivation, target alteration, persister cells’ selection, protection of bacterial communities/malignant tissue by an extracellular matrix, etc.). Both individual and collective stress responses triggered by the chemotherapeutic agent involving complex intercellular communication processes, as well as with the surrounding microenvironment, will be considered. The common themes in antimicrobial and anticancer drug resistance recommend the utility of bacterial experimental models for unraveling the mechanisms that facilitate the evolution and adaptation of malignant cells to antineoplastic drugs.
Collapse
Affiliation(s)
- Mariana Carmen Chifiriuc
- Faculty of Biology, University of Bucharest, Bucharest, Romania
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, Bucharest, Romania
- The Romanian Academy, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, Suceava, Romania
- Suceava Emergency County Hospital, Suceava, Romania
| | | | - Gratiela Gradisteanu Pircalabioru
- Faculty of Biology, University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- *Correspondence: Gratiela Gradisteanu Pircalabioru,
| | - Coralia Bleotu
- Stefan S. Nicolau Institute of Virology, Bucharest, Romania
- Romanian Academy of Scientists, Bucharest, Romania
- Coralia Bleotu, ;
| | | | - Elena Ionica
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine—King Michael I of Romania, Timisoara, Romania
| | | |
Collapse
|
157
|
Hu T, Gong H, Xu J, Huang Y, Wu F, He Z. Nanomedicines for Overcoming Cancer Drug Resistance. Pharmaceutics 2022; 14:pharmaceutics14081606. [PMID: 36015232 PMCID: PMC9412887 DOI: 10.3390/pharmaceutics14081606] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Clinically, cancer drug resistance to chemotherapy, targeted therapy or immunotherapy remains the main impediment towards curative cancer therapy, which leads directly to treatment failure along with extended hospital stays, increased medical costs and high mortality. Therefore, increasing attention has been paid to nanotechnology-based delivery systems for overcoming drug resistance in cancer. In this respect, novel tumor-targeting nanomedicines offer fairly effective therapeutic strategies for surmounting the various limitations of chemotherapy, targeted therapy and immunotherapy, enabling more precise cancer treatment, more convenient monitoring of treatment agents, as well as surmounting cancer drug resistance, including multidrug resistance (MDR). Nanotechnology-based delivery systems, including liposomes, polymer micelles, nanoparticles (NPs), and DNA nanostructures, enable a large number of properly designed therapeutic nanomedicines. In this paper, we review the different mechanisms of cancer drug resistance to chemotherapy, targeted therapy and immunotherapy, and discuss the latest developments in nanomedicines for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Hanlin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Jiayue Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Yuan Huang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Fengbo Wu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| |
Collapse
|
158
|
Pandey P, Khan F, Qari HA, Upadhyay TK, Alkhateeb AF, Oves M. Evidence of Metallic and Polyether Ionophores as Potent Therapeutic Drug Candidate in Cancer Management. Molecules 2022; 27:4708. [PMID: 35897885 PMCID: PMC9329979 DOI: 10.3390/molecules27154708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer remains one of the most crucial human malignancies with a higher mortality rate globally, and is predicted to escalate soon. Dysregulated ion homeostasis in cancerous cells prompted the researchers to investigate further ion homeostasis impeding agents as potent anticancerous agents. Reutilization of FDA-approved non-cancerous drugs has emerged as a practical approach to developing potent, cost-effective drugs for cancer treatment. Across the globe, most nations are incapable of fulfilling the medical demands of cancer patients due to costlier cancerous drugs. Therefore, we have inclined our review towards emphasizing recent advancements in cancer therapies involving ionophores utilization in exploring potent anticancer drugs. Numerous research reports have established the significant anticancerous potential of ionophores in several pre-clinical reports via modulating aberrant cell signaling pathways and enhancing antitumor immunity in immune cells. This review has mainly summarized the most significant ion homeostasis impeding agents, including copper, zinc, calcium, and polyether, that presented remarkable potential in cancer therapeutics via enhanced antitumor immunity and apoptosis induction. Altogether, this study could provide a robust future perspective for developing cost-effective anticancerous drugs rapidly and cost-effectively, thereby combating the limitations of currently available drugs used in cancer treatment.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida 201306, India;
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida 201306, India;
| | - Huda A. Qari
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India;
| | - Abdulhameed F. Alkhateeb
- Department of Electrical & Computer Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad Oves
- Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Sendai 980-8577, Japan
| |
Collapse
|
159
|
Drug Discovery Using Evolutionary Similarities in Chemical Binding to Inhibit Patient-Derived Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23147971. [PMID: 35887321 PMCID: PMC9322808 DOI: 10.3390/ijms23147971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
Drug resistance causes therapeutic failure in refractory cancer. Cancer drug resistance stems from various factors, such as patient heterogeneity and genetic alterations in somatic cancer cells, including those from identical tissues. Generally, resistance is intrinsic for cancers; however, cancer resistance becomes common owing to an increased drug treatment. Unfortunately, overcoming this issue is not yet possible. The present study aimed to evaluate a clinical approach using candidate compounds 19 and 23, which are sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) inhibitors, discovered using the evolutionary chemical binding similarity method. mRNA sequencing indicated SERCA as the dominant marker of patient-derived anti-cancer drug-resistant hepatocellular carcinoma (HCC), but not of patient-derived anti-cancer drug-sensitive HCC. Candidate compounds 19 and 23 led to significant tumor shrinkage in a tumor xenograft model of anti-cancer drug-resistant patient-derived HCC cells. Our results might be clinically significant for the development of novel combinatorial strategies that selectively and efficiently target highly malignant cells such as drug-resistant and cancer stem-like cells.
Collapse
|
160
|
Jing G, Yang L, Wang H, Niu J, Li Y, Wang S. Interference of layered double hydroxide nanoparticles with pathways for biomedical applications. Adv Drug Deliv Rev 2022; 188:114451. [PMID: 35843506 DOI: 10.1016/j.addr.2022.114451] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/18/2022] [Accepted: 07/09/2022] [Indexed: 11/01/2022]
Abstract
Recent decades have witnessed a surge of explorations into the application of multifarious materials, especially biomedical applications. Among them, layered double hydroxides (LDHs) have been widely developed as typical inorganic layer materials to achieve remarkable advancements. Multiple physicochemical properties endow LDHs with excellent merits in biomedical applications. Moreover, LDH nanoplatforms could serve as "molecular switches", which are capable of the controlled release of payloads under specific physiological pH conditions but are stable during circulation in the bloodstream. In addition, LDHs themselves are composed of several specific cations and possess favorable biological effects or regulatory roles in various cellular functions. These advantages have caused LDHs to become increasingly of interest in the area of nanomedicine. Recent efforts have been devoted to revealing the potential factors that interfere with the biological pathways of LDH-based nanoparticles, such as their applications in shaping the functions of immune cells and in determining the fate of stem cells and tumor treatments, which are comprehensively described herein. In addition, several intracellular signaling pathways interfering with by LDHs in the above applications were also systematically expatiated. Finally, the future development and challenges of LDH-based nanomedicine are discussed in the context of the ultimate goal of practical clinical application.
Collapse
Affiliation(s)
- Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Linnan Yang
- Central Laboratory, First Affiliated Hospital, Anhui Medical University, Hefei, PR China
| | - Hong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Jintong Niu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Youyuan Li
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China.
| |
Collapse
|
161
|
Wang C, Xu X, Zhang P, Xiong S, Yuan J, Gao X, Guan W, Wang F, Li X, Dou H, Xu G. Lipid-coated albumin-paclitaxel nanoparticles loaded with sorcin-siRNA reverse cancer chemoresistance via restoring intracellular calcium ion homeostasis. J Nanobiotechnology 2022; 20:319. [PMID: 35799174 PMCID: PMC9264675 DOI: 10.1186/s12951-022-01487-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/31/2022] [Indexed: 12/17/2022] Open
Abstract
Chemoresistance is often a cause of the failure of chemotherapy in cancer treatment. Sorcin (SRI) is a soluble resistance-related calcium-binding protein involved in chemoresistant processes and is overexpressed in many chemoresistant cancer cells, including paclitaxel (PTX)-resistant ovarian cancer. Increased SRI can reduce the concentration of calcium ions in the cytosol and mitochondria and the decrease of calcium ion concentration prevents the occurrence of apoptosis. Here we examined the SRI expression in multiple cancers using a human TissueArray and found that SRI expression was significantly higher in malignant tumor tissues. Furthermore, SRI was overexpressed, while intracellular calcium concentration was decreased, in chemoresistant cancer cells. To restore intracellular calcium homeostasis and overcome chemoresistance, we developed lipid-coated albumin-PTX nanoparticles loaded with SRI-siRNA (LANP-PTX-siSRI) for PTX and SRI-siRNA co-delivery. LANP-PTX-siSRI had dual-target roles in the regulation of SRI and the delivery of PTX into chemoresistant cells. The LANP-PTX-siSRI inhibited the expression of SRI and enhanced intracellular calcium, leading to the induction of apoptosis and the inhibition of the growth of PTX-resistant cancer cells in vitro and in vivo. In addition, the mechanism study revealed that the overexpression of SRI was associated with an impaired TGF-β signaling pathway. The administration of TGF-β1 inhibited two calcium-binding proteins SRI and S100A14. In conclusion, our data unveil that restoring intracellular calcium ion homeostasis via reducing SRI expression can reverse chemoresistance. Thus, the fabricated LANP-PTX-siSRI has a potentially therapeutical application.
