1
|
Rayginia TP, Keerthana CK, Shifana SC, Pellissery MJ, Abhishek A, Anto RJ. Phytochemicals as Potential Lead Molecules against Hepatocellular Carcinoma. Curr Med Chem 2024; 31:5199-5221. [PMID: 38213177 DOI: 10.2174/0109298673275501231213063902] [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: 08/03/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 01/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of liver cancer, accounting for 85-90% of liver cancer cases and is a leading cause of cancer-related mortality worldwide. The major risk factors for HCC include hepatitis C and B viral infections, along with chronic liver diseases, such as cirrhosis, fibrosis, and non-alcoholic steatohepatitis associated with metabolic syndrome. Despite the advancements in modern medicine, there is a continuous rise in the annual global incidence rate of HCC, and it is estimated to reach >1 million cases by 2025. Emerging research in phytomedicine and chemotherapy has established the anti-cancer potential of phytochemicals, owing to their diverse biological activities. In this review, we report the major phytochemicals that have been explored in combating hepatocellular carcinoma and possess great potential to be used as an alternative or in conjunction with the existing HCC treatment modalities. An overview of the pre-clinical observations, mechanism of action and molecular targets of some of these phytochemicals is also incorporated.
Collapse
Affiliation(s)
- Tennyson Prakash Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, 695011, India
| | - Chenicheri Kizhakkeveettil Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, 695011, India
| | | | - Maria Joy Pellissery
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Ajmani Abhishek
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, Kerala, 695317, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, Kerala, 695317, India
| |
Collapse
|
2
|
Sahin TK, Bilir B, Kucuk O. Modulation of inflammation by phytochemicals to enhance efficacy and reduce toxicity of cancer chemotherapy. Crit Rev Food Sci Nutr 2023; 63:2494-2508. [DOI: https:/doi.org/10.1080/10408398.2021.1976721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Taha Koray Sahin
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Birdal Bilir
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Aghabozorgi R, Hesam M, Zahed G, Babaee M, Hashemi M, Rayegani SM. Efficacy of Duloxetine on electrodiagnostic findings of Paclitaxel-induced peripheral neuropathy, does it have a prophylactic effect? A randomized clinical trial. Anticancer Drugs 2023; 34:680-685. [PMID: 36730548 DOI: 10.1097/cad.0000000000001429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study aimed to evaluate the efficacy of Duloxetine on electrodiagnostic findings of Paclitaxel-induced peripheral neuropathy in patients with breast cancer. This randomized, double-blind clinical trial was conducted on 40 patients with breast cancer who received Paclitaxel as their first chemotherapy session. All the patients were randomly allocated into two groups, intervention (20 subjects) and placebo (20 subjects). The intervention group received 30 mg duloxetine/day in the first week, followed by 60 mg (twice daily) until 8 weeks. The patient neurotoxicity questionnaire (PNQ) was used to evaluate the severity of neuropathy. Nerve conduction study was also performed. The evaluations were performed at the baseline and 8 weeks after the treatment. Out of 20 subjects in the placebo group, 10 (50%) patients had neurotoxicity (two milds, three moderate, four severe, and one incapacitated), according to PNQ. However, in the duloxetine group, two patients had mild neurotoxicity ( P = 0.03). Significant differences between groups related to the mean of Median Sensory Latency ( P <0.001), Median Motor Latency ( P < 0.001), and Median Motor velocity ( P = 0.001) were reported. However, the relative risk of polyneuropathy between the two groups (relative risk: 1) was not significant. Regarding the results, duloxetine could be an effective treatment for preventing paclitaxel-induced peripheral neuropathy in patients with breast cancer, and an electrodiagnostic study confirmed this effect.
Collapse
Affiliation(s)
| | - Marzieh Hesam
- School of Medicine, Arak University of Medical Sciences, Arak
| | - Ghazal Zahed
- Child and Adolescent Psychiatry Division, Shahid Beheshti University of Medical Sciences
| | - Marzieh Babaee
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdis Hashemi
- Department of Physical medicine and Rehabilitation, Vancouver Island Health Authority
- International Collaboration on Repair Discoveries (ICORD) British Colombia, Vancouver, British Columbia, Canada
| | - Seyed Mansoor Rayegani
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Pouliquen DL, Trošelj KG, Anto RJ. Curcuminoids as Anticancer Drugs: Pleiotropic Effects, Potential for Metabolic Reprogramming and Prospects for the Future. Pharmaceutics 2023; 15:1612. [PMID: 37376060 DOI: 10.3390/pharmaceutics15061612] [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: 04/28/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The number of published studies on curcuminoids in cancer research, including its lead molecule curcumin and synthetic analogs, has been increasing substantially during the past two decades. Insights on the diversity of inhibitory effects they have produced on a multitude of pathways involved in carcinogenesis and tumor progression have been provided. As this wealth of data was obtained in settings of various experimental and clinical data, this review first aimed at presenting a chronology of discoveries and an update on their complex in vivo effects. Secondly, there are many interesting questions linked to their pleiotropic effects. One of them, a growing research topic, relates to their ability to modulate metabolic reprogramming. This review will also cover the use of curcuminoids as chemosensitizing molecules that can be combined with several anticancer drugs to reverse the phenomenon of multidrug resistance. Finally, current investigations in these three complementary research fields raise several important questions that will be put among the prospects for the future research related to the importance of these molecules in cancer research.
