1
|
García-Díaz N, Solli E, Hajjar E, Cornillot-Clément S, Landskron J, Ahmad R, Wei Q, Taskén K. MAPK and STAT3 Inhibitors Modulate FoxP3 Expression and Regulatory T Cell Function. Eur J Immunol 2025; 55:e202451225. [PMID: 39955647 DOI: 10.1002/eji.202451225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Regulatory T cells (Tregs) are a subset of T cells defined by the expression of Forkhead box protein P3 (FoxP3) playing a crucial role in regulating effector T cell activity. Tregs accumulate in the tumor microenvironment facilitating tumor growth. Thus, targeting FoxP3+ Tregs could improve cancer immunotherapies. Here, we conducted a high-throughput, phenotypic screening of a drug repurposing library to identify compounds downregulating FoxP3 expression in human primary T cells. We identified the tyrosine kinase inhibitor bosutinib and the STAT3 inhibitor nifuroxazide effectively downregulating FoxP3 expression. To identify more potent compounds, structural analogs of these two compounds were searched and validated. These analogs were found to reduce FoxP3 expression in a similar- or more potent manner than the original hits. All compounds inhibited Treg suppressive functions and reduced the expression of Treg activation markers. Importantly, bosutinib disrupted FAK and CaMKII signaling more potently in Tregs, whilst nifuroxazide and its analog NA16 targeted STAT3 protein levels more effectively in Tregs. Additionally, bosutinib and NA16 targeted effector Tregs more effectively than other Treg subsets. In summary, bosutinib, nifuroxazide, and their analogs inhibited FoxP3 expression, Treg suppressive abilities, and Treg activation effectively, which could serve as tools for the improvement of current cancer immunotherapies.
Collapse
Affiliation(s)
- Nuria García-Díaz
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elise Solli
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ehsan Hajjar
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Selma Cornillot-Clément
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Johannes Landskron
- Centre for Molecular Medicine, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Rafi Ahmad
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Biotechnology, University of Inland Norway, Hamar, Norway
| | - Qian Wei
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for B-cell Malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Liu Y, Xu M, Xia B, Qiao Z, He Y, Liu Y, Pan Z, Zhang C, Peng H, Liang X, Zhao P, Tang H, Zheng X. Nifuroxazide Prevents Chikungunya Virus Infection Both In Vitro and In Vivo via Suppressing Viral Replication. Viruses 2024; 16:1322. [PMID: 39205296 PMCID: PMC11360488 DOI: 10.3390/v16081322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging arbovirus causing disease on a global scale, and the potential for its epidemics remains high. CHIKV has caused millions of cases and heavy economic burdens around the world, while there are no available approved antiviral therapies to date. In this study, nifuroxazide, an FDA-approved antibiotic for acute diarrhea or colitis, was found to significantly inhibit a variety of arboviruses, although its antiviral activity varied among different target cell types. Nifuroxazide exhibited relatively high inhibitory efficiency in yellow fever virus (YFV) infection of the hepatoma cell line Huh7, tick-borne encephalitis virus (TBEV) and west nile virus (WNV) infection of the vascular endothelial cell line HUVEC, and CHIKV infection of both Huh7 cells and HUVECs, while it barely affected the viral invasion of neurons. Further systematic studies on the action stage of nifuroxazide showed that nifuroxazide mainly inhibited in the viral replication stage. In vivo, nifuroxazide significantly reduced the viral load in muscles and protected mice from CHIKV-induced footpad swelling, an inflammation injury within the arthrosis of infected mice. These results suggest that nifuroxazide has a potential clinical application as an antiviral drug, such as in the treatment of CHIKV infection.
Collapse
Affiliation(s)
- Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China; (M.X.); (X.L.)
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhuoyue Qiao
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China;
| | - Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Congcong Zhang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xuesong Liang
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China; (M.X.); (X.L.)
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xu Zheng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
3
|
Hassanein EHM, Abdel-Reheim MA, Althagafy HS, Hemeda MS, Gad RA, Abdel-Sattar AR. Nifuroxazide attenuates indomethacin-induced renal injury by upregulating Nrf2/HO-1 and cytoglobin and suppressing NADPH-oxidase, NF-κB, and JAK-1/STAT3 signals. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3985-3994. [PMID: 37994949 DOI: 10.1007/s00210-023-02851-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Indomethacin (INDO) is an NSAID with remarkable efficacy and widespread utilization for alleviating pain. Nevertheless, renal function impairment is an adverse reaction linked to INDO usage. Nifuroxazide (NFX), an oral nitrofuran antibiotic, is frequently employed as an intestinal anti-infective agent. Our study aimed to investigate the renoprotective effects of NFX against INDO-induced nephrotoxicity and explore the protection mechanisms. Four groups of rats were allocated to (I) the normal control, (II) the NFX-treated (50 mg/kg), (III) INDO control (20 mg/kg), and (IV) NFX + INDO. NFX attenuates renal impairment in INDO-induced renal injury, proved by decreasing serum levels of urea, creatinine, uric acid, and NGAL while the albumin was elevated. NFX mitigates renal oxidative stress by decreasing MDA levels and restoring the antioxidants' GSH and SOD levels mediated by upregulating Nrf2, HO-1, and cytoglobin pathways. NFX mitigated renal inflammation and effectively decreased MPO, IL-1β, and TNF-α levels in the rat's kidney mediated by significant downregulation of NADPH-oxidase and NF-κB expression and suppression of JAK-1 and STAT3 phosphorylation. NFX mitigates renal apoptosis by decreasing the expression of cleaved caspase-3 expression. In conclusion, NFX treatment prevents INDO nephrotoxicity by regulating Nrf2/HO-1, cytoglobin, NADPH-oxidase, NF-κB, and JAK-1/STAT3 signals.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62521, Egypt.
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohamed S Hemeda
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Rania A Gad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef (NUB), Beni-Suef, 62511, Egypt
| | - Asmaa Ramadan Abdel-Sattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef (NUB), Beni-Suef, 62511, Egypt
| |
Collapse
|
4
|
Salama RM, Omar MA. Anti-aging effect of nifuroxazide on skin changes of aged male rat models via modulating immunoreactivity of IL-6/NF-κB/Caspase-3. Morphologie 2023; 107:100605. [PMID: 37353466 DOI: 10.1016/j.morpho.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/25/2023]
Abstract
PURPOSE To evaluate nifuroxazide's (NIF's) anti-aging characteristics in a skin-aging rat model for the first time in order to create effective preventive measures and anti-aging skin therapies. MATERIALS AND METHODS Thirty randomly selected aged male rats were assorted into three equal groups; aged control group, treated NIF I, aged rats were treated with NIF (10mg/kg, orally once daily for 14 consecutive days), and treated NIF II, aged rats were treated with NIF (20mg/kg, orally once daily for 14 consecutive days). Skin samples were obtained from the dorsal skin of the aged male rats and processed for tissue biochemical MDA, histological (Hx&E and Masson's Trichrome stains), and immunohistochemical (IL-6, NF-κB, and caspase-3) analysis. RESULTS Group I aged male albino rat skin illustrated evident distorted epidermis and dermis, disorganization of collagen fibers with marked multiple spaces of collagen fibers loss in the dermis, marked reduction of total epidermal thickness and mean area percent of collagen fibers, elevated tissue MDA level and strong positive IL-6, NF-κB, and caspase-3 immune reaction. The anti-aging benefits of NIF on skin aging are demonstrated by a marked improvement in histological alterations in the form of a well-organized epidermis and dermis, most collagen fibers in the dermis appear closely packed, significant elevation of total epidermal thickness and mean area percent of collagen fibers, a significant decrease of tissue MDA level, and immunoexpression of the inflammatory markers, IL-6, and NF-κB, and the apoptotic marker caspase-3. CONCLUSIONS This study found that group III, which received 20mg/kg of NIF, experienced more pronounced and noticeable improvements in skin aging than group II, which received 10mg/kg of NIF.
Collapse
Affiliation(s)
- R M Salama
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - M A Omar
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
5
|
Elnaggar MM, El-Yazbi AF, Belal TS, Elbardisy HM. White sustainable luminescent determination of nifuroxazide using nitrogen-sulphur co-doped carbon quantum dots nanosensor in bulk and various pharmaceutical matrices. RSC Adv 2023; 13:29830-29846. [PMID: 37829714 PMCID: PMC10566585 DOI: 10.1039/d3ra05471c] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Nifuroxazide (NFX) is an antimicrobial agent that is frequently used as an intestinal antiseptic and recently was proven to have anticancer properties. This work employs the use of nitrogen and sulphur co-doped carbon quantum dots (NSC-dots) luminescent nanoparticles to propose a highly sensitive, sustainable, white and green spectrofluorometric method for NFX detection in bulk and pharmaceutical dosage forms. l-Cysteine and citric acid were the precursors to synthesize water soluble NSC-dots by a quick and environmentally-friendly hydrothermal process. NSC-dots' native fluorescence was measured at λem = 416 nm following excitation at 345 nm. Addition of NFX resulted in quantitative quenching of NSC-dots' luminescence, which represents the principle over which this luminescent method was based. Additionally, the mechanism of fluorescence quenching was studied and discussed. The analytical procedure was validated according to the ICH-guidelines. Linear response for NFX was obtained in the dynamic range 0.04-15 μg mL-1. The estimated NFX detection and quantification limits were 0.005 and 0.015 μg mL-1, respectively. The proposed method was employed for NFX quantification into two commercial pharmaceutical dosage forms. The calculated percentage recoveries (R%), percentage relative standard deviations (RSD%), and percentage error (Er%) were satisfactory. Comparison with other reported methods showed that the proposed method is superior in several aspects. Evaluation of the whiteness of the proposed method using the RGB 12 algorithm combined with the most widely used greenness evaluation tools, the Analytical Eco-Scale and AGREE, demonstrated its superiority and sustainability over other previously published spectrofluorimetric methods for the assay of NFX in various dosage forms.