Collapse
Affiliation(s)
- Chenglong Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Xiaolin Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Peipei Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Shuhan Xiong
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Jia Yuan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Xuzhu Gao
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Wencai Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Fanchen Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Xin Li
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China.
| |
Collapse
|
162
|
Mahinfar P, Mansoori B, Rostamzadeh D, Baradaran B, Cho WC, Mansoori B. The Role of microRNAs in Multidrug Resistance of Glioblastoma. Cancers (Basel) 2022; 14:3217. [PMID: 35804989 PMCID: PMC9265057 DOI: 10.3390/cancers14133217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor that develops from neuroglial stem cells and represents a highly heterogeneous group of neoplasms. These tumors are predominantly correlated with a dismal prognosis and poor quality of life. In spite of major advances in developing novel and effective therapeutic strategies for patients with glioblastoma, multidrug resistance (MDR) is considered to be the major reason for treatment failure. Several mechanisms contribute to MDR in GBM, including upregulation of MDR transporters, alterations in the metabolism of drugs, dysregulation of apoptosis, defects in DNA repair, cancer stem cells, and epithelial-mesenchymal transition. MicroRNAs (miRNAs) are a large class of endogenous RNAs that participate in various cell events, including the mechanisms causing MDR in glioblastoma. In this review, we discuss the role of miRNAs in the regulation of the underlying mechanisms in MDR glioblastoma which will open up new avenues of inquiry for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Parvaneh Mahinfar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran;
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA 19104, USA
| |
Collapse
|
163
|
Sonbol H, Mohammed AE, Korany SM. Soil Fungi as Biomediator in Silver Nanoparticles Formation and Antimicrobial Efficacy. Int J Nanomedicine 2022; 17:2843-2863. [PMID: 35795079 PMCID: PMC9250898 DOI: 10.2147/ijn.s356724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/12/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction and Objectives Biogenic agents in nanoparticles fabrication are gaining great interest due to their lower possible negative environmental impacts. The present study aimed to isolate fungal strains from deserts in Saudi Arabia and assess their ability in silver nanoparticles (AgNPs) fabrication and evaluate their antibacterial effect. Methods Soil fungi were identified using 18s rDNA, and their ability in NPs fabrication was assessed as extracellular synthesis, then UV-vis spectroscopy, dynamic light scattering (DLS), energy-dispersive X-ray spectroscopy, and transmission electron microscopy were used for AgNPs characterization. The antibacterial activity of fungal-based NPs was assessed against one Gram-positive methicillin-resistant S. aureus (MRSA) and three Gram-negative bacteria (E. coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae). Ultrastructural changes caused by fungal-based NPs on K. pneumoniae were investigated using TEM along with SDS-PAGE for protein profile patterns. Results The three fungal isolates were identified as Phoma sp. (MN995524), Chaetomium globosum (MN995493), and Chaetomium sp. (MN995550), and their filtrate reduced Ag ions into spherical P-AgNPs, G-AgNPs, and C-AgNPs, respectively. DLS data showed an average size between 12.26 and 70.24 nm, where EDX spectrums represent Ag at 3.0 keV peak. G-AgNPs displayed strong antibacterial activities against Klebsiella pneumoniae, and the ultrastructural changes caused by NPs were noted. Additionally, SDS-PAGE analysis of treated K. pneumoniae revealed fewer bands compared to control, which could be related to protein degradation. Conclusion Present findings have consequently developed an eco-friendly approach in NPs formation by environmentally isolated fungal strains to yield NPs as antibacterial agents.
Collapse
Affiliation(s)
- Hana Sonbol
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Afrah E Mohammed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Shereen M Korany
- Botany and Microbiology Department, Faculty of Science, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
164
|
Wei Y, Amend B, Todenhöfer T, Lipke N, Aicher WK, Fend F, Stenzl A, Harland N. Urinary Tract Tumor Organoids Reveal Eminent Differences in Drug Sensitivities When Compared to 2-Dimensional Culture Systems. Int J Mol Sci 2022; 23:ijms23116305. [PMID: 35682984 PMCID: PMC9181330 DOI: 10.3390/ijms23116305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 06/01/2022] [Indexed: 12/31/2022] Open
Abstract
Generation of organoids from urinary tract tumor samples was pioneered a few years ago. We generated organoids from two upper tract urothelial carcinomas and from one bladder cancer sample, and confirmed the expression of cytokeratins as urothelial antigens, vimentin as a mesenchymal marker, and fibroblast growth factor receptor 3 by immunohistochemistry. We investigated the dose response curves of two novel components, venetoclax versus S63845, in comparison to the clinical standard cisplatin in organoids in comparison to the corresponding two-dimensional cultures. Normal urothelial cells and tumor lines RT4 and HT1197 served as controls. We report that upper tract urothelial carcinoma cells and bladder cancer cells in two-dimensional cultures yielded clearly different sensitivities towards venetoclax, S63845, and cisplatin. Two-dimensional cultures were more sensitive at low drug concentrations, while organoids yielded higher drug efficacies at higher doses. In some two-dimensional cell viability experiments, colorimetric assays yielded different IC50 toxicity levels when compared to chemiluminescence assays. Organoids exhibited distinct sensitivities towards cisplatin and to a somewhat lesser extent towards venetoclax or S63845, respectively, and significantly different sensitivities towards the three drugs investigated when compared to the corresponding two-dimensional cultures. We conclude that organoids maintained inter-individual sensitivities towards venetoclax, S63845, and cisplatin. The preclinical models and test systems employed may bias the results of cytotoxicity studies.
Collapse
Affiliation(s)
- Yi Wei
- Center for Medicine Research, Eberhard Karls University, 72072 Tuebingen, Germany; (Y.W.); (N.L.); (W.K.A.)
| | - Bastian Amend
- Department of Urology, University Hospital, 72076 Tuebingen, Germany; (B.A.); (T.T.); (A.S.)
| | - Tilman Todenhöfer
- Department of Urology, University Hospital, 72076 Tuebingen, Germany; (B.A.); (T.T.); (A.S.)
| | - Nizar Lipke
- Center for Medicine Research, Eberhard Karls University, 72072 Tuebingen, Germany; (Y.W.); (N.L.); (W.K.A.)
| | - Wilhelm K. Aicher
- Center for Medicine Research, Eberhard Karls University, 72072 Tuebingen, Germany; (Y.W.); (N.L.); (W.K.A.)
| | - Falko Fend
- Institute for Pathology, Eberhard Karls University, 72076 Tuebingen, Germany;
| | - Arnulf Stenzl
- Department of Urology, University Hospital, 72076 Tuebingen, Germany; (B.A.); (T.T.); (A.S.)
| | - Niklas Harland
- Department of Urology, University Hospital, 72076 Tuebingen, Germany; (B.A.); (T.T.); (A.S.)
- Correspondence: ; Tel.: +49-7071-298-6613
| |
Collapse
|
165
|
Khan MI, Alsayed RKME, Choudhry H, Ahmad A. Exosome-Mediated Response to Cancer Therapy: Modulation of Epigenetic Machinery. Int J Mol Sci 2022; 23:ijms23116222. [PMID: 35682901 PMCID: PMC9181065 DOI: 10.3390/ijms23116222] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Exosomes, the extracellular vesicles produced in the endosomal compartments, facilitate the transportation of proteins as well as nucleic acids. Epigenetic modifications are now considered important for fine-tuning the response of cancer cells to various therapies, and the acquired resistance against targeted therapies often involves dysregulated epigenetic modifications. Depending on the constitution of their cargo, exosomes can affect several epigenetic events, thus impacting post-transcriptional regulations. Thus, a role of exosomes as facilitators of epigenetic modifications has come under increased scrutiny in recent years. Exosomes can deliver methyltransferases to recipient cells and, more importantly, non-coding RNAs, particularly microRNAs (miRNAs), represent an important exosome cargo that can affect the expression of several oncogenes and tumor suppressors, with a resulting impact on cancer therapy resistance. Exosomes often harbor other non-coding RNAs, such as long non-coding RNAs and circular RNAs that support resistance. The exosome-mediated transfer of all this cargo between cancer cells and their surrounding cells, especially tumor-associated macrophages and cancer-associated fibroblasts, has a profound effect on the sensitivity of cancer cells to several chemotherapeutics. This review focuses on the exosome-induced modulation of epigenetic events with resulting impact on sensitivity of cancer cells to various therapies, such as, tamoxifen, cisplatin, gemcitabine and tyrosine kinase inhibitors. A better understanding of the mechanisms by which exosomes can modulate response to therapy in cancer cells is critical for the development of novel therapeutic strategies to target cancer drug resistance.
Collapse
Affiliation(s)
- Mohammad Imran Khan
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.I.K.); (H.C.)
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reem K. M. E. Alsayed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Hani Choudhry
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.I.K.); (H.C.)
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
- Correspondence: ; Tel.: +974-44390984
| |
Collapse
|
166
|
Abdelaal MR, Haffez H. The potential roles of retinoids in combating drug resistance in cancer: implications of ATP-binding cassette (ABC) transporters. Open Biol 2022; 12:220001. [PMID: 35642494 PMCID: PMC9157304 DOI: 10.1098/rsob.220001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Multidrug resistance (MDR) means that tumour cells become unresponsive during or after the course of treatment to one or more of chemotherapeutic drugs. Chemotherapeutic resistance critically limits the treatment outcomes and remains a key challenge for clinicians. The alternation in intracellular drug concentration through the modulation of its transport across the plasma membrane is the major cause for MDR and is adopted by various mediators, including ATP-requiring enzymes (ATPases). Among these ATPases, ABC transporters have been extensively studied, and found to be highly implicated in tumorigenesis and MDR. The present review sheds light on the documented effects of retinoids on ABC enzymes to understand their mechanism in combating cancer cell resistance. This would open the gate to test the mechanism and applicability of different new synthetic retinoids in literature and market as modulators of ATP-dependent efflux pumping activity, and promote their applicability in diminishing anti-cancer drug resistance.
