1
|
Nauroze T, Ali S, Andleeb S, Ara C, Liaqat I, Mushtaq H, Mumtaz S, Kanwal L, Abbas AS, Mumtaz S, Farooq MA, Khan IH. Therapeutic Potential of Aloe vera and Aloe vera-Conjugated Silver Nanoparticles on Mice Exposed to Hexavalent Chromium. Biol Trace Elem Res 2024; 202:5580-5595. [PMID: 38478315 DOI: 10.1007/s12011-024-04105-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/12/2024] [Indexed: 10/25/2024]
Abstract
Hexavalent chromium (Cr (VI)) is a hazardous heavy metal that induces hepatotoxicity and nephrotoxicity. Thus, this study was planned to explore the ameliorating capacity of Aloe vera leaf gel extract (AV) and their conjugated silver nanoparticles (AVNP) against Cr (VI) induced hepatotoxicity and renal toxicity. The organ indices, level of alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, malondialdehyde, total protein, and creatinine in blood serum were measured. The histopathological and micrometric analysis of the hepatic and renal tissue sections were studied. The hepatosomatic index was raised significantly (0.098 ± 0.13 g) in Cr treated group. The blood serum level of AST (484 ± 10.7 U/L), ALT (163 ± 5.5 U/L), ALP (336.7 ± 9.5 U/L), MDA (642.3 ± 28.3 U/L), and creatinine (4.0 ± 0.1 mg/dL) were increased significantly, whereas total protein level was declined (2.8 ± 0.3 g/dL) significantly in Cr exposed group. In the histopathological study, necrosis, disturbed hepatic cords, impaired glomeruli, and Bowman's capsule were noted. Micrometric data from the liver and kidney revealed a significant surge in the size of hepatocytes and their nuclei (1188.2 ± 467.7 µ2 and 456.5 ± 205.6 µ2) and CSA of glomeruli and Bowman's capsule (9051.8 ± 249.8 µ2 and 11,835.5 ± 336.7 µ2) in Cr (VI) exposed group, whereas the brush border (10.2 ± 4.0 µ) size declined significantly. The administration of AV and AVNP reduced the oxidative stress induced by Cr (VI).
Collapse
Affiliation(s)
- Tooba Nauroze
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
- Department of Zoology, University of Education, Lahore, Pakistan
| | - Shaukat Ali
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan.
| | - Shagufta Andleeb
- Department of Zoology, University of Education, Lahore, Pakistan
| | - Chaman Ara
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Iqra Liaqat
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Hina Mushtaq
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Samaira Mumtaz
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Lubna Kanwal
- Department of Zoology, University of Okara, Lahore, Pakistan
| | | | - Shumaila Mumtaz
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
- Department of Zoology, University of Poonch, Rawlakot, AzadKashmir, Pakistan
| | - Muhammad Adeel Farooq
- Applied Entomology and Medical Toxicology Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | | |
Collapse
|
2
|
Abtahi MS, Fotouhi A, Rezaei N, Akalin H, Ozkul Y, Hossein-Khannazer N, Vosough M. Nano-based drug delivery systems in hepatocellular carcinoma. J Drug Target 2024; 32:977-995. [PMID: 38847573 DOI: 10.1080/1061186x.2024.2365937] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024]
Abstract
The high recurrence rate of hepatocellular carcinoma (HCC) and poor prognosis after medical treatment reflects the necessity to improve the current chemotherapy protocols, particularly drug delivery methods. Development of targeted and efficient drug delivery systems (DDSs), in all active, passive and stimuli-responsive forms for selective delivery of therapeutic drugs to the tumour site has been extended to improve efficacy and reduce the severe side effects. Recent advances in nanotechnology offer promising breakthroughs in the diagnosis, treatment and monitoring of cancer cells. In this review, the specific design of DDSs based on the different nano-particles and their surface engineering is discussed. In addition, the innovative clinical studies in which nano-based DDS was used in the treatment of HCC were highlighted.