Collapse
Affiliation(s)
- Mai M Elnaggar
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt +20 34873273 +20 34871317
| | - Amira F El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt +20 34873273 +20 34871317
| | - Tarek S Belal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt +20 34873273 +20 34871317
| | - Hadil M Elbardisy
- Pharmaceutical Analysis Department, Faculty of Pharmacy, Damanhour University Damanhour 22511 Egypt
| |
Collapse
|
6
|
Li D, Liu L, Li F, Ma C, Ge K. Nifuroxazide induces the apoptosis of human non‑small cell lung cancer cells through the endoplasmic reticulum stress PERK signaling pathway. Oncol Lett 2023; 25:248. [PMID: 37153034 PMCID: PMC10161345 DOI: 10.3892/ol.2023.13834] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/26/2023] [Indexed: 05/09/2023] Open
Abstract
The aim of the present study was to investigate the molecular mechanism of nifuroxazide (NFZ) in the induction of apoptosis of NCI-H1299 human non-small cell lung cancer (NSCLC) cells through the reactive oxygen species (ROS)/Ca2+/protein kinase R-like ER kinase (PERK)-activating transcription factor 4 (ATF4)-DNA damage inducible transcript 3 (CHOP) signaling pathway. Morphological changes of cells were observed by microscopy, and the apoptosis and intracellular ROS levels of cells were observed by inverted fluorescence microscopy. Cell viability after the addition of the PERK inhibitor, GSK2606414, were detected by Cell Counting Kit-8 assay. Annexin V-FITC was used to detect cell apoptosis, Brite 670 was used to detect intracellular ROS and Fura Red AM was used to detect Ca2+ content. Western blotting was used to detect PERK, phosphorylated (P)-PERK, ATF4, CHOP, P-Janus kinase 2 and P-signal transducer and activator of transcription 3 expression levels. Compared with the dimethyl sulfoxide control group, NFZ inhibited the survival activity in the H1299 NSCLC cell line, in a time- and dose-dependent manner. However, GSK2606414 inhibited the NFZ-induced apoptosis of H1299 cells. GSK2606414 also inhibited the increase in ROS and Ca2+ in H1299 cells induced by NFZ. Western blotting results demonstrated that NFZ significantly increased the expression levels of P-PERK, ATF4 and CHOP, whereas GSK2606414 significantly reduced the NFZ-induced increase in these protein expression levels. In conclusion, NFZ may induce the apoptosis of H1299 NSCLC cells through the ROS/Ca2+/PERK-ATF4-CHOP signaling pathway.
Collapse
Affiliation(s)
- Deliang Li
- The First Clinical Medical College, Medicine College, Qingdao University, Qingdao, Shandong 266023, P.R. China
- Emergency Department, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Liping Liu
- The First Clinical Medical College, Medicine College, Qingdao University, Qingdao, Shandong 266023, P.R. China
| | - Feng Li
- Traditional Chinese Medicine Department, Zibo Wanjie Cancer Hospital, Zibo, Shandong 255200, P.R. China
| | - Chengshan Ma
- Orthopedic SurgeryDepartment, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250000, P.R. China
- Dr Chengshan Ma, Orthopedic Surgery Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia, Jinan, Shandong 250000, P.R. China, E-mail:
| | - Keli Ge
- School of Basic Medicine, Medical College, Qingdao University, Qingdao, Shandong 266023, P.R. China
- Correspondence to: Dr Keli Ge, School of Basic Medicine, Medical College, Qingdao University, 38 Dengzhou Road, Qingdao, Shandong 266023, P.R. China, E-mail:
| |
Collapse
|
7
|
Elzahhar PA, Nematalla HA, Al-Koussa H, Abrahamian C, El-Yazbi AF, Bodgi L, Bou-Gharios J, Azzi J, Al Choboq J, Labib HF, Kheir WA, Abu-Serie MM, Elrewiny MA, El-Yazbi AF, Belal ASF. Inclusion of Nitrofurantoin into the Realm of Cancer Chemotherapy via Biology-Oriented Synthesis and Drug Repurposing. J Med Chem 2023; 66:4565-4587. [PMID: 36921275 DOI: 10.1021/acs.jmedchem.2c01408] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Structural modifications of the antibacterial drug nitrofurantoin were envisioned, employing drug repurposing and biology-oriented drug synthesis, to serve as possible anticancer agents. Eleven compounds showed superior safety in non-cancerous human cells. Their antitumor efficacy was assessed on colorectal, breast, cervical, and liver cancer cells. Three compounds induced oxidative DNA damage in cancer cells with subsequent cellular apoptosis. They also upregulated the expression of Bax while downregulated that of Bcl-2 along with activating caspase 3/7. The DNA damage induced by these compounds, demonstrated by pATM nuclear shuttling, was comparable in both MCF7 and MDA-MB-231 (p53 mutant) cell lines. Mechanistic studies confirmed the dependence of these compounds on p53-mediated pathways as they suppressed the p53-MDM2 interaction. Indeed, exposure of radiosensitive prostatic cancer cells to low non-cytotoxic concentrations of compound 1 enhanced the cytotoxic response to radiation indicating a possible synergistic effect. In vivo antitumor activity was verified in an MCF7-xenograft animal model.
Collapse
Affiliation(s)
- Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22516, Egypt
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11072020, Lebanon
| | - Carla Abrahamian
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Amira F El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Jolie Bou-Gharios
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Joyce Azzi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Joelle Al Choboq
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Hala F Labib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy of Science Technology and Maritime Transport, Alexandria 21913, Egypt
| | - Wassim Abou Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Mohamed A Elrewiny
- Faculty of Pharmacy and the Research and Innovation Hub, Alamein International University, Alamein 5060335, Egypt
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11072020, Lebanon.,Faculty of Pharmacy and the Research and Innovation Hub, Alamein International University, Alamein 5060335, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
8
|
Wang X, Ye J, Gao M, Zhang D, Jiang H, Zhang H, Zhao S, Liu X. Nifuroxazide inhibits the growth of glioblastoma and promotes the infiltration of CD8 T cells to enhance antitumour immunity. Int Immunopharmacol 2023; 118:109987. [PMID: 36924564 DOI: 10.1016/j.intimp.2023.109987] [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: 11/04/2022] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Glioblastoma is a primary intracranial tumour with extremely high disability and fatality rates among adults. Existing diagnosis and treatment methods have not significantly improved the overall poor prognosis of patients. Nifuroxazide, an oral antibiotic, has been reported to act as a tumour suppressor in a variety of tumours and to participate in the process of antitumour immunity. However, whether it can inhibit the growth of glioma is still unclear. METHODS We explored the potential mechanism of nifuroxazide inhibiting the growth of glioblastoma cells through in vitro and in vivo experiments. RESULTS nifuroxazide can inhibit the proliferation of glioblastoma cells, promote G2 phase arrest, induce apoptosis, and inhibit epithelial-mesenchymal transition through the MAP3K1/JAK2/STAT3 pathway. Similarly, clinical sample analysis confirmed that MAP3K1 combined with STAT3 can affect the prognostic characteristics of patients with glioma. In addition, nifuroxazide can drive the M1 polarization of microglioma cells, inhibit the expression of CTLA4 and PD-L1 in tumour cells, and promote the infiltration of CD8 T cells to exert antitumour effects. Combination treatment with PD-L1 inhibitors can significantly prolong the survival time of mice. CONCLUSION we found that nifuroxazide can inhibit the growth of glioblastoma and enhance antitumour immunity. Thus, nifuroxazide is an effective drug for the treatment of glioblastoma and has great potential for clinical application.
Collapse
Affiliation(s)
- Xinzhuang Wang
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junyi Ye
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ming Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Dongzhi Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Haiping Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hong Zhang
- Department of Hematology, Liaocheng People's Hospital, Liaocheng, China
| | - Shiguang Zhao
- Shenzhen University General Hospital, Xueyuan AVE 1098, Nanshan District, 11, Shenzhen, Guangdong, P. R. China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Xianzhi Liu
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
9
|
Mossine VV, Kelley SP, Waters JK, Mawhinney TP. Screening a small hydrazide-hydrazone combinatorial library for targeting the STAT3 in monocyte-macrophages with insulated reporter transposons. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
AbstractThe Signal Transducer and Activator of Transcription 3 (STAT3) pharmacological targeting is regarded as a prospective approach to treat cancer, autoimmune disorders, or inflammatory diseases. We have developed a series of reporters of the STAT3, NF-κB, Nrf2, metal-responsive transcription factor-1 (MTF-1), and hypoxia-inducible factor 1α (HIF-1α) transcriptional activation in human monocyte-macrophage line THP-1. The reporter lines were employed to test a set of hydrazide-hydrazones as potential STAT3 inhibitors. A hydrazide-hydrazone library composed of 70 binary combinations of 7 carbonyl and 10 hydrazide components, including a STAT3 inhibitor clinical drug nifuroxazide, has been assembled and screened by the reporters. For the library as a whole, significant correlations between responses of the STAT3 and NF-κB or the STAT3 and HIF-1α reporters in THP-1 monocytes were found. For selected inhibitory combinations, respective hydrazide-hydrazones have been prepared and tested individually. The most potent 2-acetylpyridine 4-chlorobenzoylhydrazone exhibited the STAT3 inhibitory potential significantly exceeding that of nifuroxazide (ED50 2 vs 50 μM respectively) in THP-1 cells. We conclude that insulated reporter transposons could be a useful tool for drug discovery applications.
Graphical Abstract
Collapse
|
10
|
Saber S, El-Fattah EEA, Abdelhamid AM, Mourad AAE, Hamouda MAM, Elrabat A, Zakaria S, Haleem AA, Mohamed SZ, Elgharabawy RM, Morsy NE, El Adle Khalaf N, Mohammed OA, El-Bahouty WB, Mostafa SA, Abdelhady R, Galal O, ElSaid ZH, Yahya G, Shata A, Youssef ME. Innovative challenge for the inhibition of hepatocellular carcinoma progression by combined targeting of HSP90 and STAT3/HIF-1α signaling. Biomed Pharmacother 2023; 158:114196. [PMID: 36916405 DOI: 10.1016/j.biopha.2022.114196] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third foremost cause of cancer-related deaths. HCC has a very bad prognosis because it is asymptomatic in the early stages, resulting in a late diagnosis, and it is highly resistant to conventional chemotherapy. Such chemotherapies have been proven disappointing because they provide extremely low survival benefits. This study discloses that the STAT3/HIF-1α is an auspicious therapeutic attack site for conceivable repression of HCC development. A site that can be targeted by simultaneous administration of a STAT3 inhibitor in the context of HSP90 inhibition. 17-DMAG binds to HSP90 and constrains its function, resulting in the degradation of HSP90 client proteins HIF-1α and STAT3. Hypoxia recruits STAT3/HIF-1α complex within the VEGF promoter. Additionally, it was acknowledged that STAT3 is an essential mediator of VEGF transcription by direct binding to its promoter. Furthermore, it induces HIF-1α stability and enhances its transcriptional activity. Herein, we revealed that the combination therapy using 17-DMAG and nifuroxazide, a STAT3 inhibitor, repressed the diethylnitrosamine-induced alterations in the structure of the liver. This effect was mediated via decreasing the levels of the HSP90 client proteins HIF-1α and pSTAT3 resulting in the suppression of the STAT3/HIF-1α complex transcriptional activity. To conclude, 17-DMAG/NFXZD combination therapy-induced disruption in the STAT3/HIF-1α loop led to a potential antiangiogenic activity and showed apoptotic potential by inhibiting autophagy and inducing ROS/apoptosis signaling. Additionally, this combination therapy exhibited promising survival prolongation in mice with HCC. Consequently, the use of 17-DMAG/NFXZD renders an inspirational perspective in managing HCC. However, further investigations are compulsory.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Amir Mohamed Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Ahmed A E Mourad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Port-Said University, Port-Said 42511, Egypt.