Collapse
Affiliation(s)
- Mohamed R. Abdelaal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt,Centre of Scientific Excellence ‘Helwan Structural Biology Research (HSBR)’, Helwan University, Cairo 11795, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt,Centre of Scientific Excellence ‘Helwan Structural Biology Research (HSBR)’, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
167
|
Hamilton G. Comparative characteristics of small cell lung cancer and Ewing's sarcoma: a narrative review. Transl Lung Cancer Res 2022; 11:1185-1198. [PMID: 35832443 PMCID: PMC9271444 DOI: 10.21037/tlcr-22-58] [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: 01/19/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022]
Abstract
Background and Objective Small cell lung cancer (SCLC) and Ewing's sarcoma (ES) at the disseminated stage are not amenable to therapy and have a dismal prognosis with low survival rates. Despite representing different tumor entities, treatment for both malignancies relies on cytotoxic chemotherapy that has not considerably changed for the past decades. The genomic background has been extensively studied and found to comprise inactivation of p53 and RB1 in case of SCLC and EWSR1/FLI1 rearrangement in case of ES resulting in aggressive tumors in adults with heavy tobacco consumption and as bone tumor in juveniles, respectively. New therapeutic modalities are urgently needed to improve the outcomes of both tumor entities, especially in patients with metastatic disease or recurrences. This review summarizes the common cell biologic and clinical characteristics of difficult-to-treat SCLC and ES and discusses their refractoriness and options to improve the therapeutic efficacy. Methods PubMed and Euro PMC were searched from January 1st, 2012 to January 16th, 2022 using the following key words: "SCLC", "Ewing´s sarcoma", "Genomics" and "Chemoresistance" as well as own work. Key Content and Findings Therapy of SCLC and ES involves the use of undirected cytotoxic drugs in multimodal chemotherapy and administration of topotecan for 2nd line SCLC regimens. Despite highly aggressive chemotherapies, outcomes are dismal for patients with disseminated tumors. A host of unrelated drugs and targeted therapeutics have failed to result in progress for the patients and the underlying mechanisms of chemoresistance are still not clear. Identification of chemoresistance-reversing modulators in vitro and patient-derived xenografts of SCLC and ES has not translated into new therapies. Conclusions The global chemoresistance of SCLC and ES may be explained by physiological resistance at the tumor level and formation of larger spheroids that contain quiescent and hypoxic tumor cells in regions that occlude therapeutics. This type of chemoresistance is difficult to overcome and prevent the accumulation of effective drug concentration at the tumor cell level to a significant degree leaving therapeutic interventions of any kind ineffective.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
168
|
Wang H, Man Q, Huo F, Gao X, Lin H, Li S, Wang J, Su F, Cai, L, Shi Y, Liu, B, Bu L. STAT3 pathway in cancers: Past, present, and future. MedComm (Beijing) 2022; 3:e124. [PMID: 35356799 PMCID: PMC8942302 DOI: 10.1002/mco2.124] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/13/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, discovered in the cytoplasm of almost all types of mammalian cells, plays a significant role in biological functions. The duration of STAT3 activation in normal tissues is a transient event and is strictly regulated. However, in cancer tissues, STAT3 is activated in an aberrant manner and is induced by certain cytokines. The continuous activation of STAT3 regulates the expression of downstream proteins associated with the formation, progression, and metastasis of cancers. Thus, elucidating the mechanisms of STAT3 regulation and designing inhibitors targeting the STAT3 pathway are considered promising strategies for cancer treatment. This review aims to introduce the history, research advances, and prospects concerning the STAT3 pathway in cancer. We review the mechanisms of STAT3 pathway regulation and the consequent cancer hallmarks associated with tumor biology that are induced by the STAT3 pathway. Moreover, we summarize the emerging development of inhibitors that target the STAT3 pathway and novel drug delivery systems for delivering these inhibitors. The barriers against targeting the STAT3 pathway, the focus of future research on promising targets in the STAT3 pathway, and our perspective on the overall utility of STAT3 pathway inhibitors in cancer treatment are also discussed.
Collapse
Affiliation(s)
- Han‐Qi Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Qi‐Wen Man
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Oral & Maxillofacial Head Neck OncologySchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Fang‐Yi Huo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Xin Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Hao Lin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Su‐Ran Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Jing Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Fu‐Chuan Su
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Lulu Cai,
- Personalized Drug Therapy Key Laboratory of Sichuan ProvinceDepartment of PharmacySchool of MedicineSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory MedicineSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Bing Liu,
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Oral & Maxillofacial Head Neck OncologySchool & Hospital of StomatologyWuhan UniversityWuhanChina
| | - Lin‐Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedicine Ministry of EducationSchool & Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Oral & Maxillofacial Head Neck OncologySchool & Hospital of StomatologyWuhan UniversityWuhanChina
| |
Collapse
|
169
|
Guo Y, Shen Y, Yu B, Ding L, Meng Z, Wang X, Han M, Dong Z, Wang X. Hydrophilic Poly(glutamic acid)-Based Nanodrug Delivery System: Structural Influence and Antitumor Efficacy. Polymers (Basel) 2022; 14:2242. [PMID: 35683914 PMCID: PMC9182916 DOI: 10.3390/polym14112242] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Poly(amino acids) have advanced characteristics, including unique secondary structure, enzyme degradability, good biocompatibility, and stimuli responsibility, and are suitable as drug delivery nanocarriers for tumor therapy. The isoform structure of poly(amino acids) plays an important role in their antitumor efficacy and should be researched in detail. In this study, two kinds of pH-sensitive isoforms, including α-poly(glutamic acid) (α-PGA) and γ-PGA, were selected and used as nanocarriers to prepare a nanodrug delivery system. According to the preparation results, α-PGA can be used as an ideal drug carrier. Selecting doxorubicin (DOX) as the model drug, an α-PGA/DOX nanoparticle (α-PGA/DOX NPs) with a particle size of 110.4 nm was prepared, and the drug-loading content was 66.2%. α-PGA/DOX NPs presented obvious sustained and pH-dependent release characteristics. The IC50 value of α-PGA/DOX NPs was 1.06 ± 0.77 μg mL-1, decreasing by approximately 8.5 fold in vitro against 4T1 cells after incubation for 48 h. Moreover, α-PGA/DOX NPs enhanced antitumor efficacy in vivo, the tumor inhibition rate was 67.4%, increasing 1.5 fold over DOX injection. α-PGA/DOX NPs also reduced the systemic toxicity and cardiotoxicity of DOX. In sum, α-PGA is a biosafe nanodrug delivery carrier with potential clinical application prospects.
Collapse
Affiliation(s)
- Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yiping Shen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Bo Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Lijuan Ding
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Zheng Meng
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Xiaotong Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| | - Zhengqi Dong
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (Y.G.); (Y.S.); (B.Y.); (L.D.); (Z.M.); (X.W.); (M.H.)
| |
Collapse
|
170
|
Nie Z, Chen M, Gao Y, Huang D, Cao H, Peng Y, Guo N, Wang F, Zhang S. Ferroptosis and Tumor Drug Resistance: Current Status and Major Challenges. Front Pharmacol 2022; 13:879317. [PMID: 35668934 PMCID: PMC9163417 DOI: 10.3389/fphar.2022.879317] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a novel type of regulated cell death, whose unique metabolic characteristics are commonly used to evaluate the conditions of various diseases especially in tumors. Accumulating evidence supports that ferroptosis can regulate tumor development, metastasis, and therapeutic responses. Considering to the important role of chemotherapy in tumor treatment, drug resistance has become the most serious challenge. Revealing the molecular mechanism of ferroptosis is expected to solve tumor drug resistance and find new therapies to treat cancers. In this review, we discuss the relationship between ferroptosis and tumor drug resistance, summarize the abnormal ferroptosis in tissues of different cancer types and current research progress and challenges in overcoming treatment resistance, and explore the concept of targeting ferroptosis to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Fei Wang
- Department of Urology, Hainan General Hospital, Affiliated Hainan Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| |
Collapse
|
171
|
Mteremko D, Shadrack DM, Ntie-Kang F, Chilongola J, Chacha M. Finding alternatives to 5-fluorouracil: application of ensemble-based virtual screening for drug repositioning against human thymidylate synthase. J Biomol Struct Dyn 2022:1-17. [PMID: 35538714 DOI: 10.1080/07391102.2022.2074140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
5-fluorouracil and analogs are used in the treatment of many solid tumours. However, there are many cases of resistance and high toxicity associated with 5-fluorouracil chemotherapy. Repurposing FDA drugs against human thymidylate synthase revealed a number of FDA drugs that have a potential to be further developed for the treatment of various cancers for which 5-fluorouracil and analogs have been used for chemotherapy. Four FDA drugs prioritized for further validation included Erismodegib, Irinotecan, Conivaptan and Ergotamine. The role of water in mediating drug interactions and its contribution to the total binding energy was also shown. MM-PBSA calculations revealed that the binding affinity was the lowest for the hTS-Ergotamine complex (-66.702 ± 1.807 kJ/mol) suggesting moderate inhibition despite a large energetic contribution from van der Waal interactions (-190.889 ± 1.027 kJ/mol).Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Denis Mteremko
- Global Health and Biomedical Sciences, The Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania
| | | | | | - Jaffu Chilongola
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Musa Chacha
- Global Health and Biomedical Sciences, The Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania
| |
Collapse
|
172
|
Kumar S, Mishra S. MALAT1 as master regulator of biomarkers predictive of pan-cancer multi-drug resistance in the context of recalcitrant NRAS signaling pathway identified using systems-oriented approach. Sci Rep 2022; 12:7540. [PMID: 35534592 PMCID: PMC9085754 DOI: 10.1038/s41598-022-11214-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/18/2022] [Indexed: 11/25/2022] Open
Abstract
NRAS, a protein mutated in several cancer types, is involved in key drug resistance mechanisms and is an intractable target. The development of drug resistance is one of the major impediments in targeted therapy. Currently, gene expression data is used as the most predictive molecular profile in pan-cancer drug sensitivity and resistance studies. However, the common regulatory mechanisms that drive drug sensitivity/resistance across cancer types are as yet, not fully understood. We focused on GDSC data on NRAS-mutant pan-cancer cell lines, to pinpoint key signaling targets in direct or indirect associations with NRAS, in order to identify other druggable targets involved in drug resistance. Large-scale gene expression, comparative gene co-expression and protein–protein interaction network analyses were performed on selected drugs inducing drug sensitivity/resistance. We validated our data from cell lines with those obtained from primary tissues from TCGA. From our big data studies validated with independent datasets, protein-coding hub genes FN1, CD44, TIMP1, SNAI2, and SPARC were found significantly enriched in signal transduction, proteolysis, cell adhesion and proteoglycans pathways in cancer as well as the PI3K/Akt-signaling pathway. Further studies of the regulation of these hub/driver genes by lncRNAs revealed several lncRNAs as prominent regulators, with MALAT1 as a possible master regulator. Transcription factor EGR1 may control the transcription rate of MALAT1 transcript. Synergizing these studies, we zeroed in on a pan-cancer regulatory axis comprising EGR1-MALAT1-driver coding genes playing a role. These identified gene regulators are bound to provide new paradigms in pan-cancer targeted therapy, a foundation for precision medicine, through the targeting of these key driver genes in the improvement of multi-drug sensitivity or resistance.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Seema Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
173
|
Pahwa R, Chhabra J, Kumar R, Narang R. Melphalan: Recent insights on synthetic, analytical and medicinal aspects. Eur J Med Chem 2022; 238:114494. [DOI: 10.1016/j.ejmech.2022.114494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/15/2022] [Accepted: 05/24/2022] [Indexed: 12/17/2022]
|
174
|
Cao L, Zhu S, Lu H, Soutto M, Bhat N, Chen Z, Peng D, Lin J, Lu J, Li P, Zheng C, Huang C, El-Rifai W. Helicobacter pylori-induced RASAL2 Through Activation of Nuclear Factor-κB Promotes Gastric Tumorigenesis via β-catenin Signaling Axis. Gastroenterology 2022; 162:1716-1731.e17. [PMID: 35134322 PMCID: PMC9038683 DOI: 10.1053/j.gastro.2022.01.046] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori infection is the predominant risk factor for gastric cancer. RAS protein activator like 2 (RASAL2) is considered a double-edged sword in carcinogenesis. Herein, we investigated the role of RASAL2 in response to H pylori infection and gastric tumorigenesis. METHODS Bioinformatics analyses of local and public databases were applied to analyze RASAL2 expression, signaling pathways, and clinical significance. In vitro cell culture, spheroids, patient-derived organoids, and in vivo mouse models were used. Molecular assays included chromatin immunoprecipitation, co-immunoprecipitation, Western blotting, quantitative polymerase chain reaction, and immunocyto/histochemistry. RESULTS H pylori infection induced RASAL2 expression via a nuclear factor-κB (NF-κB)-dependent mechanism whereby NF-κB was directly bound to the RASAL2 promoter activating its transcription. By gene silencing and ectopic overexpression, we found that RASAL2 upregulated β-catenin transcriptional activity. RASAL2 inhibited protein phosphatase 2A activity through direct binding with subsequent activation of the AKT/β-catenin signaling axis. Functionally, RASAL2 silencing decreased nuclear β-catenin levels and impaired tumor spheroids and organoids formation. Furthermore, the depletion of RASAL2 impaired tumor growth in gastric tumor xenograft mouse models. Clinicopathological analysis indicated that abnormal overexpression of RASAL2 correlated with poor prognosis and chemoresistance in human gastric tumors. CONCLUSIONS These studies uncovered a novel signaling axis of NF-κB/RASAL2/β-catenin, providing a novel link between infection, inflammation and gastric tumorigenesis.