Collapse
Affiliation(s)
- Maryam Sadat Abtahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Fotouhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hilal Akalin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nikoo Hossein-Khannazer
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
3
|
Santavanond JP, Chiu YH, Tixeira R, Liu Z, Yap JKY, Chen KW, Li CL, Lu YR, Roncero-Carol J, Hoijman E, Rutter SF, Shi B, Ryan GF, Hodge AL, Caruso S, Baxter AA, Ozkocak DC, Johnson C, Day ZI, Mayfosh AJ, Hulett MD, Phan TK, Atkin-Smith GK, Poon IKH. The small molecule raptinal can simultaneously induce apoptosis and inhibit PANX1 activity. Cell Death Dis 2024; 15:123. [PMID: 38336804 PMCID: PMC10858176 DOI: 10.1038/s41419-024-06513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Discovery of new small molecules that can activate distinct programmed cell death pathway is of significant interest as a research tool and for the development of novel therapeutics for pathological conditions such as cancer and infectious diseases. The small molecule raptinal was discovered as a pro-apoptotic compound that can rapidly trigger apoptosis by promoting the release of cytochrome c from the mitochondria and subsequently activating the intrinsic apoptotic pathway. As raptinal is very effective at inducing apoptosis in a variety of different cell types in vitro and in vivo, it has been used in many studies investigating cell death as well as the clearance of dying cells. While examining raptinal as an apoptosis inducer, we unexpectedly identified that in addition to its pro-apoptotic activities, raptinal can also inhibit the activity of caspase-activated Pannexin 1 (PANX1), a ubiquitously expressed transmembrane channel that regulates many cell death-associated processes. By implementing numerous biochemical, cell biological and electrophysiological approaches, we discovered that raptinal can simultaneously induce apoptosis and inhibit PANX1 activity. Surprisingly, raptinal was found to inhibit cleavage-activated PANX1 via a mechanism distinct to other well-described PANX1 inhibitors such as carbenoxolone and trovafloxacin. Furthermore, raptinal also interfered with PANX1-regulated apoptotic processes including the release of the 'find-me' signal ATP, the formation of apoptotic cell-derived extracellular vesicles, as well as NLRP3 inflammasome activation. Taken together, these data identify raptinal as the first compound that can simultaneously induce apoptosis and inhibit PANX1 channels. This has broad implications for the use of raptinal in cell death studies as well as in the development new PANX1 inhibitors.
Collapse
Affiliation(s)
- Jascinta P Santavanond
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Yu-Hsin Chiu
- Departments of Medical Science, Life Science, and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
| | - Rochelle Tixeira
- Unit for Cell Clearance in Health and Disease, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Zonghan Liu
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Jeremy K Y Yap
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Kaiwen W Chen
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Chen-Lu Li
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ru Lu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Joan Roncero-Carol
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Esteban Hoijman
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Stephanie F Rutter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Bo Shi
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Gemma F Ryan
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Amy L Hodge
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Sarah Caruso
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Amy A Baxter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Dilara C Ozkocak
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Chad Johnson
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Zoe I Day
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Alyce J Mayfosh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
| | - Thanh K Phan
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
- The Walter and Eliza Hall Institute of Medial Research, Parkville, Vic, Australia
| | - Georgia K Atkin-Smith
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia
- The Walter and Eliza Hall Institute of Medial Research, Parkville, Vic, Australia
- University of Melbourne, Melbourne, VIC, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
- Research Centre of Extracellular Vesicles, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Mohamed MR, Osman SA, Hassan AA, Raafat AI, Refaat MM, Fathy SA. Gemcitabine and synthesized silver nanoparticles impact on chemically induced hepatocellular carcinoma in male rats. Int J Immunopathol Pharmacol 2024; 38:3946320241263352. [PMID: 39046434 PMCID: PMC11271163 DOI: 10.1177/03946320241263352] [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: 08/30/2023] [Accepted: 06/02/2024] [Indexed: 07/25/2024] Open
Abstract
Objective: Gemcitabine (GEM) is a deoxycytidine analog chemotherapeutic drug widely used to treat many cancers. Silver nanoparticles (AgNPs) are important nanomaterials used to treat many diseases. Using gamma radiation in nanoparticle preparation is a new eco-friendly method. This study aims to evaluate the efficiency of co-treating gemcitabine and silver nanoparticles in treating hepatocellular carcinoma. Method: The AgNPs were characterized using UV-visible spectroscopy, XRD, TEM, and EDX. The MTT cytotoxicity in vitro assay of gemcitabine, doxorubicin, and cyclophosphamide was assessed against Wi38 normal fibroblast and HepG2 HCC cell lines. After HCC development, rats received (10 µg/g b.wt.) of AgNPs three times a week for 4 weeks and/or GEM (5 mg/kg b.wt.) twice weekly for 4 weeks. Liver function enzymes were investigated. Cytochrome P450 and miR-21 genes were studied. Apoptosis was determined by using flow cytometry, and apoptotic modifications in signaling pathways were evaluated via Bcl-2, Bax, Caspase-9, and SMAD-4. Results: The co-treatment of GEM and AgNPs increased apoptosis by upregulating Bax and caspase 9 while diminishing Bcl2 and SMAD4. It also improved cytochrome P450 m-RNA relative expression. The results also proved the cooperation between GEM and AgNPs in deactivating miR21. The impact of AgNPs as an adjuvant treatment with GEM was recognized. Conclusions: The study showed that co-treating AgNPs and GEM can improve the efficiency of GEM alone in treating HCC. This is achieved by enhancing intrinsic and extrinsic apoptotic pathways while diminishing some drawbacks of using GEM alone.