| | | | - Amr Elrabat
- Gastroenterology and Hepatology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Sahar Zakaria
- Department of Tropical Medicine, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Amira A Haleem
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Sherin Z Mohamed
- Department of Internal Medicine, Faculty of Medicine, Horus University, New Damietta 34518, Egypt.
| | | | - Nesreen Elsayed Morsy
- Pulmonary Medicine Department, Mansoura University Sleep Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Noura El Adle Khalaf
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Osama A Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt.
| | | | - Sally Abdallah Mostafa
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt.
| | - Omneya Galal
- Department of Pharmacology, Faculty of Pharmacy, Ahram Canadian University, Giza 12451, Egypt.
| | - Zeinab H ElSaid
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharqia 44519, Egypt.
| | - Ahmed Shata
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| |
Collapse
|
11
|
Discovery and SAR Study of Quinoxaline–Arylfuran Derivatives as a New Class of Antitumor Agents. Pharmaceutics 2022; 14:pharmaceutics14112420. [DOI: 10.3390/pharmaceutics14112420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/11/2022] Open
Abstract
A novel class of quinoxaline–arylfuran derivatives were designed, synthesized, and preliminarily evaluated for their antiproliferative activities in vitro against several cancer cell lines and normal cells. The representative derivative QW12 exerts a potent antiproliferative effect against HeLa cells (IC50 value of 10.58 μM), through inducing apoptosis and triggering ROS generation and the accumulation of HeLa cells in vitro. Western blot analysis showed that QW12 inhibits STAT3 phosphorylation (Y705) in a dose-dependent manner. The BLI experiment directly demonstrated that QW12 binds to the STAT3 recombination protein with a KD value of 67.3 μM. Furthermore, molecular docking investigation showed that QW12 specifically occupies the pY+1 and pY-X subpocket of the SH2 domain, thus blocking the whole transmission signaling process. In general, these findings indicated that the study of new quinoxaline–aryfuran derivatives as inhibitors of STAT3 may lead to new therapeutic medical applications for cancer in the future.
Collapse
|
12
|
The Use of Novel, Rapid Analytical Tools in the Assessment of the Stability of Tablets—A Pilot Analysis of Expired and Unexpired Tablets Containing Nifuroxazide. Processes (Basel) 2022. [DOI: 10.3390/pr10101934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In the analysis of finished pharmaceutical products, numerous innovative analytical techniques are often used, i.e., Raman spectroscopy, scanning electron microscopy, computer microtomography, directional hemispherical reflectance, and hyperspectral analyses. These techniques allow for the identification of changes in solid phases. Many advantages over other techniques can be attributed to these techniques, e.g., they are rapid, non-destructive, and comprehensive. They allow for the identification of changes occurring in solid phases. However, the above-mentioned methods are still not standard procedures in pharmaceutical research. The present study aimed to assess the possible usefulness of total directional hemispherical reflectance (THR), hyperspectral imaging, and computer microtomography to evaluate the stability of tablets containing nifuroxazide during storage. In the study, expired and unexpired coating tablets containing nifuroxazide (n = 10 each) were analyzed. In addition, four unexpired tablets were stored at 40°C over 3 months (stressed tablets). Reflectance was determined with seven wavelength bands from 335 nm to 2500 nm using an SOC-410 Directional Hemispherical Reflectometer (Surface Optics Corporation, San Diego, CA, USA). A Specim IQ hyperspectral camera (Spectral Imaging Ltd., Oulu, Finland) was used with a wavelength range of 400–1030 nm. Tablets were also scanned using X-ray microtomography (Phoenix vǀtomeǀx, GE Sensing & Inspection Technologies GmbH, Wunstorf, Germany). The results indicated that total reflectance was lower in expired tablets than in unexpired tablets in all spectral bands, except for 700–1100 nm and 1700–2500 nm. In turn, the stressed tablets showed higher THR values than expired tablets in all spectral bands, except for 1000–1700 nm. In addition, hyperspectral analysis of the homogeneity of the tablets, as well as X-ray microtomographic analysis of tablet density and coating thickness, indicated that these parameters differed significantly between the analyzed tablets.
Collapse
|
13
|
van Geffen C, Heiss C, Deißler A, Kolahian S. Pharmacological modulation of myeloid-derived suppressor cells to dampen inflammation. Front Immunol 2022; 13:933847. [PMID: 36110844 PMCID: PMC9468781 DOI: 10.3389/fimmu.2022.933847] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population with potent suppressive and regulative properties. MDSCs’ strong immunosuppressive potential creates new possibilities to treat chronic inflammation and autoimmune diseases or induce tolerance towards transplantation. Here, we summarize and critically discuss different pharmacological approaches which modulate the generation, activation, and recruitment of MDSCs in vitro and in vivo, and their potential role in future immunosuppressive therapy.
Collapse
|
14
|
Nifuroxazide in combination with CpG ODN exerts greater efficacy against hepatocellular carcinoma. Int Immunopharmacol 2022; 108:108911. [PMID: 35729838 DOI: 10.1016/j.intimp.2022.108911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
|
15
|
Saber S, Nasr M, Kaddah MMY, Mostafa-Hedeab G, Cavalu S, Mourad AAE, Gaafar AGA, Zaghlool SS, Saleh S, Hafez MM, Girgis S, Elgharabawy RM, Nader K, Alsharidah M, Batiha GES, El-Ahwany E, Amin NA, Elagamy HI, Shata A, Nader R, Khodir AE. Nifuroxazide-loaded cubosomes exhibit an advancement in pulmonary delivery and attenuate bleomycin-induced lung fibrosis by regulating the STAT3 and NF-κB signaling: A new challenge for unmet therapeutic needs. Pharmacotherapy 2022; 148:112731. [PMID: 35220029 DOI: 10.1016/j.biopha.2022.112731] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 02/08/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic progressive disease that portends a very poor prognosis. It has been suggested that STAT3 is a potential target in PF. This study highlights the importance of cubosomes as a drug delivery system in enhancing the bioavailability of nifuroxazide (NXZD), a poorly soluble STAT3 inhibitor. NXZD-loaded cubosomes (NXZD-LC) were in vitro and in vivo evaluated. In vitro, cubosomes presented a poly-angular nanosized particles with a mean size and zeta potential of 223.73 ± 4.73 nm and - 20.93 ± 2.38 mV, respectively. The entrapment efficiency of nifuroxazide was 90.56 ± 4.25%. The in vivo pharmacokinetic study and the lung tissue accumulation of NXZD were performed by liquid chromatography-tandem mass spectrometry after oral administration to rats. The nanoparticles exhibited a two-fold increase and 1.33 times of bioavailability and lung tissue concentration of NXZD compared to NXZD dispersion, respectively. In view of this, NXZD-LC effectively attenuated PF by targeting STAT3 and NF-κB signals. As a result, NXZD-LC showed a potential anti-inflammatory effect as revealed by the significant decrease in MCP-1, ICAM-1, IL-6, and TNF-α and suppressed fibrogenic mediators as indicated by the significant reduction in TGF-β, TIMP-1, and PDGF-BB in lung tissues. Besides, NXZD-LC improved antioxidant defense mechanisms and decreased LDH and BALF total protein. These effects contributed to decreased collagen deposition. To conclude, cubosomes represent an advantageous pharmaceutical delivery system for enhancing pulmonary delivery of poorly soluble drugs. Additionally, repurposing NXZD as an antifibrotic agent is a promising challenge and new therapeutic approach for unmet therapeutic needs.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Mohamed Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Helwan University, Cairo 11790, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Mohamed M Y Kaddah
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab 21934, Alexandria, Egypt.
| | - Gomaa Mostafa-Hedeab
- Pharmacology Department & Health Research Unit, Medical College, Jouf University, Saudi Arabia; Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni Suef, Egypt.
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania.
| | - Ahmed A E Mourad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt.
| | - Ahmed Gaafar Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt.
| | - Sameh S Zaghlool
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Mokattam, Cairo 11571, Egypt.
| | - Safaa Saleh
- Department of Clinical Physiology, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| | - Mohamed M Hafez
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt.
| | - Samuel Girgis
- Department of Pharmaceutics, Faculty of Pharmacy, Alsalam University, Egypt.
| | | | - Karim Nader
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Mansour Alsharidah
- Department of Physiology, College of Medicine, Qassim University, Qassim 51452, Kingdom of Saudi Arabia.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt.
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Noha A Amin
- Department of Haematology, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Heba I Elagamy
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Ahmed Shata
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Reem Nader
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Ahmed E Khodir
- Department of Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt.
| |
Collapse
|
16
|
Zanoni M, Bravaccini S, Fabbri F, Arienti C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front Med (Lausanne) 2022; 9:795762. [PMID: 35299840 PMCID: PMC8920988 DOI: 10.3389/fmed.2022.795762] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/10/2022] [Indexed: 12/19/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes often upregulated in cancer cells and associated with therapeutic resistance. In humans, the ALDH family comprises 19 isoenzymes active in the majority of mammalian tissues. Each ALDH isoform has a specific differential expression pattern and most of them have individual functional roles in cancer. ALDHs are overexpressed in subpopulations of cancer cells with stem-like features, where they are involved in several processes including cellular proliferation, differentiation, detoxification and survival, participating in lipids and amino acid metabolism and retinoic acid synthesis. In particular, ALDH enzymes protect cancer cells by metabolizing toxic aldehydes in less reactive and more soluble carboxylic acids. High metabolic activity as well as conventional anticancer therapies contribute to aldehyde accumulation, leading to DNA double strand breaks (DSB) through the generation of reactive oxygen species (ROS) and lipid peroxidation. ALDH overexpression is crucial not only for the survival of cancer stem cells but can also affect immune cells of the tumour microenvironment (TME). The reduction of ROS amount and the increase in retinoic acid signaling impairs immunogenic cell death (ICD) inducing the activation and stability of immunosuppressive regulatory T cells (Tregs). Dissecting the role of ALDH specific isoforms in the TME can open new scenarios in the cancer treatment. In this review, we summarize the current knowledge about the role of ALDH isoforms in solid tumors, in particular in association with therapy-resistance.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | | | | | - Chiara Arienti
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
17
|
Nifuroxazide Mitigates Angiogenesis in Ehlrich's Solid Carcinoma: Molecular Docking, Bioinformatic and Experimental Studies on Inhibition of Il-6/Jak2/Stat3 Signaling. Molecules 2021; 26:molecules26226858. [PMID: 34833950 PMCID: PMC8621155 DOI: 10.3390/molecules26226858] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Nifuroxazide is an antidiarrheal medication that has promising anticancer activity against diverse types of tumors. The present study tested the anticancer activity of nifuroxazide against Ehrlich’s mammary carcinoma grown in vivo. Furthermore, we investigated the effect of nifuroxazide on IL-6/jak2/STAT3 signaling and the possible impact on tumor angiogenesis. The biological study was supported by molecular docking and bioinformatic predictions for the possible effect of nifuroxazide on this signaling pathway. Female albino mice were injected with Ehrlich carcinoma cells to produce Ehrlich’s solid tumors (ESTs). The experimental groups were as follows: EST control, EST + nifuroxazide (5 mg/kg), and EST + nifuroxazide (10 mg/kg). Nifuroxazide was found to reduce tumor masses (730.83 ± 73.19 and 381.42 ± 109.69 mg vs. 1099.5 ± 310.83) and lessen tumor pathologies. Furthermore, nifuroxazide downregulated IL-6, TNF-α, NFk-β, angiostatin, and Jak2 proteins, and it also reduced tumoral VEGF, as indicated by ELISA and immunohistochemical analysis. Furthermore, nifuroxazide dose-dependently downregulated STAT3 phosphorylation (60% and 30% reductions, respectively). Collectively, the current experiment shed light on the antitumor activity of nifuroxazide against mammary solid carcinoma grown in vivo. The antitumor activity was at least partly mediated by inhibition of IL-6/Jak2/STAT3 signaling that affected angiogenesis (low VEGF and high angiostatin) in the EST. Therefore, nifuroxazide might be a promising antitumor medication if appropriate human studies will be conducted.