Collapse
Affiliation(s)
- Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Shoumin Zhu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Heng Lu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Mohammed Soutto
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Nadeem Bhat
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Zheng Chen
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Dunfa Peng
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Wael El-Rifai
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida; Department of Veterans Affairs, Miami Healthcare System, Miami, Florida; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
175
|
The Mechanisms of lncRNA-Mediated Multidrug Resistance and the Clinical Application Prospects of lncRNAs in Breast Cancer. Cancers (Basel) 2022; 14:cancers14092101. [PMID: 35565231 PMCID: PMC9103444 DOI: 10.3390/cancers14092101] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Multidrug resistance (MDR) is a major cause of breast cancer (BC) chemotherapy failure. Long noncoding RNAs (lncRNAs) have been shown closely related to the chemoresistance of BC. In this work, the mechanisms of lncRNA-mediated MDR in BC were elaborated from eight sections, including apoptosis, autophagy, DNA repair, cell cycle, drug efflux, epithelial-mesenchymal transition, epigenetic modification and the tumor microenvironment. Additionally, we also discuss the clinical significance of lncRNAs, which may be biomarkers for diagnosis, therapy and prognosis. Abstract Breast cancer (BC) is a highly heterogeneous disease and presents a great threat to female health worldwide. Chemotherapy is one of the predominant strategies for the treatment of BC; however, multidrug resistance (MDR) has seriously affected or hindered the effect of chemotherapy. Recently, a growing number of studies have indicated that lncRNAs play vital and varied roles in BC chemoresistance, including apoptosis, autophagy, DNA repair, cell cycle, drug efflux, epithelial-mesenchymal transition (EMT), epigenetic modification and the tumor microenvironment (TME). Although thousands of lncRNAs have been implicated in the chemoresistance of BC, a systematic review of their regulatory mechanisms remains to be performed. In this review, we systematically summarized the mechanisms of MDR and the functions of lncRNAs mediated in the chemoresistance of BC from the latest literature. These findings significantly enhance the current understanding of lncRNAs and suggest that they may be promising prognostic biomarkers for BC patients receiving chemotherapy, as well as therapeutic targets to prevent or reverse chemoresistance.
Collapse
|
176
|
Mehmood S, Faheem M, Ismail H, Farhat SM, Ali M, Younis S, Asghar MN. ‘Breast Cancer Resistance Likelihood and Personalized Treatment Through Integrated Multiomics’. Front Mol Biosci 2022; 9:783494. [PMID: 35495618 PMCID: PMC9048735 DOI: 10.3389/fmolb.2022.783494] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
In recent times, enormous progress has been made in improving the diagnosis and therapeutic strategies for breast carcinoma, yet it remains the most prevalent cancer and second highest contributor to cancer-related deaths in women. Breast cancer (BC) affects one in eight females globally. In 2018 alone, 1.4 million cases were identified worldwide in postmenopausal women and 645,000 cases in premenopausal females, and this burden is constantly increasing. This shows that still a lot of efforts are required to discover therapeutic remedies for this disease. One of the major clinical complications associated with the treatment of breast carcinoma is the development of therapeutic resistance. Multidrug resistance (MDR) and consequent relapse on therapy are prevalent issues related to breast carcinoma; it is due to our incomplete understanding of the molecular mechanisms of breast carcinoma disease. Therefore, elucidating the molecular mechanisms involved in drug resistance is critical. For management of breast carcinoma, the treatment decision not only depends on the assessment of prognosis factors but also on the evaluation of pathological and clinical factors. Integrated data assessments of these multiple factors of breast carcinoma through multiomics can provide significant insight and hope for making therapeutic decisions. This omics approach is particularly helpful since it identifies the biomarkers of disease progression and treatment progress by collective characterization and quantification of pools of biological molecules within and among the cancerous cells. The scrupulous understanding of cancer and its treatment at the molecular level led to the concept of a personalized approach, which is one of the most significant advancements in modern oncology. Likewise, there are certain genetic and non-genetic tests available for BC which can help in personalized therapy. Genetically inherited risks can be screened for personal predisposition to BC, and genetic changes or variations (mutations) can also be identified to decide on the best treatment. Ultimately, further understanding of BC at the molecular level (multiomics) will define more precise choices in personalized medicine. In this review, we have summarized therapeutic resistance associated with BC and the techniques used for its management.
Collapse
Affiliation(s)
- Sabba Mehmood
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
- *Correspondence: Sabba Mehmood, ; Muhammad Nadeem Asghar,
| | - Muhammad Faheem
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Hammad Ismail
- Department of Biochemistry & Biotechnology University of Gujrat, Gujrat, Pakistan
| | - Syeda Mehpara Farhat
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Mahwish Ali
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Sidra Younis
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Muhammad Nadeem Asghar
- Department of Medical Biology, University of Québec at Trois-Rivieres, Trois-Rivieres, QC, Canada
- *Correspondence: Sabba Mehmood, ; Muhammad Nadeem Asghar,
| |
Collapse
|
177
|
Shao N, Yuan L, Ma P, Zhou M, Xiao X, Cong Z, Wu Y, Xiao G, Fei J, Liu R. Heterochiral β-Peptide Polymers Combating Multidrug-Resistant Cancers Effectively without Inducing Drug Resistance. J Am Chem Soc 2022; 144:7283-7294. [PMID: 35420800 DOI: 10.1021/jacs.2c00452] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multidrug resistance to chemotherapeutic drugs is one of the major causes for the failure of cancer treatment. Therefore, there is an urgent need to develop anticancer agents that can combat multidrug-resistant cancers effectively and mitigate drug resistance. Here, we report a rational design of anticancer heterochiral β-peptide polymers as synthetic mimics of host defense peptides to combat multidrug-resistant cancers. The optimal polymer shows potent and broad-spectrum anticancer activities against multidrug-resistant cancer cells and is insusceptible to anticancer drug resistance owing to its membrane-damaging mechanism. The in vivo study indicates that the optimal polymer efficiently inhibits the growth and distant transfer of solid tumors and the metastasis and seeding of circulating tumor cells. Moreover, the polymer shows excellent biocompatibility during anticancer treatment on animals. In addition, the β-peptide polymers address those prominent shortcomings of anticancer peptides and have superior stability against proteolysis, easy synthesis in large scale, and low cost. Collectively, the structural diversity and superior anticancer performance of β-peptide polymers imply an effective strategy in designing and finding anticancer agents to combat multidrug-resistant cancers effectively while mitigating drug resistance.
Collapse
Affiliation(s)
- Ning Shao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ling Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Pengcheng Ma
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Min Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ximian Xiao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zihao Cong
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yueming Wu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Guohui Xiao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Fei
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
178
|
Liposomal Nanoformulation as a Carrier for Curcumin and pEGCG—Study on Stability and Anticancer Potential. NANOMATERIALS 2022; 12:nano12081274. [PMID: 35457986 PMCID: PMC9028936 DOI: 10.3390/nano12081274] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 02/08/2023]
Abstract
Nanoformulations are regarded as a promising tool to enable the efficient delivery of active pharmaceutical ingredients to the target site. One of the best-known and most studied nanoformulations are liposomes—spherical phospholipid bilayered nanocarriers resembling cell membranes. In order to assess the possible effect of a mixture of polyphenols on both the stability of the formulation and its biological activity, two compounds were embedded in the liposomes—(i) curcumin (CUR), (ii) a peracetylated derivative of (−)-epigallocatechin 3-O-gallate (pEGCG), and (iii) a combination of the aforementioned. The stability of the formulations was assessed in two different temperature ranges (4–8 and 20 °C) by monitoring both the particle size and their concentration. It was found that after 28 days of the experiment, the liposomes remained largely unchanged in terms of the particle size distribution, with the greatest change from 130 to 146 nm. The potential decomposition of the carried substances was evaluated using HPLC. The combined CUR and pEGCG was sensitive to temperature conditions; however its stability was greatly increased when compared to the solutions of the individual compounds alone—up to 9.67% of the initial concentration of pEGCG in liposomes after 28 days storage compared to complete decomposition within hours for the non-encapsulated sample. The potential of the prepared formulations was assessed in vitro on prostate (LNCaP) and bladder cancer (5637) cell lines, as well as on a non-cancerous human lung fibroblast cell line (MRC-5), with the highest activity of IC50 equal 15.33 ± 2.03 µM for the mixture of compounds towards the 5637 cell line.