Collapse
Affiliation(s)
- Mohamed R Mohamed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Soheir A Osman
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Asmaa A Hassan
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Amany I Raafat
- Polymer Chemistry Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mahmoud M Refaat
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Shadia A Fathy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
5
|
Roshani M, Rezaian-Isfahni A, Lotfalizadeh MH, Khassafi N, Abadi MHJN, Nejati M. Metal nanoparticles as a potential technique for the diagnosis and treatment of gastrointestinal cancer: a comprehensive review. Cancer Cell Int 2023; 23:280. [PMID: 37981671 PMCID: PMC10657605 DOI: 10.1186/s12935-023-03115-1] [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: 04/11/2023] [Accepted: 10/27/2023] [Indexed: 11/21/2023] Open
Abstract
Gastrointestinal (GI) cancer is a major health problem worldwide, and current diagnostic and therapeutic approaches are often inadequate. Various metallic nanoparticles (MNPs) have been widely studied for several biomedical applications, including cancer. They may potentially overcome the challenges associated with conventional chemotherapy and significantly impact the overall survival of GI cancer patients. Functionalized MNPs with targeted ligands provide more efficient localization of tumor energy deposition, better solubility and stability, and specific targeting properties. In addition to enhanced therapeutic efficacy, MNPs are also a diagnostic tool for molecular imaging of malignant lesions, enabling non-invasive imaging or detection of tumor-specific or tumor-associated antigens. MNP-based therapeutic systems enable simultaneous stability and solubility of encapsulated drugs and regulate the delivery of therapeutic agents directly to tumor cells, which improves therapeutic efficacy and minimizes drug toxicity and leakage into normal cells. However, metal nanoparticles have been shown to have a cytotoxic effect on cells in vitro. This can be a concern when using metal nanoparticles for cancer treatment, as they may also kill healthy cells in addition to cancer cells. In this review, we provide an overview of the current state of the field, including preparation methods of MNPs, clinical applications, and advances in their use in targeted GI cancer therapy, as well as the advantages and limitations of using metal nanoparticles for the diagnosis and treatment of gastrointestinal cancer such as potential toxicity. We also discuss potential future directions and areas for further research, including the development of novel MNP-based approaches and the optimization of existing approaches.