Collapse
|
18
|
Gamov G, Kiselev A, Murekhina A, Zavalishin M, Aleksandriiskii V, Kosterin D. Synthesis, protolytic equilibria, and antimicrobial action of nifuroxazide analogs. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
19
|
Design, Synthesis, and Anticancer Screening for Repurposed Pyrazolo[3,4-d]pyrimidine Derivatives on Four Mammalian Cancer Cell Lines. Molecules 2021; 26:molecules26102961. [PMID: 34065773 PMCID: PMC8156061 DOI: 10.3390/molecules26102961] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
The present study reports the synthesis of new purine bioisosteres comprising a pyrazolo[3,4-d]pyrimidine scaffold linked to mono-, di-, and trimethoxy benzylidene moieties through hydrazine linkages. First, in silico docking experiments of the synthesized compounds against Bax, Bcl-2, Caspase-3, Ki67, p21, and p53 were performed in a trial to rationalize the observed cytotoxic activity for the tested compounds. The anticancer activity of these compounds was evaluated in vitro against Caco-2, A549, HT1080, and Hela cell lines. Results revealed that two (5 and 7) of the three synthesized compounds (5, 6, and 7) showed high cytotoxic activity against all tested cell lines with IC50 values in the micro molar concentration. Our in vitro results show that there is no significant apoptotic effect for the treatment with the experimental compounds on the viability of cells against A549 cells. Ki67 expression was found to decrease significantly following the treatment of cells with the most promising candidate: drug 7. The overall results indicate that these pyrazolopyrimidine derivatives possess anticancer activity at varying doses. The suggested mechanism of action involves the inhibition of the proliferation of cancer cells.
Collapse
|
20
|
El-Wakil MH, Meheissen MA, Abu-Serie MM. Nitrofurazone repurposing towards design and synthesis of novel apoptotic-dependent anticancer and antimicrobial agents: Biological evaluation, kinetic studies and molecular modeling. Bioorg Chem 2021; 113:104971. [PMID: 34051413 DOI: 10.1016/j.bioorg.2021.104971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
Drug repurposing has gained much attention as a cost-effective strategy that plays an exquisite role in identifying undescribed biological activities in clinical drugs. In the present work, we report the repurposing of the antibacterial drug nitrofurazone (NFZ) as a potential anticancer agent against CaCo-2, MDA-MB 231 and HepG-2 cancer cell lines. Novel series of nitrofurazone analogs were then designed considering the important pharmacologic features present in NFZ. Synthesis and biological evaluation of the target compounds revealed their promising anticancer activities endowed with antimicrobial potential and possessing better lipophilicity than NFZ. Compound 7, exclusively, inhibited the growth of all tested cancer cells more potently than NFZ with the least cytotoxicity against normal cells, displaying anti Gram-positive bacterial activities and antifungal potential. Analysis of the stereo-electronic properties of compound 7 via investigating the energies of HOMO, LUMO, HOMO-LUMO energy gap and MEP maps demonstrated its high reactivity and the expected molecular mechanism of action through reduction of the 5-nitrofuryl moiety. Data of the bioactivity studies indicated that the potent anticancer activity of 7 is mainly through increasing intracellular ROS levels and induction of apoptosis via significantly down-regulating the expression of Bcl-2 while up-regulating BAX, p53 and caspase 3 expression levels. Compound 7 potently inhibited the cellular expression levels of antioxidant enzymes GPx1 and GR compared to NFZ. Antioxidant enzymes kinetic studies and blind molecular docking simulations disclosed the mechanistic and structural aspects of the interaction between 7 and both GR and GPx1. Thus, the successful discovery of 7 as a potential dual anticancer-antimicrobial nitrofurazone analog might validate the applicability of drug repurposing strategy in unravelling the unrecognized bioactivity of the present conventional drugs, besides furnishing the way towards more optimization and development studies.
Collapse
Affiliation(s)
- Marwa H El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Marwa Ahmed Meheissen
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria 21521, Egypt
| | - Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Egypt
| |
Collapse
|
21
|
da Costa MOL, Pavani TFA, Lima AN, Scott AL, Ramos DFV, Lazarini M, Rando DGG. Nifuroxazide as JAK2 inhibitor: A binding mode proposal and Hel cell proliferation assay. Eur J Pharm Sci 2021; 162:105822. [PMID: 33775828 DOI: 10.1016/j.ejps.2021.105822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/01/2021] [Accepted: 03/21/2021] [Indexed: 12/23/2022]
Abstract
Nifuroxazide has been employed as an anti-diarrheic agent since 1966, but in the last decade has brought to the research spotlight again due to its recently described antitumoral activity through the JAK2 inhibitory potential. Since 2008, more than 70 papers have been published about the issue and more are expected to the following years. Herein we discuss the findings of molecular modelling studies which were performed to elucidate the potential binding mode of this drug into the JAK2 ATP recognition site and also into the allosteric region near the catalytic site. Molecular modelling followed by dynamics simulations indicated the NFZ could bind at both sites, such as a Type II kinase inhibitor since residues from both ATP and modulatory site would exhibit contacts with the drug when in a stable complex. Synthesis of NFZ and its sulfur bioisosteric analogue GPQF-63 were performed and experimental assays against HEL cells indicate the potential of NFZ and, mainly of its analogue GPQF-63 in acting as inhibitors of cell growth. HEL-cells present the JAK2 V617F mutation which leads to an enhanced JAK/STAT pathway and they have never been tested by the NFZ activity before. A mechanistic approach was also performed and revealed that both compounds induce cell apoptosis.Taken together, both the theoretical and experimental approaches point out the N-acylhydrazones as good starting points in the search for JAK2 modulatory small molecules which could then, be studied as promising leads toward new alternatives to control the JAK-STAT pathway related pathologies. This is the first study, as far as we have known, to propose a potential binding mode for NFZ as well as reporting the activity of this drug against HEL cells, which are a usual cellular model to human erythroleukemia and other myeloproliferative diseases.
Collapse
Affiliation(s)
- Marcela Oliveira Legramanti da Costa
- Grupo de Pesquisas Químico-Farmacêuticas, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema
| | - Thais Fernanda Amorim Pavani
- Grupo de Pesquisas Químico-Farmacêuticas, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema
| | - Angélica Nakagawa Lima
- Laboratório de Biologia Computacional e Bioinformática, Universidade Federal do ABC; Centro de Engenharia, Modelagem e Ciências Sociais Aplicadas, Universidade Federal do ABC
| | - Ana Lígia Scott
- Laboratório de Biologia Computacional e Bioinformática, Universidade Federal do ABC
| | - Débora Felicia Vieira Ramos
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema
| | - Mariana Lazarini
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema
| | - Daniela Gonçales Galasse Rando
- Grupo de Pesquisas Químico-Farmacêuticas, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema.
| |
Collapse
|
22
|
Ali AAA, Lee YR, Wu AT, Yadav VK, Yu DS, Huang HS. Structure-based strategies for synthesis, lead optimization and biological evaluation of N-substituted anthra[1,2-c][1,2,5]thiadiazole-6,11-dione derivatives as potential multi-target anticancer agents. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.10.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
23
|
Canh NX, Giang NV, Nghia VX, Sopjani M, Ngan NTT, Hoang NH, Xuan NT. Regulation of cell activation by A20 through STAT signaling in acute lymphoblastic leukemia. J Recept Signal Transduct Res 2020; 41:331-338. [PMID: 32808859 DOI: 10.1080/10799893.2020.1808678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the hematologic malignancy characterized by the aberrant proliferation of immature lymphoid cells. A20 is a deubiquitinase gene that inhibits functional activation of immune cells mediated through NF-κB/STAT pathways and frequently found inactivated in lymphoma. IL-6 is a pro-inflammatory cytokine secreted by immune cells under the pathogenic conditions and regulated by STAT signaling. Little is known about the role of A20 in regulating the function of ALL blasts and underlying molecular mechanisms. The present study, therefore, explored whether A20 expression contributes to IL-6 induced cell migration and activation of myeloid cells in ALL. To this end, blood samples of thirty-five adult ALL patients were examined. Gene expression profile was determined by quantitative RT-PCR, immunophenotype by flow cytometry, secretion of inflammatory cytokines by ELISA, and cell migration by a transwell migration assay. As a result, the expression of A20 was inactivated in ALL. Immunophenotypic analysis indicated that percent of CD11b+CD40+ expressing cells present in ALL was significantly reduced when transfected with PEM-T easy A20. Importantly, IL6-induced CXCL12-mediated migration of ALL blasts was dependent on the presence of A20. The inhibitory effects of A20 on activated myeloid cells and migration of ALL blasts were mediated through the STAT pathway upon IL-6 challenge. In addition, the CA-125 level was much higher in elderly females than either young female or male ALL patients or healthy donors. In conclusion, the inhibitory effects of A20 on activation of ALL blasts are expected to affect the immune response to treatment for adult ALL patients.