Collapse
|
179
|
Peh KH, Przybylski DJ, Fallon MJ, Bergsbaken JJ, Hutson PR, Yu M, Deming DA, Burkard ME. Clinical utility of a regional precision medicine molecular tumor board and challenges to implementation. J Oncol Pharm Pract 2022:10781552221091282. [DOI: 10.1177/10781552221091282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose Molecular tumor boards provide precision treatment recommendations based on cancer genomic profile. However, practical barriers limit their benefits. We studied the clinical utility of the precision medicine molecular tumor board (PMMTB) and described challenges with PMMTB implementation. Methods An observational cohort study included patients reviewed by the PMMTB between September 2015 to December 2017. Patients who had consented to the registry study were included. The primary endpoint of this study was time on treatment (ToT) ratio. Clinical utility was established if the primary endpoint had least 15% of patients achieving a ToT ratio of ≥1.3. Results Overall, 278 patients were presented to the PMMTB and 113 cases were included in the final analysis. The PMMTB identified at least one nonstandard of care (SOC) clinically actionable mutation for 69.0% (78/113) of cases. In patients who received non-SOC treatment, 43.8% (7/16) achieved a ToT ratio of 1.3 or more (p < 0.001). Fifty-nine patients did not receive non-SOC recommendations. Reasons for not pursuing treatment included 35.6% having response to current treatment, 20.3% died prior to starting or considering PMMTB recommendations, 13.6% pursued other treatment options based on clinician discretion, another 10.2% pursued other treatment options because clinical trials recommended were not geographically accessible, 8.5% had rapid decline of performance status, 6.8% lacked of financial support for treatment, and 5.1% were excluded from clinical trials due to abnormal laboratory values. Conclusion The regional PMMTB non-SOC recommendations benefitted a majority of patients and additional processes were implemented to assist with non-SOC treatment accessibility.
Collapse
Affiliation(s)
- Keng Hee Peh
- University of Kentucky College of Pharmacy, Lexington, KY, United States
| | | | | | | | - Paul R Hutson
- School of Pharmacy, University of Wisconsin - Madison, Madison, WI, United States
| | - Menggang Yu
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| | - Dustin A Deming
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
- Department of Medicine, Hematology/Oncology and McArdle Laboratories, University of Wisconsin, Madison, WI, United States
| | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
- Department of Medicine, Hematology/Oncology and McArdle Laboratories, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
180
|
TRIM46 activates AKT/HK2 signaling by modifying PHLPP2 ubiquitylation to promote glycolysis and chemoresistance of lung cancer cells. Cell Death Dis 2022; 13:285. [PMID: 35354796 PMCID: PMC8967906 DOI: 10.1038/s41419-022-04727-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 12/13/2022]
Abstract
The incidence of lung cancer is increasing worldwide. Although great progress in lung cancer treatment has been made, the clinical outcome is still unsatisfactory. Tripartite motif (TRIM)-containing proteins has been shown to be closely related to tumor progression. However, the function of TRIM46 in lung cancer is largely unknown. Here, TRIM46 amplification was found in lung adenocarcinoma (LUAD) tissues and TRIM46 amplification was significantly associated with a poor survival rate. Overexpression of wild type TRIM46 increased the proliferation of LUAD cells and glycolysis, promoted xenografts growth, and enhanced cisplatin (DDP) resistance of LUAD cells via increased ubiquitination of pleckstrin homology domain leucine-rich repeat protein phosphatase 2 (PHLPP2) and upregulation of p-AKT. In contrast, overexpression of RING-mutant TRIM46 did not show any effects, suggesting the function of TRIM46 was dependent on the E3 ligase activity. Furthermore, we found that TRIM46 promoted LUAD cell proliferation and DDP resistance by enhancing glycolysis. PHLPP2 overexpression reversed the effects of TRIM46 overexpression. Amplification of TRIM46 also promoted LUAD growth and enhanced its DDP resistance in a patient-derived xenograft (PDX) model. In conclusion, our data highlight the importance of TRIM46/PHLPP2/AKT signaling in lung cancer and provide new insights into therapeutic strategies for lung cancer.
Collapse
|
181
|
Zhu J, Guo T, Wang Z, Zhao Y. Triggered azobenzene-based prodrugs and drug delivery systems. J Control Release 2022; 345:475-493. [PMID: 35339578 DOI: 10.1016/j.jconrel.2022.03.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 12/18/2022]
Abstract
Azobenzene-based molecules show unique trans-cis isomerization upon ultraviolet light irradiation, which induce the change of polarity, crystallinity, stability, and binding affinity with pharmacological target. Moreover, azobenzene is the substrate of azoreductase that is often overexpressed in many pathological sites, e.g. hypoxic solid tumor. Therefore, azobenzene can be a multifunctional molecule in material science, pharmaceutical science and biomedicine because of its sensitivity to light, hypoxia and certain enzymes, hence showing potential application in site-specific smart therapy. Herein we focus on the employment of azobenzene and its derivatives for engineering triggered prodrug and drug delivery systems, and provide an overview of photoswitchable azo-based prodrugs, the associated problems regarding ultraviolet light and reversible isomerization, as well as the potential solutions. We also present the advance of azo-bearing delivery vehicles wherein azobenzene act as the linker, capping agent, and building block, and discuss the corresponding mechanisms for controlled cargo release, endocytosis enhancement and sensitization of free radical cancer therapy.
Collapse
Affiliation(s)
- Jundong Zhu
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
182
|
Khan SU, Pathania AS, Wani A, Fatima K, Mintoo MJ, Hamza B, Paddar MA, Bhumika W, Anand LK, Maqbool MS, Mir SA, Kour J, Venkateswarlu V, Mondhe DM, Sawant SD, Malik F. Activation of lysosomal mediated cell death in the course of autophagy by mTORC1 inhibitor. Sci Rep 2022; 12:5052. [PMID: 35322026 PMCID: PMC8943151 DOI: 10.1038/s41598-022-07955-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 01/10/2022] [Indexed: 01/13/2023] Open
Abstract
Lysosomal biogenesis plays a vital role in cell fate. Under certain conditions, excessive lysosomal biogenesis leads to susceptibility for lysosomal membrane permeabilization resulting in various pathological conditions including cell death. In cancer cells apoptosis machinery becomes dysregulated during the course of treatment, thus allows cancer cells to escape apoptosis. So it is therefore imperative to identify cytotoxic agents that exploit non-apoptotic mechanisms of cell death. Our study showed that pancreatic cancer cells treated with SDS-203 triggered an incomplete autophagic response and a nuclear translocation of transcriptional factor TFEB. This resulted in abundant biosynthesis and accumulation of autophagosomes and lysosomes into the cells leading to their death. It was observed that the silencing of autophagy genes didn’t alter the cell fate, whereas siRNA-mediated silencing of TFEB subdued SDS-203 mediated lysosomal biogenesis and associated cell death. Further mouse tumors treated with SDS-203 showed a significant reduction in tumor burden and increased expression of lysosomal markers. Taken together this study demonstrates that SDS-203 treatment triggers non-apoptotic cell death in pancreatic cancer cells through a mechanism of lysosome over accumulation.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Anup Singh Pathania
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Abubakar Wani
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Kaneez Fatima
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Mubashir Javed Mintoo
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Baseerat Hamza
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India
| | - Masroor Ahmad Paddar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Wadhwa Bhumika
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Loveleena Kour Anand
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Mir Shahid Maqbool
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Sameer Ahmad Mir
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Jaspreet Kour
- Medicinal Chemistry, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Vunnam Venkateswarlu
- Medicinal Chemistry, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Dilip Manikrao Mondhe
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India
| | - Sanghapal D Sawant
- Medicinal Chemistry, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Fayaz Malik
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, Jammu and Kashmir, 190005, India.
| |
Collapse
|
183
|
Rezaei A, Asgari S, Komijani S, Sadat SN, Sabatier JM, Nasrabadi D, Pooshang Bagheri K, Shahbazzadeh D, Akbari Eidgahi MR, De Waard M, Mirzahoseini H. Discovery of Leptulipin, a New Anticancer Protein from theIranian Scorpion, Hemiscorpius lepturus. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072056. [PMID: 35408455 PMCID: PMC9000277 DOI: 10.3390/molecules27072056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022]
Abstract
Cancer is one of the leading causes of mortality in the world. Unfortunately, the present anticancer chemotherapeutics display high cytotoxicity. Accordingly, the discovery of new anticancer agents with lower side effects is highly necessitated. This study aimed to discover an anticancer compound from Hemiscorpius lepturus scorpion venom. Bioactivity-guided chromatography was performed to isolate an active compound against colon and breast cancer cell lines. 2D electrophoresis and MALDI-TOF were performed to identify the molecule. A partial protein sequence was obtained by mass spectrometry, while the full-length was deciphered using a cDNA library of the venom gland by bioinformatics analyses and was designated as leptulipin. The gene was cloned in pET-26b, expressed, and purified. The anticancer effect and mechanism action of leptulipin were evaluated by MTT, apoptosis, and cell cycle assays, as well as by gene expression analysis of apoptosis-related genes. The treated cells displayed inhibition of cell proliferation, altered morphology, DNA fragmentation, and cell cycle arrest. Furthermore, the treated cells showed a decrease in BCL-2 expression and an increase in Bax and Caspase 9 genes. In this study, we discovered a new anticancer protein from H. lepturus scorpion venom. Leptulipin showed significant anticancer activity against breast and colon cancer cell lines.
Collapse
Affiliation(s)
- Ali Rezaei
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 3514799422, Iran; (A.R.); (D.N.)
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
| | - Saeme Asgari
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran 5157944533, Iran;
| | - Samira Komijani
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
| | - Seyedeh Narjes Sadat
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
| | - Jean-Marc Sabatier
- Institute of NeuroPhysiopathology (INP), Faculté de Pharmacie, Université D’Aix-Marseille, UMR 7051, 27 Bd Jean Moulin, CEDEX 05, 13385 Marseille, France;
| | - Davood Nasrabadi
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 3514799422, Iran; (A.R.); (D.N.)
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
- Correspondence: (K.P.B.); (M.R.A.E.); (M.D.W.); (H.M.)
| | - Delavar Shahbazzadeh
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
| | - Mohammad Reza Akbari Eidgahi
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 3514799422, Iran; (A.R.); (D.N.)