Collapse
Affiliation(s)
- Mohammad Roshani
- Internal Medicine and Gastroenterology, Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arya Rezaian-Isfahni
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Negar Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hassan Jafari Najaf Abadi
- Research Center for Health Technology Assessment and Medical Informatics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Li Y, Zou H, Zheng Z, Liu Z, Hu H, Wu W, Wang T. Advances in the Study of Bioactive Nanoparticles for the Treatment of HCC and Its Postoperative Residual Cancer. Int J Nanomedicine 2023; 18:2721-2735. [PMID: 37250475 PMCID: PMC10216871 DOI: 10.2147/ijn.s399146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Primary hepatocellular carcinoma (HCC, hepatocellular carcinoma) is the third leading cause of tumor death in the world and the second leading cause in China. The high recurrence rate at 5 years after surgery also seriously affects the long-term survival of HCC patients. For reasons such as poor liver function, large tumors, or vascular invasion, only relatively limited palliative treatment is available. Therefore, effective diagnostic and therapeutic strategies are needed to improve the complex microenvironment and block the mechanism of tumor development in order to treat the tumor and prevent recurrence. A variety of bioactive nanoparticles have been shown to have therapeutic effects on hepatocellular carcinoma and have the advantages of improving drug solubility, reducing drug side effects, preventing degradation in the blood, increasing drug exposure time, and reducing drug resistance. The development of bioactive nanoparticles is expected to complete the current clinical therapeutic approach. In this review, we discuss the therapeutic advances of different nanoparticles for hepatocellular carcinoma and discuss their potential for postoperative applications with respect to possible mechanisms of hepatocellular carcinoma recurrence. We further discuss the limitations regarding the application of NPs and the safety of NPs.
Collapse
Affiliation(s)
- Yanxu Li
- Medical College of Yangzhou University, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Hao Zou
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Zekun Zheng
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Zhuoheng Liu
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Huiyuan Hu
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Wei Wu
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| |
Collapse
|
7
|
Owis AI, Sherif NH, Hassan AA, El-Naggar EMB, El-Khashab IH, El-Ghaly ES. Tropaeolum majus L. and low dose gamma radiation suppress liver carcinoma development via EGFR-HER2 signaling pathway. Nat Prod Res 2023; 37:1030-1035. [PMID: 35834717 DOI: 10.1080/14786419.2022.2098958] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most fatal cancers around the world and remain asymptomatic in early stage. An alcoholic extract prepared from leaves of Tropaeolum majus L. (Tropaeolaceae) was assessed for its potential activity against diethylnitrosamine-induced liver carcinoma in vivo. Oral administration of the extract significantly decreased the inflammatory marker translation NF-kB and supressed HCC progression in combination with 0.5 Gy gamma radiation via EGF-HER-2 pathway. Histopathological and immunohistopathological features also showed the recovery of a hepatic architecture. Immunohistochemical study showed the T. majus and LDR enhancement effect on proapoptotic markers (caspase-3 and Bax) and inhibition of anti-apoptotic factor (BCl2). HPLC-DAD-MSn analysis of the extract revealed the annotation of twelve compounds. T. majus could mediate a defensive influence against diethylnitrosamine-induced hepatocarcinogenesis and serve as a respectable option in amelioration of the hepatocellular carcinoma development in combination with low dose of gamma radiation.
Collapse
Affiliation(s)
- Asmaa I Owis
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.,Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Noheir H Sherif
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.,Drug Radiation Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Asmaa A Hassan
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | - Iman H El-Khashab
- Department of Zoology, Faculty of Girls, Ain Shams University, Cairo, Egypt
| | - El-Sayed El-Ghaly
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
8
|
Kabir MA, Kharel A, Malla S, Kreis ZJ, Nath P, Wolfe JN, Hassan M, Kaur D, Sari-Sarraf H, Tiwari AK, Ray A. Automated detection of apoptotic versus nonapoptotic cell death using label-free computational microscopy. JOURNAL OF BIOPHOTONICS 2022; 15:e202100310. [PMID: 34936215 DOI: 10.1002/jbio.202100310] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Identification of cell death mechanisms, particularly distinguishing between apoptotic versus nonapoptotic pathways, is of paramount importance for a wide range of applications related to cell signaling, interaction with pathogens, therapeutic processes, drug discovery, drug resistance, and even pathogenesis of diseases like cancers and neurogenerative disease among others. Here, we present a novel high-throughput method of identifying apoptotic versus necrotic versus other nonapoptotic cell death processes, based on lensless digital holography. This method relies on identification of the temporal changes in the morphological features of mammalian cells, which are unique to each cell death processes. Different cell death processes were induced by known cytotoxic agents. A deep learning-based approach was used to automatically classify the cell death mechanism (apoptotic vs necrotic vs nonapoptotic) with more than 93% accuracy. This label free approach can provide a low cost (<$250) alternative to some of the currently available high content imaging-based screening tools.