Collapse
Affiliation(s)
- Nguyen Xuan Canh
- Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Nguyen Van Giang
- Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | | | - Mentor Sopjani
- Faculty of Medicine, University of Prishtina, Prishtinë, Kosova
| | - Nguyen Thi Thanh Ngan
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi, Vietnam
| | - Nguyen Huy Hoang
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi, Vietnam
| | - Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi, Vietnam
| |
Collapse
|
24
|
MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071760. [PMID: 32630675 PMCID: PMC7409324 DOI: 10.3390/cancers12071760] [Citation(s) in RCA: 274] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, major advances have been made in cancer immunotherapy. This has led to significant improvement in prognosis of cancer patients, especially in the hematological setting. Nonetheless, translation of these successes to solid tumors was found difficult. One major mechanism through which solid tumors can avoid anti-tumor immunity is the downregulation of major histocompatibility complex class I (MHC-I), which causes reduced recognition by- and cytotoxicity of CD8+ T-cells. Downregulation of MHC-I has been described in 40-90% of human tumors, often correlating with worse prognosis. Epigenetic and (post-)transcriptional dysregulations relevant in the stabilization of NFkB, IRFs, and NLRC5 are often responsible for MHC-I downregulation in cancer. The intrinsic reversible nature of these dysregulations provides an opportunity to restore MHC-I expression and facilitate adaptive anti-tumor immunity. In this review, we provide an overview of the mechanisms underlying reversible MHC-I downregulation and describe potential strategies to counteract this reduction in MHC-I antigen presentation in cancer.
Collapse
|
25
|
Li G, Henry SA, Liu H, Kang TS, Nao SC, Zhao Y, Wu C, Jin J, Zhang JT, Leung CH, Wai Hong Chan P, Ma DL. A robust photoluminescence screening assay identifies uracil-DNA glycosylase inhibitors against prostate cancer. Chem Sci 2020; 11:1750-1760. [PMID: 34123270 PMCID: PMC8148385 DOI: 10.1039/c9sc05623h] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many cancers have developed resistance to 5-FU, due to removal by the enzyme uracil-DNA glycosylase (UDG), a type of base excision repair enzyme (BER) that can excise uracil and 5-fluorouracil (5-FU) from DNA. However, the development of UDG inhibitor screening methods, especially for the rapid and efficient screening of natural product/natural product-like compounds, is still limited so far. We developed herein a robust time-resolved photoluminescence method for screening UDG inhibitors, which could significantly improve sensitivity over the screening method based on the conventional steady-state spectroscopy, reducing the substantial fluorescence background interference. As a proof-of-concept, two potential UDG inhibitors were identified from a database of natural products and approved drugs. Co-treatment of these two compounds with 5-FU showed synergistic cytotoxicity, providing the basis for treating drug-resistant cancers. Overall, this method provides an avenue for the rapid screening of small molecule regulators of other BER enzyme activities that can avoid false negatives arising from the background fluorescence. The discovery of UDG inhibitors against prostate cancer by using a robust photoluminescence screening assay that can avoid false negatives arising from the background fluorescence.![]()
Collapse
Affiliation(s)
- Guodong Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Macau
| | | | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University Kowloon Tong Hong Kong
| | - Tian-Shu Kang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Macau
| | - Sang-Cuo Nao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Macau
| | - Yichao Zhao
- School of Chemistry, Monash University Clayton Victoria 3800 Australia
| | - Chun Wu
- Department of Chemistry, Hong Kong Baptist University Kowloon Tong Hong Kong
| | - Jianwen Jin
- School of Chemistry, Monash University Clayton Victoria 3800 Australia
| | - Jia-Tong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Macau
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Macau
| | - Philip Wai Hong Chan
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK.,School of Chemistry, Monash University Clayton Victoria 3800 Australia
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University Kowloon Tong Hong Kong
| |
Collapse
|
26
|
Aldehyde dehydrogenase-positive melanoma stem cells in tumorigenesis, drug resistance and anti-neoplastic immunotherapy. Mol Biol Rep 2019; 47:1435-1443. [PMID: 31838656 DOI: 10.1007/s11033-019-05227-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs), a rare subset of cancer cells, are well known for their self-renewing capacity. CSCs play a critical role in therapeutic failure and are responsible for poor prognosis in leukemia and various solid tumors. However, it is still unclear how CSCs initiate carcinogenesis and evade the immune response. In humans, the melanoma initiating cells (MICs) are recognized as the CSCs in melanomas, and were verified to possess CSC potentials. The enzymatic system, aldehyde dehydrogenase (ALDH) is considered to be a specific marker for CSCs in several tumors. The expression of ALDH in MICs may be closely correlated with phenotypic heterogeneity, melanoma-genesis, metastasis, and drug resistance. The ALDH+ CSCs/MICs not only serve as an indicator for therapeutic efficacy, but have also become a target for the treat of melanoma. In this review, we initially introduce the multiple capacities of MICs in melanoma. Then, we summarize in vivo and in vitro studies that illustrate the relationship between ALDH and MICs. Furthermore, understanding of chemotherapy resistance in melanoma relies on ALDH+ MICs. Finally, we review studies that focus on melanoma immunotherapies, rendering ALDH a potential marker to evaluate the efficacy of anti-neoplastic therapies or an adjuvant anti-melanoma target.
Collapse
|
27
|
Luo L, Xu F, Peng H, Luo Y, Tian X, Battaglia G, Zhang H, Gong Q, Gu Z, Luo K. Stimuli-responsive polymeric prodrug-based nanomedicine delivering nifuroxazide and doxorubicin against primary breast cancer and pulmonary metastasis. J Control Release 2019; 318:124-135. [PMID: 31838206 DOI: 10.1016/j.jconrel.2019.12.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/08/2019] [Accepted: 12/11/2019] [Indexed: 02/05/2023]
Abstract
Functionalized drug delivery systems against malignant lung metastasis of breast cancer have been extensively studied, while metastasis remains a challenging issue. We propose a new strategy to achieve eradication of primary breast cancer cells and inhibition of pulmonary metastasis. A cathepsin B/pH dual-sensitive block copolymer with a molecular weight of 92 kDa was synthesized to conjugate with doxorubicin (DOX). The copolymer-DOX was further loaded with nifuroxazide (NFX) to self-assemble co-prodrug-loaded micelles (CLM). CLM displayed a drug release pattern in response to pH/enzyme dual stimuli and was enzymatically biodegradable. CLM was demonstrated to reduce viability and inhibit migration and invasion of 4T1 murine breast cancer cells in vitro. After i.v. injection of CLM, its nanoscale size and stimuli-responsiveness facilitated delivery of drugs to the tumor site in mice. Enhanced anti-tumor efficacy and great anti-metastatic effects were found in both orthotropic and lung metastasis 4T1 breast cancer mice models. Meanwhile, histological immunofluorescence and immunohistochemical analyses revealed a high level of apoptosis, suppressed expression of matrix metalloproteinases and reduction in MDSCs infiltration, and all these contributed to inhibit pulmonary metastasis. CLM may be explored as a potential nanomedicine against breast cancer metastasis.
Collapse
Affiliation(s)
- Lei Luo
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| | - Fanshu Xu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Huilan Peng
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Yonghuang Luo
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Xiaohe Tian
- Department of Chemistry, Key Laboratory of Functional Inorganic Material Chemistry of Anhui Province, Anhui University, Hefei 230039, PR China
| | - Giuseppe Battaglia
- Department of Chemistry, Department of Chemical Engineering, University College London, UK
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
28
|
Zuma NH, Aucamp J, N'Da DD. An update on derivatisation and repurposing of clinical nitrofuran drugs. Eur J Pharm Sci 2019; 140:105092. [DOI: 10.1016/j.ejps.2019.105092] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 10/25/2022]
|
29
|
Thilakasiri PS, Dmello RS, Nero TL, Parker MW, Ernst M, Chand AL. Repurposing of drugs as STAT3 inhibitors for cancer therapy. Semin Cancer Biol 2019; 68:31-46. [PMID: 31711994 DOI: 10.1016/j.semcancer.2019.09.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
Drug repurposing is a valuable approach in delivering new cancer therapeutics rapidly into the clinic. Existing safety and patient tolerability data for drugs already in clinical use represent an untapped resource in terms of identifying therapeutic agents for off-label protein targets. The multicellular effects of STAT3 mediated by a range of various upstream signaling pathways make it an attractive therapeutic target with utility in a range of diseases including cancer, and has led to the development of a variety of STAT3 inhibitors. Moreover, heightened STAT3 transcriptional activation in tumor cells and within the cells of the tumor microenvironment contribute to disease progression. Consequently, there are many STAT3 inhibitors in preclinical development or under evaluation in clinical trials for their therapeutic efficacy predominantly in inflammatory diseases and cancer. Despite these advances, many challenges remain in ultimately providing STAT3 inhibitors to patients as cancer treatments, highlighting the need not only for a better understanding of the mechanisms associated with STAT3 activation, but also how various pharmaceutical agents suppress STAT3 activity in various cancers. In this review we discuss the importance of STAT3-dependent functions in cancer, review the status of compounds designed as direct-acting STAT3 inhibitors, and describe some of the strategies for repurposing of drugs as STAT3 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Pathum S Thilakasiri
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Rhynelle S Dmello
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Tracy L Nero
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia; Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia; Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Matthias Ernst
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashwini L Chand
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.
| |
Collapse
|
30
|
Li Y, Gan C, Zhang Y, Yu Y, Fan C, Deng Y, Zhang Q, Yu X, Zhang Y, Wang L, He F, Xie Y, Ye T, Yin W. Inhibition of Stat3 Signaling Pathway by Natural Product Pectolinarigenin Attenuates Breast Cancer Metastasis. Front Pharmacol 2019; 10:1195. [PMID: 31649548 PMCID: PMC6796319 DOI: 10.3389/fphar.2019.01195] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/17/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Breast cancer is the most common female cancer with considerable metastatic potential, which urges the need for developing novel potential drug candidate to inhibit tumor metastasis. Signal transducer and activator of transcription 3 (Stat3) have critical roles in cancer growth and metastasis and have been confirmed as a promising anticancer target. Here, we report our finding with pectolinarigenin, a flavonoid compound isolated from the aerial parts of Cirsium chanroenicum. Methods: The role of Pec. in cell proliferation, cell apoptosis, and cell migration and invasion in three breast cancer cells (4T1, MDA-MB-231, MCF-7) was investigated. Cell proliferation was determined by MTT assay, cell apoptosis was determined by flow cytometry, and protein expression was detected by western blotting. Tumor xenograft mice model and breast tumor metastasis model in vivo were built to further assess the effects of Pec. on 4T1 cells. Results: Intraperitoneal administrations of pectolinarigenin significantly inhibited breast cancer metastasis to lungs without affecting the tumor growth of incubated 4T1 breast cancer cells. Pectolinarigenin could also recruit CD8+ T cells to mediate tumor immune response. Furthermore, pectolinarigenin markedly impaired cancer cell migration and invasion by down-regulating phosphorylated-Stat3, and expression of matrix metalloproteinase (MMP)-2, MMP-9, while up-regulating the expression of TIMP2. We also found that pectolinarigenin inhibited breast cancer cell proliferation and induced apoptosis via mitochondrial-related apoptosis pathway, reduced mitochondrial membrane potential and the expression of Bcl-2, increased expression of Bax, and cleaved caspase-3 as well as disturbed the ROS generation. Conclusions: Pectolinarigenin might potentially be a candidate for metastasis of breast cancer by mediating Stat3 pathway.