- Correspondence: (K.P.B.); (M.R.A.E.); (M.D.W.); (H.M.)
| | - Michel De Waard
- L’Institut du Thorax, INSERM, CNRS, University of Nantes, 44000 Nantes, France
- LabEx “Ion Channels, Science & Therapeutics”, 65560 Valbonne, France
- Smartox Biotechnology, 6 Rue Des Platanes, 38120 Saint-Egrève, France
- Correspondence: (K.P.B.); (M.R.A.E.); (M.D.W.); (H.M.)
| | - Hasan Mirzahoseini
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran; (S.K.); (S.N.S.); (D.S.)
- Correspondence: (K.P.B.); (M.R.A.E.); (M.D.W.); (H.M.)
| |
Collapse
|
184
|
Cyran AM, Zhitkovich A. Heat Shock Proteins and HSF1 in Cancer. Front Oncol 2022; 12:860320. [PMID: 35311075 PMCID: PMC8924369 DOI: 10.3389/fonc.2022.860320] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/07/2022] [Indexed: 12/23/2022] Open
Abstract
Fitness of cells is dependent on protein homeostasis which is maintained by cooperative activities of protein chaperones and proteolytic machinery. Upon encountering protein-damaging conditions, cells activate the heat-shock response (HSR) which involves HSF1-mediated transcriptional upregulation of a group of chaperones - the heat shock proteins (HSPs). Cancer cells experience high levels of proteotoxic stress due to the production of mutated proteins, aneuploidy-induced excess of components of multiprotein complexes, increased translation rates, and dysregulated metabolism. To cope with this chronic state of proteotoxic stress, cancers almost invariably upregulate major components of HSR, including HSF1 and individual HSPs. Some oncogenic programs show dependence or coupling with a particular HSR factor (such as frequent coamplification of HSF1 and MYC genes). Elevated levels of HSPs and HSF1 are typically associated with drug resistance and poor clinical outcomes in various malignancies. The non-oncogene dependence ("addiction") on protein quality controls represents a pancancer target in treating human malignancies, offering a potential to enhance efficacy of standard and targeted chemotherapy and immune checkpoint inhibitors. In cancers with specific dependencies, HSR components can serve as alternative targets to poorly druggable oncogenic drivers.
Collapse
Affiliation(s)
- Anna M Cyran
- Legoretta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Anatoly Zhitkovich
- Legoretta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| |
Collapse
|
185
|
Homayoonfal M, Asemi Z, Yousefi B. Potential anticancer properties and mechanisms of thymoquinone in osteosarcoma and bone metastasis. Cell Mol Biol Lett 2022; 27:21. [PMID: 35236304 PMCID: PMC8903697 DOI: 10.1186/s11658-022-00320-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
Despite great advances, therapeutic approaches of osteosarcoma, the most prevalent class of preliminary pediatric bone tumors, as well as bone-related malignancies, continue to demonstrate insufficient adequacy. In recent years, a growing trend toward applying natural bioactive compounds, particularly phytochemicals, as novel agents for cancer treatment has been observed. Bioactive phytochemicals exert their anticancer features through two main ways: they induce cytotoxic effects against cancerous cells without having any detrimental impact on normal cell macromolecules such as DNA and enzymes, while at the same time combating the oncogenic signaling axis activated in tumor cells. Thymoquinone (TQ), the most abundant bioactive compound of Nigella sativa, has received considerable attention in cancer treatment owing to its distinctive properties, including apoptosis induction, cell cycle arrest, angiogenesis and metastasis inhibition, and reactive oxygen species (ROS) generation, along with inducing immune system responses and reducing side effects of traditional chemotherapeutic drugs. The present review is focused on the characteristics and mechanisms by which TQ exerts its cytotoxic effects on bone malignancies.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
186
|
Elgiushy HR, Mohamed SH, Taha H, Sawaf H, Hassan Z, Abou-Taleb NA, El-labbad EM, Hassan AS, Abouzid KA, Hammad SF. Identification of a promising hit from a new series of pyrazolo[1,5-a]pyrimidine based compounds as a potential anticancer agent with potent CDK1 inhibitory and pro-apoptotic properties through a multistep in vitro assessment. Bioorg Chem 2022; 120:105646. [DOI: 10.1016/j.bioorg.2022.105646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022]
|
187
|
Terrasson J, Seigneur É, Rault A, El Mellah L, Dolbeault S, Brédart A. The announcement of treatment resistance from the pediatric oncologist's point of view: a qualitative study. Pediatr Hematol Oncol 2022; 39:132-144. [PMID: 34392770 DOI: 10.1080/08880018.2021.1956030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Breaking bad news in pediatric oncology covers widely diverse clinical situations. The aim of this study was to highlight the specificities of the announcement of treatment resistance as perceived by pediatric oncologists, particularly in comparison with the disclosure of a cancer diagnosis. Semi-structured interviews were conducted in two pediatric oncology departments in France, with 15 pediatric oncologists (66.7% were women, aged 44.7 years on average). Interviews were audio-recorded and transcribed and a thematic content analysis was conducted. Most pediatric oncologists reported emotional difficulties in announcing treatment resistance. Some of them mentioned a personal need to accept resistance to treatment and to mourn the child's chances of recovery, and reported feelings of medical failure. This disclosure was considered more difficult than the announcement of the cancer diagnosis because it was associated with less optimism and more complex and fewer therapeutic options. The attachment bond created with families in the course of treatment seemed to exacerbate the emotional difficulties associated with this announcement. In conclusion, resistance to treatment has an impact on prognosis. It makes it more uncertain. Its announcement for pediatric oncologists is a turning point that affects their initial optimistic perspective. Their emotional difficulties are accentuated by the attachment that has been created with the families. Focusing on difficulties experienced by pediatricians could help to improve parent-pediatrician communication.
Collapse
Affiliation(s)
- Johanna Terrasson
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France
| | - Étienne Seigneur
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France.,Institut Curie, SIREDO Oncology Center, Paris, France
| | - Aude Rault
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France
| | - Leïla El Mellah
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France
| | - Sylvie Dolbeault
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France.,Research Center in Epidemiology and Health Population, INSERM, U1018, Paris-Saclay University, Villejuif, France
| | - Anne Brédart
- Psycho-Oncology Unit, Institut Curie, PSL Research University, Paris, France.,Psychopathology and Health Process Laboratory, University of Paris, Boulogne-Billancourt, France
| |
Collapse
|
188
|
Lei Y, Chen L, Liu J, Zhong Y, Deng L. The MicroRNA-Based Strategies to Combat Cancer Chemoresistance via Regulating Autophagy. Front Oncol 2022; 12:841625. [PMID: 35211417 PMCID: PMC8861360 DOI: 10.3389/fonc.2022.841625] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance frequently occurs in cancer treatment, which results in chemotherapy failure and is one of the most leading causes of cancer-related death worldwide. Understanding the mechanism of chemoresistance and exploring strategies to overcome chemoresistance have become an urgent need. Autophagy is a highly conserved self-degraded process in cells. The dual roles of autophagy (pro-death or pro-survival) have been implicated in cancers and chemotherapy. MicroRNA (miRNA) is a class of small non-coding molecules that regulate autophagy at the post-transcriptional level in cancer cells. The association between miRNAs and autophagy in cancer chemoresistance has been emphasized. In this review, we focus on the dual roles of miRNA-mediated autophagy in facilitating or combating chemoresistance, aiming to shed lights on the potential role of miRNAs as targets to overcome chemoresistance.
Collapse
Affiliation(s)
- Yuhe Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lei Chen
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Junshan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yinqin Zhong
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
189
|
Wang HT, Ju J, Wang SC, Zhang YH, Liu CY, Wang T, Yu X, Wang F, Cheng XR, Wang K, Chen ZY. Insights Into Ferroptosis, a Novel Target for the Therapy of Cancer. Front Oncol 2022; 12:812534. [PMID: 35280796 PMCID: PMC8914339 DOI: 10.3389/fonc.2022.812534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death (PCD) characterized by an excess iron accumulation and subsequent unbalanced redox states. Ferroptosis is different from the already reported PCD and has unique morphological features and biochemical processes. Ferroptosis was first elaborated by Brent R. Stockwell’s lab in 2012, in which small molecules erastin and RSL-3 induce PCD in Ras mutant cell lines. Ferroptosis involves various physiological processes and occurrence of disease and especially shows strong potential in cancer treatment. Development of small molecule compounds based on Stockwell’s research was found to kill cancer cells, and some FDA-approved drugs were discovered to result in ferroptosis of cancer cells. Radiotherapy and checkpoint therapy have been widely used as a treatment for many types of cancer. Recently, some papers have reported that chemotherapy, radiotherapy, and checkpoint therapy induce ferroptosis of cancer cells, which provides new strategies for cancer treatment. Nevertheless, the limitless proliferation of tumor cells and the lack of cell death mechanisms are important reasons for drug resistance for tumor therapy. Therefore, we reviewed the molecular mechanism of ferroptosis and sensitivity to ferroptosis of different cancer cells and tumor treatment strategy.
Collapse
Affiliation(s)
- Hong-Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- Science and Technology Department, Qingdao University, Qingdao, China
| | - Jie Ju
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Shao-Cong Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cui-Yun Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Tao Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue Yu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Fei Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue-Ru Cheng
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| | - Zhao-Yang Chen
- Cardiology Department, Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, China
- *Correspondence: Kun Wang, ; Zhao-Yang Chen,
| |
Collapse
|
190
|
Kim MJ, Kawk HW, Kim SH, Lee HJ, Seo JW, Lee CY, Kim YM. The p53-Driven Anticancer Effect of Ribes fasciculatum Extract on AGS Gastric Cancer Cells. Life (Basel) 2022; 12:life12020303. [PMID: 35207590 PMCID: PMC8876336 DOI: 10.3390/life12020303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer metastasis is directly related to the survival rate of cancer patients. Although cancer metastasis proceeds by the movement of cancer cells, it is fundamentally caused by its resistance to anoikis, a mechanism of apoptosis caused by the loss of adhesion of cancer cells. Therefore, it was found that inhibiting cancer migration and reducing anoikis resistance are important for cancer suppression, and natural compounds can effectively control it. Among them, Ribes fasciculatum, which has been used as a medicinal plant, was confirmed to have anticancer potential, and experiments were conducted to prove various anticancer effects by extracting Ribes fasciculatum (RFE). Through various experiments, it was observed that RFE induces apoptosis of AGS gastric cancer cells, arrests the cell cycle, induces oxidative stress, and reduces mobility. It was also demonstrated that anoikis resistance was attenuated through the downregulation of proteins, such as epidermal growth factor receptor (EGFR). Moreover, the anticancer effect of RFE depends upon the increase in p53 expression, suggesting that RFE is suitable for the development of p53-targeted anticancer materials. Moreover, through xenotransplantation, it was found that the anticancer effect of RFE confirmed in vitro was continued in vivo.