Collapse
Affiliation(s)
- Md Alamgir Kabir
- Department of Physics and Astronomy, University of Toledo, Toledo, OH, USA
| | - Ashish Kharel
- Department of Electrical and Computer Science, University of Toledo, Toledo, OH, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | | | - Peuli Nath
- Department of Physics and Astronomy, University of Toledo, Toledo, OH, USA
| | - Jared Neil Wolfe
- Department of Mechanical Engineering, University of Toledo, Toledo, OH, USA
| | - Marwa Hassan
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Devinder Kaur
- Department of Electrical and Computer Science, University of Toledo, Toledo, OH, USA
| | - Hamed Sari-Sarraf
- Department of Electrical & Computer Engineering, Texas Tech University, Lubbock, TX, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Aniruddha Ray
- Department of Physics and Astronomy, University of Toledo, Toledo, OH, USA
| |
Collapse
|
9
|
Uehara T, Pogribny IP, Rusyn I. The DEN and CCl 4 -Induced Mouse Model of Fibrosis and Inflammation-Associated Hepatocellular Carcinoma. Curr Protoc 2021; 1:e211. [PMID: 34370903 PMCID: PMC8744072 DOI: 10.1002/cpz1.211] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human hepatocellular carcinoma (HCC) develops most often as a complication of fibrosis or cirrhosis. Although most human studies of HCC provide crucial insights into the molecular signatures of HCC, they seldom address its etiology. Mouse models provide essential tools for investigating the pathogenesis of HCC, but the majority of rodent cancer models do not feature liver fibrosis. Detailed here is a protocol for an experimental mouse model of HCC that arises in association with advanced liver fibrosis. The disease model is induced by a single injection of N-nitrosodiethylamine (DEN) at 2 weeks of age followed by repeated administration of carbon tetrachloride (CCl4 ) from 8 weeks of age for up to 14 consecutive weeks. A dramatic potentiation of liver tumor incidence is observed following administration of DEN and CCl4 , with 100% of mice developing liver tumors at 5 months of age. This model has been employed for studying the molecular mechanisms of fibrogenesis and HCC development, as well as for cancer hazard/chemotherapy testing of drug candidates. © 2021 Wiley Periodicals LLC. Basic Protocol: The DEN and CCl4 -induced mouse model of fibrosis and inflammation-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Takeki Uehara
- Strategic Development Department, Shionogi & Co., Osaka, Japan
| | - Igor P. Pogribny
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
10
|
Molecular and hormonal changes caused by long-term use of high dose pregabalin on testicular tissue: the role of p38 MAPK, oxidative stress and apoptosis. Mol Biol Rep 2020; 47:8523-8533. [PMID: 33051752 DOI: 10.1007/s11033-020-05894-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/03/2020] [Indexed: 01/20/2023]
Abstract
In 1990, pregabalin was introduced as a novel antiepileptic drug that acts by binding selectively to the alpha-2-delta subunits of voltage-gated calcium channels resulting in increasing neuronal GABA levels and inhibiting the release of exciting neurotransmitters. The aim of our study is to assess the hazardous effects of prolonged high-dose pregabalin (like that abused by addicts) on testes and to clarify the potential causative mechanisms. The current study was conducted on 70 adult male Wistar albino rats which were divided into 7 groups. In our study we evaluated the effect of pregabalin, at concentrations 150 and 300 mg/kg/day for 90 days, on hormones; FSH, LH, testosterone and prolactin secretion. Our study also evaluated the expression of apoptosis-related genes BAX and BCL2 in testicular tissue in addition to the western blotted analysis of p38 Mitogen activated protein kinases (p38 MAPK). The levels of reduced glutathione, malondialdehyde and superoxide dismutase were also measured. Pregabalin decreased testosterone level while FSH, LH and prolactin showed a significant increase. It also produced genotoxicity through reversal of the BAX/BCL2 ratio; increased p38 MAPK level and induction of oxidative stress markers. The concomitant administration of vitamin E significantly reduced all the previously mentioned biochemical and hormonal adverse effects caused by pregabalin. Pregabalin can adversely affect male fertility particularly in addicts and patients who are being treated with it for long periods as those suffering from neuropathies and seizures. Antioxidants like vitamin E could have a role in amelioration.
Collapse
|