Collapse
Affiliation(s)
- Yali Li
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Cailing Gan
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yange Zhang
- Cosmetic Plastic and Burn Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yan Yu
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chen Fan
- School of Pharmacy, Southwest University for Nationalities, Chengdu, China
| | - Yuanle Deng
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Qianyu Zhang
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Xi Yu
- Carey Business School, Johns Hopkins University, Baltimore, MD, United States
| | - Yiwen Zhang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Liqun Wang
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Fang He
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Yongmei Xie
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tinghong Ye
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenya Yin
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Toward a repositioning of the antibacterial drug nifuroxazide for cancer treatment. Drug Discov Today 2019; 24:1930-1936. [DOI: 10.1016/j.drudis.2019.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/22/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
|
32
|
Zhu Y, Zuo W, Chen L, Bian S, Jing J, Gan C, Wu X, Liu H, Su X, Hu W, Guo Y, Wang Y, Ye T. Repurposing of the anti-helminthic drug niclosamide to treat melanoma and pulmonary metastasis via the STAT3 signaling pathway. Biochem Pharmacol 2019; 169:113610. [PMID: 31465777 DOI: 10.1016/j.bcp.2019.08.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/14/2019] [Indexed: 02/05/2023]
Abstract
The incidence of melanoma is increasing rapidly worldwide. Additionally, new and effective candidates for treating melanoma are needed because of the increase in drug resistance and the high metastatic potential of this cancer. The STAT3 signaling pathway plays a pivotal role in pathogenesis of melanoma, making STAT3 a promising anticancer target for melanoma therapy. Niclosamide, an FDA-approved anti-helminthic drug, has been identified as a potent STAT3 inhibitor that suppresses STAT3 phosphorylation at Tyr705 and its transcript activity. In this study, we evaluated the biological activities of niclosamide in melanoma in vitro and in vivo. Niclosamide potently inhibited the growth of four melanoma cell lines and induced the apoptosis of melanoma cells via the mitochondrial apoptotic pathway. Further, western blot analysis indicated that cell apoptosis was correlated with activation of Bax and cleaved caspase-3 and decreased expression of Bcl-2. Moreover, niclosamide markedly impaired melanoma cell migration and invasion, reduced phosphorylated STAT3Tyr705 levels, and inhibited matrix metalloproteinase-2 and -9 expression. Additionally, in a xenograft model of A375, intraperitoneal administration of niclosamide inhibited tumor growth and tumor weight in a dose-dependent manner without obvious side effects. Histological and immunohistochemical analyses revealed a decrease in Ki-67-positive cells and p-STAT3Try705-positive cells and increase in cleaved caspase-3-positive cells. Notably, niclosamide significantly inhibited pulmonary metastasis in a B16-F10 melanoma lung metastasis model, including the number of lung metastatic nodules and lung/body coefficient. Importantly, a marked reduction in myeloid-derived suppressor cells (Gr1+CD11b+) infiltration in the pulmonary metastasis tissue was observed. Taken together, these results demonstrate that niclosamide is a promising candidate for treating melanoma.
Collapse
Affiliation(s)
- Yongxia Zhu
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Weiqiong Zuo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lijuan Chen
- Department of Radiology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Shasha Bian
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Jiayu Jing
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Cailin Gan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xiuli Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Hongyao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xingping Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Wanglai Hu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Yuqi Guo
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Yue Wang
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China.
| | - Tinghong Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
33
|
Pijuan J, Barceló C, Moreno DF, Maiques O, Sisó P, Marti RM, Macià A, Panosa A. In vitro Cell Migration, Invasion, and Adhesion Assays: From Cell Imaging to Data Analysis. Front Cell Dev Biol 2019; 7:107. [PMID: 31259172 PMCID: PMC6587234 DOI: 10.3389/fcell.2019.00107] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023] Open
Abstract
Cell migration is a key procedure involved in many biological processes including embryological development, tissue formation, immune defense or inflammation, and cancer progression. How physical, chemical, and molecular aspects can affect cell motility is a challenge to understand migratory cells behavior. In vitro assays are excellent approaches to extrapolate to in vivo situations and study live cells behavior. Here we present four in vitro protocols that describe step-by-step cell migration, invasion and adhesion strategies and their corresponding image data quantification. These current protocols are based on two-dimensional wound healing assays (comparing traditional pipette tip-scratch assay vs. culture insert assay), 2D individual cell-tracking experiments by live cell imaging and three-dimensional spreading and transwell assays. All together, they cover different phenotypes and hallmarks of cell motility and adhesion, providing orthogonal information that can be used either individually or collectively in many different experimental setups. These optimized protocols will facilitate physiological and cellular characterization of these processes, which may be used for fast screening of specific therapeutic cancer drugs for migratory function, novel strategies in cancer diagnosis, and for assaying new molecules involved in adhesion and invasion metastatic properties of cancer cells.
Collapse
Affiliation(s)
- Jordi Pijuan
- Flow Cytometry and Confocal Microscopy Unit, IRBLleida, University of Lleida, Lleida, Spain
| | | | - David F Moreno
- Molecular Biology Institute of Barcelona, CSIC, Barcelona, Spain
| | | | - Pol Sisó
- IRBLleida, University of Lleida, Lleida, Spain
| | - Rosa M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, Lleida, Spain.,Center of Biomedical Research on Cancer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Macià
- IRBLleida, University of Lleida, Lleida, Spain
| | - Anaïs Panosa
- Flow Cytometry and Confocal Microscopy Unit, IRBLleida, University of Lleida, Lleida, Spain
| |
Collapse
|
34
|
Anticancer activity of grassy Hystrix brachyura bezoar and its mechanisms of action: An in vitro and in vivo based study. Biomed Pharmacother 2019; 114:108841. [PMID: 30981106 DOI: 10.1016/j.biopha.2019.108841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 01/17/2023] Open
Abstract
Porcupine bezoar (PB) is a calcified undigested material generally found in porcupine's (Hystrix brachyura) gastrointestinal tract. The bezoar is traditionally used in South East Asia and Europe for the treatment of cancer, poisoning, dengue, typhoid, etc. However, limited scientific studies have been performed to verify its anticancer potential to substantiate its traditional claims in the treatment of cancers. Hence, this study was aimed at investigating the in vitro and in vivo anticancer properties of two grassy PB aqueous extract (PB-A and PB-B) using A375 cancer cell line and zebrafish model, respectively. This paper presents the first report on in vitro A375 cell viability assay, apoptosis assay, cell cycle arrest assay, migration assay, invasion assay, qPCR experimental assay and in vivo anti-angiogenesis assay using the grassy PBs. Experimental findings revealed IC50 value are 26.59 ± 1.37 μg/mL and 30.12 ± 3.25 μg/mL for PB-A and PB-B respectively. PBs showed anti-proliferative activity with no significant cytotoxic effect on normal human dermal fibroblast (NHDF). PBs were also found to induce apoptosis via intrinsic pathway and arrest cell cycle at G2/M phase. Additionally, the findings indicated its ability to debilitate migration and invasion of A375 cells. Further evaluation using embryo zebrafish model revealed LC50 = 450.0 ± 2.50 μg/mL and 58.7 ± 5.0 μg/mL for PB-A and PB-B which also exerted anti-angiogenesis effect in zebrafish. Moreover, stearic acid, ursodeoxycholic acid and pregnenolone were identified as possible metabolites that might contribute to the anticancer effect of the both PBs. Overall, this study demonstrated that PB-A and PB-B possess potential in vitro and in vivo anticancer effects which are elicited through selective cytotoxic effect, induction of apoptosis, inhibition of migration and invasion and anti-angiogenesis. This study provides scientific evidence that the porcupine bezoar do possess anti-cancer efficacy and further justifies its traditional utility. However, more experiments with higher vertebrae models are still warranted to validate its traditional claims as an anticancer agent.
Collapse
|
35
|
Luo Y, Zeng A, Fang A, Song L, Fan C, Zeng C, Ye T, Chen H, Tu C, Xie Y. Nifuroxazide induces apoptosis, inhibits cell migration and invasion in osteosarcoma. Invest New Drugs 2019; 37:1006-1013. [PMID: 30680584 DOI: 10.1007/s10637-019-00724-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/03/2019] [Indexed: 02/05/2023]
Abstract
Osteosarcoma is the most common primary malignancy of bone and characterized by an appendicular primary tumor with a high rate of metastasis to the lungs. Unfortunately, there is no effective strategy to treat osteosarcoma in current clinical practice. In this study, the anticancer effects and potential mechanisms of nifuroxazide, an oral nitrofuran antibiotic, on two osteosarcoma cell lines were investigated. The results of the antiproliferative activity in vitro showed that nifuroxazide inhibited cell proliferation of UMR106 and MG63 cells in a dose- and time-dependent manner. Interestingly, nifuroxazide showed low toxicity to non-tumor cells (HEK 293 T). In addition, ROS-mitochondrial mediated apoptosis was observed after treatment of nifuroxazide. Moreover, nifuroxazide could significantly inhibit osteosarcoma cells migration and invasion via p-Stat3, MMP-2 and MMP-9 mediated signaling pathway. Taken together, our results suggested that nifuroxazide could be a promising agent for osteosarcoma treatment by inhibiting cell proliferation, inducing cell apoptosis and impairing cell migration and invasion.