Collapse
|
191
|
Sun CJ, Hu RY, Li ZC, Jin L, Lu H, He ZX, Shu LP. An engineered abcb4 expression model reveals the central role of NF-κB in the regulation of drug resistance in zebrafish. Drug Dev Res 2022; 83:927-939. [PMID: 35165900 DOI: 10.1002/ddr.21917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
Abstract
Multi-drug resistance (MDR) is a phenomenon that tumor cells are exposed to a chemotherapeutic drug for a long time and then develop resistance to a variety of other anticancer drugs with different structures and different mechanisms. The in vitro studies of tumor cell lines cannot systematically reflect the role of MDR gene in vivo, and the cost of in vivo studies of transgenic mice as animal models is high. Given the myriad merits of zebrafish relative to other animal models, we aimed to establish a screening system using zebrafish stably expressing ATP-binding cassette (ATP-cassette) superfamily transporters and unveil the potential regulatory mechanism. We first used the Tol2-mediated approach to construct a Tg (abcb4:EGFP) transgenic zebrafish line with ATP-binding cassette (ABC) subfamily B member 4 (abcb4) gene promoter to drive EGFP expression. The expression levels of abcb4 and EGFP were significantly increased when Tg(abcb4:EGFP) transgenic zebrafish embryos were exposed to doxorubicin (DOX) or vincristine (VCR), and the increases were accompanied by a marked decreased accumulation of rhodamine B (RhB) in embryos, indicating a remarkable increase in DOX or VCR efflux. Mechanistically, Akt and Erk signalings were activated upon the treatment with DOX or VCR. With the application of Akt and Erk inhibitors, drug resistance was reversed with differing responsive effects. Notably, downstream NF-κB played a central role in the regulation of abcb4-mediated drug resistance. Taken together, the data indicate that the engineered Tg(abcb4:EGFP) transgenic zebrafish model is a new platform for screening drug resistance in vivo, which may facilitate and accelerate the process of drug development.
Collapse
Affiliation(s)
- Cong-Jie Sun
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Province Key Laboratory for Regenerative Medicine, Department of Immunology, Department of Pediatrics, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Rong-Yin Hu
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Province Key Laboratory for Regenerative Medicine, Department of Immunology, Department of Pediatrics, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Zhi-Cao Li
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Province Key Laboratory for Regenerative Medicine, Department of Immunology, Department of Pediatrics, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Lu Jin
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Province Key Laboratory for Regenerative Medicine, Department of Immunology, Department of Pediatrics, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - He Lu
- National Institute of Health and Medical Research, Medical Research Unit 942/Paris University 7 and 13, Avicenne Hospital, Bobigny, France
| | - Zhi-Xu He
- National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Province Key Laboratory for Regenerative Medicine, Department of Immunology, Department of Pediatrics, Guizhou Medical University, Guiyang, China.,Department of Pediatrics, Zunyi Medical University, Zunyi, China
| | - Li-Ping Shu
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China.,National & Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, State Key Laboratory of Functions and Applications of Medicinal Plants, Guiyang, China
| |
Collapse
|
192
|
Dey A, Kundu M, Das S, Jena BC, Mandal M. Understanding the function and regulation of Sox2 for its therapeutic potential in breast cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188692. [PMID: 35122882 DOI: 10.1016/j.bbcan.2022.188692] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/11/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022]
Abstract
Sox family of transcriptional factors play essential functions in development and are implicated in multiple clinical disorders, including cancer. Sox2 being their most prominent member and performing a critical role in reprogramming differentiated adult cells to an embryonic phenotype is frequently upregulated in multiple cancers. High Sox2 levels are detected in breast tumor tissues and correlate with a worse prognosis. In addition, Sox2 expression is connected with resistance to conventional anticancer therapy. Together, it can be said that inhibiting Sox2 expression can reduce the malignant features associated with breast cancer, including invasion, migration, proliferation, stemness, and chemoresistance. This review highlights the critical roles played by the Sox gene family members in initiating or suppressing breast tumor development, while primarily focusing on Sox2 and its role in breast tumor initiation, maintenance, and progression, elucidates the probable mechanisms that control its activity, and puts forward potential therapeutic strategies to inhibit its expression.
Collapse
Affiliation(s)
- Ankita Dey
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur (IIT Kharagpur), Kharagpur, West Bengal, India..
| | - Moumita Kundu
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur (IIT Kharagpur), Kharagpur, West Bengal, India..
| | - Subhayan Das
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur (IIT Kharagpur), Kharagpur, West Bengal, India..
| | - Bikash Chandra Jena
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur (IIT Kharagpur), Kharagpur, West Bengal, India..
| | - Mahitosh Mandal
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur (IIT Kharagpur), Kharagpur, West Bengal, India..
| |
Collapse
|
193
|
Liu Y, Khan S, Li L, ten Hagen TL, Falahati M. Molecular mechanisms of thyroid cancer: A competing endogenous RNA (ceRNA) point of view. Biomed Pharmacother 2022; 146:112251. [DOI: 10.1016/j.biopha.2021.112251] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 12/25/2022] Open
|
194
|
Khan NH, Mir M, Qian L, Baloch M, Ali Khan MF, Rehman AU, Ngowi EE, Wu DD, Ji XY. Skin cancer biology and barriers to treatment: Recent applications of polymeric micro/nanostructures. J Adv Res 2022; 36:223-247. [PMID: 35127174 PMCID: PMC8799916 DOI: 10.1016/j.jare.2021.06.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background Skin cancer has been the leading type of cancer worldwide. Melanoma and non-melanoma skin cancers are now the most common types of skin cancer that have been reached to epidemic proportion. Based on the rapid prevalence of skin cancers, and lack of efficient drug delivery systems, it is essential to surge the possible ways to prevent or cure the disease. Aim of review Although surgical modalities and therapies have been made great progress in recent years, however, there is still an urgent need to alleviate its increased burden. Hence, understanding the precise pathophysiological signaling mechanisms and all other factors of such skin insults will be beneficial for the development of more efficient therapies. Key scientific concepts of review In this review, we explained new understandings about onset and development of skin cancer and described its management via polymeric micro/nano carriers-based therapies, highlighting the current key bottlenecks and future prospective in this field. In therapeutic drug/gene delivery approaches, polymeric carriers-based system is the most promising strategy. This review discusses that how polymers have successfully been exploited for development of micro/nanosized systems for efficient delivery of anticancer genes and drugs overcoming all the barriers and limitations associated with available conventional therapies. In addition to drug/gene delivery, intelligent polymeric nanocarriers platforms have also been established for combination anticancer therapies including photodynamic and photothermal, and for theranostic applications. This portfolio of latest approaches could promote the blooming growth of research and their clinical availability.
Collapse
Key Words
- 5-ALA, 5-aminolevulinic acid
- 5-FU, 5-fluorouracil
- AIDS, Acquired immune deficiency syndrome
- BCC, Basal cell carcinoma
- BCCs, Basal cell carcinomas
- Basal cell carcinoma
- CREB, response element-binding protein
- DDS, Drug delivery system
- DIM-D, Di indolyl methane derivative
- Drug delivery
- GNR-PEG-MN, PEGylated gold nanorod microneedle
- Gd, Gadolinium
- Gene delivery
- HH, Hedgehog
- HPMC, Hydroxypropyl methylcellulose
- IPM, Isopropyl myristate
- MCIR, Melanocortin-1 receptor
- MNPs, Magnetic nanoparticle
- MNs, Microneedles
- MRI, Magnetic Resonance Imaging
- MSC, Melanoma skin cancer
- Microneedles
- Mn, Manganese
- NMSC, Non melanoma skin cancer
- NPs, Nano Particles
- OTR, Organ transplant recipients
- PAMAM, Poly-amidoamines
- PAN, Polyacrylonitrile
- PATCH1, Patch
- PCL, Poly (ε-caprolactone)
- PDT, Photodynamic therapy
- PEG, Polyethylene glycol
- PLA, Poly lactic acid
- PLA-HPG, Poly (d-l-lactic acid)-hyperbranched polyglycerol
- PLGA, Poly (lactide-co-glycolide) copolymers
- PLL, Poly (L-lysine)
- Polymeric nanocarriers
- QDs, Quantum dots
- SC, Skin cancer
- SCC, Squamous cell Carcinoma
- SMO, Smoothen
- SPIO, Superparamagnetic iron oxide
- Squamous cell carcinoma
- UV, Ultra Violet
- cAMP, Cyclic adenosine monophosphate
- dPG, Dendritic polyglycerol
- hTERT, Human telomerase reverse transcriptase
Collapse
Affiliation(s)
- Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Life Sciences. Henan University, Kaifeng, Henan 475004, China
| | - Maria Mir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Lei Qian
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Mahnoor Baloch
- School of Natural Sciences, National University of Science and Technology, Islamabad 44000, Pakistan
| | - Muhammad Farhan Ali Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim-ur- Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- Department of Biological Sciences, Faculty of Sciences, Dar es Salaam University College of Education, Dar es Salaam 2329, Tanzania
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| |
Collapse
|
195
|
Tamtaji OR, Derakhshan M, Rashidi Noshabad FZ, Razaviyan J, Hadavi R, Jafarpour H, Jafari A, Rajabi A, Hamblin MR, Mahabady MK, Taghizadieh M, Mirzaei H. Non-Coding RNAs and Brain Tumors: Insights Into Their Roles in Apoptosis. Front Cell Dev Biol 2022; 9:792185. [PMID: 35111757 PMCID: PMC8801811 DOI: 10.3389/fcell.2021.792185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/08/2021] [Indexed: 12/18/2022] Open
Abstract
A major terrifying ailment afflicting the humans throughout the world is brain tumor, which causes a lot of mortality among pediatric and adult solid tumors. Several major barriers to the treatment and diagnosis of the brain tumors are the specific micro-environmental and cell-intrinsic features of neural tissues. Absence of the nutrients and hypoxia trigger the cells' mortality in the core of the tumors of humans' brains: however, type of the cells' mortality, including apoptosis or necrosis, has been not found obviously. Current studies have emphasized the non-coding RNAs (ncRNAs) since their crucial impacts on carcinogenesis have been discovered. Several investigations suggest the essential contribution of such molecules in the development of brain tumors and the respective roles in apoptosis. Herein, we summarize the apoptosis-related non-coding RNAs in brain tumors.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Derakhshan