Collapse
Affiliation(s)
- Yi Luo
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Anqi Zeng
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan Province, 610064, People's Republic of China
| | - Aiping Fang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Chen Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Chenjuan Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Tinghong Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Healthy Science Center, Memphis, TN, 38163, USA
| | - Chongqi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan Province, 610041, People's Republic of China.
| |
Collapse
|
36
|
Elloumi-Mseddi J, Msalbi D, Fakhfakh R, Aifa S. Anti-Diarrheal Drug Repositioning in Tumour Cell Cytotoxicity. Anticancer Agents Med Chem 2019; 19:1037-1047. [PMID: 30657046 DOI: 10.2174/1871520619666190118120030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/18/2018] [Accepted: 01/03/2019] [Indexed: 12/07/2022]
Abstract
BACKGROUND Drug repositioning is becoming an ideal strategy to select new anticancer drugs. In particular, drugs treating the side effects of chemotherapy are the best candidates. OBJECTIVE In this present work, we undertook the evaluation of anti-tumour activity of two anti-diarrheal drugs (nifuroxazide and rifaximin). METHODS Anti-proliferative effect against breast cancer cells (MDA-MB-231, MCF-7 and T47D) was assessed by MTT analysis, the Brdu incorporation, mitochondrial permeability and caspase-3 activity. RESULTS Both the drugs displayed cytotoxic effects on MCF-7, T47D and MDA-MB-231 cells. The lowest IC50 values were obtained on MCF-7 cells after 24, 48 and 72 hours of treatment while T47D and MDA-MB-231 were more resistant. The IC50 values on T47D and MDA-MB-231 cells became significantly low after 72 hours of treatment showing a late cytotoxicity effect especially of nifuroxazide but still less important than that of MCF-7 cells. According to the IC50 values, the non-tumour cell line HEK293 seems to be less sensitive to cytotoxicity especially against rifaximin. Both the drugs have shown an accumulation of rhodamine 123 as a function of the rise of their concentrations while the Brdu incorporation decreased. Despite the absence of a significant difference in the cell cycle between the treated and non-treated MCF-7 cells, the caspase-3 activity increased with the drug concentrations rise suggesting an apoptotic effect. CONCLUSION Nifuroxazide and rifaximin are used to overcome the diarrheal side effect of anticancer drugs. However, they have shown to be anti-tumour drugs which make them potential dual effective drugs against cancer and the side effects of chemotherapy.
Collapse
Affiliation(s)
- Jihene Elloumi-Mseddi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, Sidi Mansour Road Km 6, BP 1177, 3018 Sfax, Tunisia
| | - Dhouha Msalbi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, Sidi Mansour Road Km 6, BP 1177, 3018 Sfax, Tunisia
| | - Raouia Fakhfakh
- Immunology Department, Habib Bourguiba Hospital, 3029 Sfax, Tunisia
| | - Sami Aifa
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, Sidi Mansour Road Km 6, BP 1177, 3018 Sfax, Tunisia
| |
Collapse
|
37
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
38
|
ALDH1 Bio-activates Nifuroxazide to Eradicate ALDH High Melanoma-Initiating Cells. Cell Chem Biol 2018; 25:1456-1469.e6. [PMID: 30293938 PMCID: PMC6309505 DOI: 10.1016/j.chembiol.2018.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/25/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023]
Abstract
5-Nitrofurans are antibiotic pro-drugs that have potential as cancer therapeutics. Here, we show that 5-nitrofurans can be bio-activated by aldehyde dehydrogenase (ALDH) 1A1/1A3 enzymes that are highly expressed in a subpopulation of cancer-initiating (stem) cells. We discover that the 5-nitrofuran, nifuroxazide, is selective for bio-activation by ALDH1 isoforms over ALDH2, whereby it both oxidizes ALDH1 and is converted to cytotoxic metabolites in a two-hit pro-drug mechanism. We show that ALDH1High melanoma cells are sensitive to nifuroxazide, while ALDH1A3 loss-of-function mutations confer drug resistance. In tumors, nifuroxazide targets ALDH1High melanoma subpopulations with the subsequent loss of melanoma-initiating cell potential. BRAF and MEK inhibitor therapy increases ALDH1 expression in patient melanomas, and effectively combines with nifuroxazide in melanoma cell models. The selective eradication of ALDH1High cells by nifuroxazide-ALDH1 activation goes beyond current strategies based on inhibiting ALDH1 and provides a rational basis for the nifuroxazide mechanism of action in cancer. ALDH1 bio-activates nifuroxazide leading to ALDH1 inactivation and cytotoxicity Nifuroxazide selectively eradicates ALDH1High melanoma tumor-initiating cells Targeted therapy increases ALDH1 in some patient melanomas and cell line models Targeting ALDH1High cells with nifuroxazide is an orthogonal therapeutic strategy
Collapse
|
39
|
Elsherbiny NM, Zaitone SA, Mohammad HMF, El-Sherbiny M. Renoprotective effect of nifuroxazide in diabetes-induced nephropathy: impact on NFκB, oxidative stress, and apoptosis. Toxicol Mech Methods 2018; 28:467-473. [DOI: 10.1080/15376516.2018.1459995] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Nehal M. Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Hala M. F. Mohammad
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mohamed El-Sherbiny
- Anatomy Department, Almaarefa College of Medicine, Riyadh, Saudi Arabia
- Anatomy Department, Mansoura Faculty of Medicine, Mansoura, Egypt
| |
Collapse
|
40
|
Yang S, Zhang Y, Luo Y, Xu B, Yao Y, Deng Y, Yang F, Ye T, Wang G, Cheng Z, Zheng Y, Xie Y. Hinokiflavone induces apoptosis in melanoma cells through the ROS-mitochondrial apoptotic pathway and impairs cell migration and invasion. Biomed Pharmacother 2018; 103:101-110. [PMID: 29635122 DOI: 10.1016/j.biopha.2018.02.076] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 02/05/2023] Open
Abstract
Melanoma, the highest degree of malignancy, is one of the most common skin tumors. However, there is no effective strategy to treat melanoma in current clinical practice. Therefore, it is urgent to find an efficient drug to overcome melanoma. Here, the in vitro anticancer effects of a natural product named hinokiflavone on three melanoma carcinoma cell lines (human melanoma A375 and CHL-1 cells, murine melanoma B16-F10 cells) and mechanisms of action were explored. The results of MTT assay revealed that hinokiflavone inhibited cell proliferation of these cell lines in a dose- and time-dependent manner. Interestingly, hinokiflavone showed low toxicity to normal liver cells. Flow cytometry assay and EdU incorporation assay indicated that hinokiflavone affected A375 and B16 cells survival by inducing apoptosis and blocking cell cycle progression at S phase in a concentration-dependent manner. Moreover, hinokiflavone enhanced the reactive oxygen species (ROS) and decreased the mitochondrial membrane potential obviously. Furthermore, hinokiflavone effectively impaired A375 cells migration and invasion, and down-regulated the expression of matrix metalloproteinase (MMP) MMP2 and MMP9. The above-mentioned results demonstrated that hinokiflavone could be a novel chemotherapeutic agent in melanoma treatment by inhibiting cell proliferation, inducing apoptosis and cell cycle arresting and blocking cell migration and invasion.
Collapse
Affiliation(s)
- Shuping Yang
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Yange Zhang
- Cosmetic Plastic and Burn Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Yi Luo
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Bocheng Xu
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Yuqin Yao
- Research Center for Occupational Respiratory Diseases, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Yuanle Deng
- Research Center for Occupational Respiratory Diseases, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Fangfang Yang
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Tinghong Ye
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Gang Wang
- School of Pharmacy, Zunyi Medical College, Zunyi, Guizhou Province 563003, PR China
| | - Zhiqiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yu Zheng
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China.
| | - Yongmei Xie
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China.
| |
Collapse
|
41
|
Nam ST, Kim HW, Kim HS, Park YH, Lee D, Lee MB, Min KY, Kim YM, Choi WS. Furaltadone suppresses IgE-mediated allergic response through the inhibition of Lyn/Syk pathway in mast cells. Eur J Pharmacol 2018; 828:119-125. [PMID: 29588153 DOI: 10.1016/j.ejphar.2018.03.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 11/25/2022]
Abstract
Mast cells are critical cells that prompt various allergic response-inducing factors, contributing to allergic diseases. While used as an antibiotic for livestock, there is no study on the effect of furaltadone on allergic response. This study investigated the effect of furaltadone on mast cells and passive cutaneous anaphylaxis (PCA). Furaltadone inhibited the degranulation of mast cells stimulated by antigen (IC50, ~ 3.9 μM), and also suppressed the production of tumor necrosis factor (TNF)-α and interleukin (IL)-4 in a concentration dependent manner. In addition, furaltadone inhibited allergic responses in an acute allergy animal model, PCA. Further investigation on the mechanism for these inhibitory effects of furaltadone found that the activities of Lyn/Syk and Syk-dependent downstream proteins such as mitogen-activated protein (MAP) kinases were inhibited by furaltadone in mast cells. Taken together, this study demonstrates that furaltadone inhibits the activation of mast cells by antigen via the suppression of the Lyn/Syk pathway and ameliorates allergic responses in vivo.
Collapse
Affiliation(s)
- Seung Taek Nam
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyun Woo Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Bum Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea.
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea.
| |
Collapse
|
42
|
Nifuroxazide, a STAT3 inhibitor, mitigates inflammatory burden and protects against diabetes-induced nephropathy in rats. Chem Biol Interact 2018; 281:111-120. [DOI: 10.1016/j.cbi.2017.12.030] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/30/2017] [Accepted: 12/26/2017] [Indexed: 01/17/2023]
|
43
|
Hassanain WA, Hegazy MA, Abdel Fattah LE, El-Fatatry HM. Simple chromatographic detection modes for antitumor agent and its degradants. J LIQ CHROMATOGR R T 2017. [DOI: 10.1080/10826076.2017.1378674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Waleed A. Hassanain
- Analytical Chemistry Department, Faculty of Pharmacy, Misr University for Science and Technology, Al-Motamayez District, 6th of October City, Egypt
| | - Maha A. Hegazy
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila E. Abdel Fattah
- Analytical Chemistry Department, Faculty of Pharmacy, Misr University for Science and Technology, Al-Motamayez District, 6th of October City, Egypt
| | - Hamed M. El-Fatatry
- Analytical Chemistry Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
44
|
Jia H, Cui J, Jia X, Zhao J, Feng Y, Zhao P, Zang D, Yu J, Zhao T, Wang H, Xu K. Therapeutic effects of STAT3 inhibition by nifuroxazide on murine acute graft graft-vs.-host disease: Old drug, new use. Mol Med Rep 2017; 16:9480-9486. [PMID: 29152660 PMCID: PMC5780006 DOI: 10.3892/mmr.2017.7825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 05/26/2017] [Indexed: 12/30/2022] Open
Abstract
Graft-vs.-host disease (GvHD) is a major and lethal complication of allogeneic bone marrow transplantation (allo-BMT). Although great development has been made, the treatment progress of this disorder is slow. Research has illustrated that STAT3 was critical for T cell alloactivation in GvHD. In the present study, the authors hypothesized that nifuroxazide, as the STAT3 inhibitor, treatment may attenuate the development of acute GvHD (aGvHD). The results demonstrated that nifuroxazide suppressed the development of aGvHD and significantly delayed aGvHD-induced lethality. Mice receiving nifuroxazide had mostly normal-appearing skin with minimal focal ulceration, mild edema and congestion in the liver, and a less-pronounced villus injury and less inflammatory infiltrate in the small intestine. Treatment with nifuroxazide inhibited the activation of STAT3, resulting in the regulation of the CD4+ T cells and CD4+CD25+ T cells and reduction of interferon-γ and tumor necrosis factor-α levels. In conclusion, nifuroxazide may be efficacious for post-transplant of GvHD, providing a potent drug for use as a prophylactic or as a second-line therapy for aGvHD in clinical trials.