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Javad Razaviyan
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Razie Hadavi
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Jafarpour
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Rajabi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
196
|
Brédart A, Rault A, Terrasson J, Seigneur E, De Koning L, Hess E, Savignoni A, Cottu P, Pierga JY, Piperno-Neumann S, Rodrigues M, Bouleuc C, Dolbeault S. Helping Patients Communicate With Oncologists When Cancer Treatment Resistance Occurs to Develop, Test, and Implement a Patient Communication Aid: Sequential Collaborative Mixed Methods Study. JMIR Res Protoc 2022; 11:e26414. [PMID: 35019850 PMCID: PMC8792782 DOI: 10.2196/26414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Most cancer-related deaths result from disseminated diseases that develop resistance to anticancer treatments. Inappropriate communication in this challenging situation may result in unmet patient information and support needs. Patient communication aids such as question prompt lists (QPLs) may help. OBJECTIVE This study aims to develop and pilot-test a specific QPL in the following two contrasting clinical contexts in France after cancer resistance has developed: triple-negative and luminal B metastatic breast cancer (MBC) and metastatic uveal melanoma (MUM). METHODS A sequential study design with a mixed methods collaborative approach will be applied. The first step aims to build a specific QPL. Step 1a will explore oncologist-patient communication issues from oncology professionals' interviews (n=20 approximately). Step 1b will appraise information and support needs experienced by patients with MBC or MUM both quantitatively (n=80) and qualitatively (n=40 approximately). These data will be used to develop and pilot-test a QPL specific to patients with cancer experiencing initial or acquired resistance to treatment. We expect to obtain a core QPL that comprises questions and concerns commonly expressed by patients with resistant cancer and is complemented by specific issues for either MBC or MUM cancer sites. In step 1c, 2 focus groups of patients with any type of metastatic cancer (n=4) and health care professionals (n=4) will be conducted to revise the content of a preliminary QPL and elaborate an acceptable and feasible clinical implementation. In step 1d, the content of the QPL version 1 and implementation guidance will be validated using a Delphi process. Step 2 will pilot-test the QPL version 1 in real practice with patients with MBC or MUM (n=80). Clinical utility will be assessed by comparing responses to questionnaires administered in step 1b (QPL-naive historical control group) and step 2 (QPL intervention group). RESULTS This study received grants in March and December 2019 and was approved by the French national ethics committee in July 2019. As of October 2021, interviews with oncology professionals have been conducted and analyzed (N=26 to reach saturation), and 39 and 27 patients with MBC and MUM, respectively, have been recruited. CONCLUSIONS A clinically and culturally tailored QPL is expected to facilitate patients' participation in consultations, improve oncologists' responses to patients' information and support needs, and thus foster patients' psychological adjustment to the diagnosis and follow-up of cancer resistance to treatment. TRIAL REGISTRATION ClinicalTrials.gov NCT04118062; http://clinicaltrials.gov/ct2/show/NCT04118062. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/26414.
Collapse
Affiliation(s)
- Anne Brédart
- Psycho-Oncology Unit, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
- Psychopathology and Health Process, Paris University, Boulogne Billancourt, France
| | - Aude Rault
- Psycho-Oncology Unit, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Johanna Terrasson
- Psycho-Oncology Unit, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Etienne Seigneur
- Psycho-Oncology Unit, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Leanne De Koning
- Research Centre, Paris Sciences et Lettres Research University, Institut Curie, Paris, France
| | - Elisabeth Hess
- Research Centre, Paris Sciences et Lettres Research University, Institut Curie, Paris, France
| | - Alexia Savignoni
- Direction Recherche Ensemble Hospitalier, Data Management Unit, Biometry Department, Institut Curie, Saint-Cloud, France
| | - Paul Cottu
- Medical Oncology Department, Institut Curie, Paris, France
| | - Jean-Yves Pierga
- Medical Oncology Department, Institut Curie, Paris, France
- Faculty of medicine, Paris University, Paris, France
| | | | | | - Carole Bouleuc
- Département Interdisciplinaire de Soins de Support pour le Patient en Oncologie, Department of Supportive Care, Institut Curie, Paris, France
| | - Sylvie Dolbeault
- Psycho-Oncology Unit, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
- Research Centre in Epidemiology and Population Health (CESP), INSERM, U1018, University Paris-Sud, Villejuif, France
| |
Collapse
|
197
|
Maleki Dana P, Sadoughi F, Asemi Z, Yousefi B. The role of polyphenols in overcoming cancer drug resistance: a comprehensive review. Cell Mol Biol Lett 2022; 27:1. [PMID: 34979906 PMCID: PMC8903685 DOI: 10.1186/s11658-021-00301-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapeutic drugs are used to treat advanced stages of cancer or following surgery. However, cancers often develop resistance against drugs, leading to failure of treatment and recurrence of the disease. Polyphenols are a family of organic compounds with more than 10,000 members which have a three-membered flavan ring system in common. These natural compounds are known for their beneficial properties, such as free radical scavenging, decreasing oxidative stress, and modulating inflammation. Herein, we discuss the role of polyphenols (mainly curcumin, resveratrol, and epigallocatechin gallate [EGCG]) in different aspects of cancer drug resistance. Increasing drug uptake by tumor cells, decreasing drug metabolism by enzymes (e.g. cytochromes and glutathione-S-transferases), and reducing drug efflux are some of the mechanisms by which polyphenols increase the sensitivity of cancer cells to chemotherapeutic agents. Polyphenols also affect other targets for overcoming chemoresistance in cancer cells, including cell death (i.e. autophagy and apoptosis), EMT, ROS, DNA repair processes, cancer stem cells, and epigenetics (e.g. miRNAs).
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
198
|
Rault A, Terrasson J, Dolbeault S, Brédart A. Réflexions cliniques et éthiques à partir d’une recherche menée en psycho-oncologie sur la communication lors de l’annonce de l’échec d’un traitement antitumoral. PSYCHO-ONCOLOGIE 2022. [DOI: 10.3166/pson-2022-0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cet article rend compte des divers questionnements cliniques et éthiques apparus au cours de notre recherche en psycho-oncologie. Celle-ci porte sur la communication entre les oncologues et leurs patients, et les pédiatres-oncologues et les parents d’enfants malades à un moment particulier de la prise en charge : l’annonce de la résistance au traitement antitumoral et l’absence d’alternative curative connue. La mise en place d’une telle étude nécessite de prendre en compte les aspects émotionnels et psychologiques en jeu. Dans un premier temps, nous présentons les interrogations portant sur le recrutement des participants : comment et quand les contacter ? Par la suite, nous questionnons le potentiel impact de cette recherche pour les participants : quels mots utiliser et quelles questions aborder ? Enfin, nous revenons sur les précautions à prendre dans l’interprétation et la transmission des résultats : quelles sont les incidences des choix méthodologiques ? À chaque étape, nos réflexions sont illustrées par des exemples concrets. Cet article met en exergue la nécessité de la réflexion à la fois clinique et éthique dans le domaine de la recherche en psycho-oncologie.
Collapse
|
199
|
Ren X, Wang Y, Jia L, Guo X, He X, Zhao Z, Gao D, Yang Z. Intelligent Nanomedicine Approaches Using Medical Gas-Mediated Multi-Therapeutic Modalities Against Cancer. J Biomed Nanotechnol 2022; 18:24-49. [PMID: 35180898 DOI: 10.1166/jbn.2022.3224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The emerging area of gas-mediated cancer treatment has received widespread attention in the medical community. Featuring unique physical, chemical, and biological properties, nanomaterials can facilitate the delivery and controllable release of medicinal gases at tumor sites, and also serve as ideal platforms for the integration of other therapeutic modalities with gas therapy to augment cancer therapeutic efficacy. This review presents an overview of anti-cancer mechanisms of several therapeutic gases: nitric oxide (NO), hydrogen sulfide (H₂S), carbon monoxide (CO), oxygen (O₂), and hydrogen (H₂). Controlled release behaviors of gases under different endogenous and exogenous stimuli are also briefly discussed, followed by their synergistic effects with different therapeutic modes. Moreover, the potential challenges and future prospects regarding gas therapy based on nanomaterials are also described, aiming to facilitate the advancement of gas therapeutic nanomedicine in new frontiers for highly efficient cancer treatment.
Collapse
Affiliation(s)
- Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoqing Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhipeng Zhao
- School of Physical Education, Xizang Minzu University, Xianyang, 712000, Shaanxi, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
200
|
Ding H, Yu X, Yan Z. Ailanthone suppresses the activity of human colorectal cancer cells through the STAT3 signaling pathway. Int J Mol Med 2021; 49:21. [PMID: 34958109 PMCID: PMC8722763 DOI: 10.3892/ijmm.2021.5076] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022] Open
Abstract
Ailanthone (AIL) is a major quassinoid extracted from the Chinese medicinal herb, Ailanthus altissima, which has been reported to exert anti-proliferative effects on various cancer cells. The present study aimed to investigate the anti-tumor effects of AIL on HCT116 and SW620 colon cancer cells, and to analyze the underlying molecular mechanisms. CCK-8 assay was used to detect cell viability. Furthermore, colony formation and Transwell assays, and flow cytometry were used to examine the effects of AIL on cell proliferation, apoptosis and migration. Finally, the expression levels of cell cycle control proteins, and caspase and Bcl-2 family-related proteins involved in the regulation of apoptosis, as well as those of cell migration- and pathway-related proteins were examined using western blot analysis. Reverse transcription-quantitative PCR was used to quantitatively analyze the changes in the JAK and STAT3 gene levels in each group. The in vitro cell function tests revealed that AIL inhibited the proliferation and migration, and induced the apoptosis and cell cycle arrest of HCT116 and SW620 cells. It was further found exerted these effects via the JAK/STAT3 signaling pathway, as well as through caspase and Bcl-2 family proteins. On the whole, the present study demonstrates that AIL suppresses the activity of colon cancer cells via the STAT3 pathway.
Collapse
Affiliation(s)
- Haixiang Ding
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Xiuchong Yu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Zhilong Yan
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|