Collapse
Affiliation(s)
- Huijie Jia
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jing Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Xiaolong Jia
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jingjing Zhao
- Department of Immunology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Yuchen Feng
- Department of Immunology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Peijuan Zhao
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Dan Zang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Jian Yu
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Tiesuo Zhao
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Hui Wang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453000, P.R. China
| | - Kailin Xu
- Laboratory of Transplantation and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
45
|
Deoxyarbutin displays antitumour activity against melanoma in vitro and in vivo through a p38-mediated mitochondria associated apoptotic pathway. Sci Rep 2017; 7:7197. [PMID: 28775302 PMCID: PMC5543205 DOI: 10.1038/s41598-017-05416-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/30/2017] [Indexed: 02/05/2023] Open
Abstract
Deoxyarbutin (DeoxyArbutin, dA), a natural compound widely used in skin lighting, displayed selectively cytotoxicity in vitro. In the study, we found that dA significantly inhibited viability/proliferation of B16F10 melanoma cells, induced tumour cell arrest and apoptosis. Furthermore, dA triggered its pro-apoptosis through damaging the mitochondrial function (membrane potential loss, ATP depletion and ROS overload generation etc.) and activating caspase-9, PARP, caspase-3 and the phosphorylation of p38. Treatment with p38 agonist confirmed the involvement of p38 pathway triggered by dA in B16F10 cells. The in vivo finding also revealed that administration of dA significantly decreased the tumour volume and tumour metastasis in B16F10 xenograft model by inhibiting tumour proliferation and inducing tumour apoptosis. Importantly, the results indicated that dA was specific against tumour cell lines and had no observed systemic toxicity in vivo. Taken together, our study demonstrated that dA could combate tumour in vitro and in vivo by inhibiting the proliferation and metastasis of tumour via a p38-mediated mitochondria associated apoptotic pathway.
Collapse
|
46
|
Shi L, Zheng H, Hu W, Zhou B, Dai X, Zhang Y, Liu Z, Wu X, Zhao C, Liang G. Niclosamide inhibition of STAT3 synergizes with erlotinib in human colon cancer. Onco Targets Ther 2017; 10:1767-1776. [PMID: 28367059 PMCID: PMC5370071 DOI: 10.2147/ott.s129449] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Niclosamide, an anthelmintic drug approved by the US Food and Drug Administration against cestodes, is used to treat tapeworm infection. In this study, we show that niclosamide can potentially inhibit signal transducer and activator of transcription 3 (STAT3) in colon cancer cell lines. Combined inhibition of epidermal growth factor receptor and STAT3 by erlotinib and niclosamide synergistically induces apoptosis and antiproliferation in colon cancer cell lines. Our findings suggest that erlotinib and niclosamide combination provides an effective therapeutic approach to improving the prognosis of colon cancer.
Collapse
Affiliation(s)
- Lingyi Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Hailun Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Wanle Hu
- Department of Coloproctology, The Second Affiliated Hospital and Yuying Children's Hospital
| | - Bin Zhou
- Department of Coloproctology, The Second Affiliated Hospital and Yuying Children's Hospital
| | - Xuanxuan Dai
- Department of Oncological Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yi Zhang
- Department of Oncological Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Xiaoping Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences
| |
Collapse
|
47
|
Ye TH, Yang FF, Zhu YX, Li YL, Lei Q, Song XJ, Xia Y, Xiong Y, Zhang LD, Wang NY, Zhao LF, Gou HF, Xie YM, Yang SY, Yu LT, Yang L, Wei YQ. Inhibition of Stat3 signaling pathway by nifuroxazide improves antitumor immunity and impairs colorectal carcinoma metastasis. Cell Death Dis 2017; 8:e2534. [PMID: 28055016 PMCID: PMC5386364 DOI: 10.1038/cddis.2016.452] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 02/05/2023]
Abstract
Colorectal carcinoma (CRC) is the one of the most common cancers with considerable metastatic potential, explaining the need for new drug candidates that inhibit tumor metastasis. The signal transducers and activators of the transcription 3 (Stat3) signaling pathway has an important role in CRC and has been validated as a promising anticancer target for CRC therapy. In the present study, we report our findings on nifuroxazide, an antidiarrheal agent identified as an inhibitor of Stat3. Our studies showed that nifuroxazide decreased the viability of three CRC cell lines and induced apoptosis of cancer cells in a concentration-dependent manner. Moreover, western blot analysis demonstrated that the occurrence of its apoptosis was correlated with the activation of Bax and cleaved caspase-3, and decreased the expression of Bcl-2. In addition, nifuroxazide markedly impaired CRC cell migration and invasion by downregulating phosphorylated-Stat3Tyr705, and also impaired the expression of matrix metalloproteinases (MMP-2 and MMP-9). Furthermore, our studies showed that nifuroxazide also significantly inhibited the tumor metastasis in lung and abdomen metastasis models of colon cancer. Meanwhile, nifuroxazide functionally reduced the proliferation index, induced tumor apoptosis and impaired metastasis. Notably, nifuroxazide reduced the number of myeloid-derived suppressor cells in the blood, spleens and tumors, accompanied by the increased infiltration of CD8+ T cells in the tumors. Importantly, a marked decrease in the number of M2-type macrophages in tumor in the abdomen metastasis model was also observed. Taken together, our results indicated that nifuroxazide could effectively inhibit tumor metastasis by mediating Stat3 pathway and it might have a therapeutic potential for the treatment of CRC.
Collapse
Affiliation(s)
- Ting-Hong Ye
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Fang-Fang Yang
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yong-Xia Zhu
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ya-Li Li
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qian Lei
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xue-Jiao Song
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ying Xiong
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China.,Department of Pharmacy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Li-Dan Zhang
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ning-Yu Wang
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Li-Feng Zhao
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hong-Feng Gou
- Department of Abdominal Cancer, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yong-Mei Xie
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Sheng-Yong Yang
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Luo-Ting Yu
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Li Yang
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yu-Quan Wei
- Department of Liver Surgery and Division of Digestive Diseases, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Prospect of Human Pluripotent Stem Cell-Derived Neural Crest Stem Cells in Clinical Application. Stem Cells Int 2016; 2016:7695836. [PMID: 28090209 PMCID: PMC5206454 DOI: 10.1155/2016/7695836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/21/2016] [Accepted: 06/20/2016] [Indexed: 12/13/2022] Open
Abstract
Neural crest stem cells (NCSCs) represent a transient and multipotent cell population that contributes to numerous anatomical structures such as peripheral nervous system, teeth, and cornea. NCSC maldevelopment is related to various human diseases including pigmentation abnormalities, disorders affecting autonomic nervous system, and malformations of teeth, eyes, and hearts. As human pluripotent stem cells including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) can serve as an unlimited cell source to generate NCSCs, hESC/hiPSC-derived NCSCs can be a valuable tool to study the underlying mechanisms of NCSC-associated diseases, which paves the way for future therapies for these abnormalities. In addition, hESC/hiPSC-derived NCSCs with the capability of differentiating to various cell types are highly promising for clinical organ repair and regeneration. In this review, we first discuss NCSC generation methods from human pluripotent stem cells and differentiation mechanism of NCSCs. Then we focus on the clinical application potential of hESC/hiPSC-derived NCSCs on peripheral nerve injuries, corneal blindness, tooth regeneration, pathological melanogenesis, Hirschsprung disease, and cardiac repair and regeneration.
Collapse
|
49
|
Ding Y, Li X, Hong D, Jiang L, He Y, Fang H. Silence of MACC1 decreases cell migration and invasion in human malignant melanoma through inhibiting the EMT. Biosci Trends 2016; 10:258-64. [PMID: 27488539 DOI: 10.5582/bst.2016.01091] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Metastasis-associated colon cancer 1 (MACC1) has been demonstrated to promote metastasis of several cancers via regulating epithelial-mesenchymal transition (EMT). However, its biological behavior in human malignant melanoma remains unclear. In this study, MACC1 downregulation was established in two melanoma cell lines (A375 and G361 cells) using RNA interference, as confirmed by quantitative real time PCR (qRT-PCR) and Western blot analysis. Subsequently, we investigated the effects of MACC1 silencing on cell mobility, migration and invasion using scratch wound and Transwell assays. Our results indicated that knockdown of MACC1 significantly suppressed cell migration and invasion ability of both melanoma cell lines. Moreover, downregulation of MACC1 upregulated E-cadherin, N-cadherin and Vimentin, as confirmed by qRT-PCR, Western blot and immunofluorescent Staining analysis. These findings suggest MACC1 might serve as a new molecular target for the treatment of melanoma by a novel mechanism underlying the metastasis of melanoma cells.
Collapse
Affiliation(s)
- Yingguo Ding
- Department of Dermatology, The First Affiliated Hospital of Zhejiang University
| | | | | | | | | | | |
Collapse
|
50
|
Benzothiazinethione is a potent preclinical candidate for the treatment of drug-resistant tuberculosis. Sci Rep 2016; 6:29717. [PMID: 27405961 PMCID: PMC4942819 DOI: 10.1038/srep29717] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/22/2016] [Indexed: 02/05/2023] Open
Abstract
New chemotherapeutic compounds are needed to combat multidrug-resistant Mycobacterium tuberculosis (Mtb), which remains a serious public-health challenge. Decaprenylphosphoryl-β-D-ribose 2′-epimerase (DprE1 enzyme) has been characterized as an attractive therapeutic target to address this urgent demand. Herein, we have identified a new class of DprE1 inhibitors benzothiazinethiones as antitubercular agents. Benzothiazinethione analogue SKLB-TB1001 exhibited excellent activity against Mtb in the Microplate Alamar blue assay and intracellular model, meanwhile SKLB-TB1001 was also highly potent against multi-drug resistant extensively and drug resistant clinical isolates. Importantly, no antagonism interaction was found with any two-drug combinations tested in the present study and the combination of SKLB-TB1001 with rifampicin (RMP) was proved to be synergistic. Furthermore, benzothiazinethione showed superb in vivo antitubercular efficacy in an acute Mtb infection mouse model, significantly better than that of BTZ043. These data combined with the bioavailability and safety profiles of benzothiazinethione indicates SKLB-TB1001 is a promising preclinical candidate for the treatment of drug-resistant tuberculosis.
Collapse
|