Collapse
Affiliation(s)
- Daniel L Pouliquen
- Université d'Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Koraljka Gall Trošelj
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ruby John Anto
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram 695317, India
| |
Collapse
|
5
|
Ali YA, Soliman HA, Abdel-Gabbar M, Ahmed NA, Attia KAA, Shalaby FM, El-Nahass ES, Ahmed OM. Rutin and Hesperidin Revoke the Hepatotoxicity Induced by Paclitaxel in Male Wistar Rats via Their Antioxidant, Anti-Inflammatory, and Antiapoptotic Activities. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:2738351. [PMID: 37275575 PMCID: PMC10238143 DOI: 10.1155/2023/2738351] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 06/07/2023]
Abstract
Paclitaxel, one of the most effective chemotherapeutic drugs, is used to treat various cancers but it is exceedingly toxic when used long-term and can harm the liver. This study aimed to see if rutin, hesperidin, and their combination could protect male Wistar rats against paclitaxel (Taxol)-induced hepatotoxicity. Adult male Wistar rats were subdivided into 5 groups (each of six rats). The normal group was orally given the equivalent volume of vehicles for 6 weeks. The paclitaxel-administered control group received intraperitoneal injection of paclitaxel at a dose of 2 mg/Kg body weight twice a week for 6 weeks. Treated paclitaxel-administered groups were given paclitaxel similar to the paclitaxel-administered control group together with oral supplementation of rutin, hesperidin, and their combination at a dose of 10 mg/Kg body weight every other day for 6 weeks. The treatment of paclitaxel-administered rats with rutin and hesperidin significantly reduced paclitaxel-induced increases in serum alanine transaminase, aspartate transaminase, lactate dehydrogenase, alkaline phosphatase, and gamma-glutamyl transferase activities as well as total bilirubin level and liver lipid peroxidation. However, the levels of serum albumin, liver glutathione content, and the activities of liver superoxide dismutase and glutathione peroxidase increased. Furthermore, paclitaxel-induced harmful hepatic histological changes (central vein and portal area blood vessel congestion, fatty changes, and moderate necrotic changes with focal nuclear pyknosis, focal mononuclear infiltration, and Kupffer cell proliferation) were remarkably enhanced by rutin and hesperidin treatments. Moreover, the elevated hepatic proapoptotic mediator (caspase-3) and pro-inflammatory cytokine (tumor necrosis factor-α) expressions were decreased by the three treatments in paclitaxel-administered rats. The cotreatment with rutin and hesperidin was the most effective in restoring the majority of liver function and histological integrity. Therefore, rutin, hesperidin, and their combination may exert hepatic protective effects in paclitaxel-administered rats by improving antioxidant defenses and inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Yasmine A. Ali
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Hanan A. Soliman
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Mohamed Abdel-Gabbar
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Noha A. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Kandil A. A. Attia
- Clinical Nutrition Department, College of Applied Medical Sciences, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
- Department of Evaluation of Natural Resources, Environmental Studies and Research Institute, El-Sadat City University, El-Sadat City 32897, Egypt
| | - Fatma M. Shalaby
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - El-Shaymaa El-Nahass
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
6
|
Talib WH, Awajan D, Hamed RA, Azzam AO, Mahmod AI, AL-Yasari IH. Combination Anticancer Therapies Using Selected Phytochemicals. Molecules 2022; 27:5452. [PMID: 36080219 PMCID: PMC9458090 DOI: 10.3390/molecules27175452] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is still one of the most widespread diseases globally, it is considered a vital health challenge worldwide and one of the main barriers to long life expectancy. Due to the potential toxicity and lack of selectivity of conventional chemotherapeutic agents, discovering alternative treatments is a top priority. Plant-derived natural products have high potential in cancer treatment due to their multiple mechanisms of action, diversity in structure, availability in nature, and relatively low toxicity. In this review, the anticancer mechanisms of the most common phytochemicals were analyzed. Furthermore, a detailed discussion of the anticancer effect of combinations consisting of natural product or natural products with chemotherapeutic drugs was provided. This review should provide a strong platform for researchers and clinicians to improve basic and clinical research in the development of alternative anticancer medicines.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Dima Awajan
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Reem Ali Hamed
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Aya O. Azzam
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 964, Iraq
| |
Collapse
|
7
|
Kim J, You HJ, Youn C. SCARA3 inhibits cell proliferation and EMT through AKT signaling pathway in lung cancer. BMC Cancer 2022; 22:552. [PMID: 35578316 PMCID: PMC9112459 DOI: 10.1186/s12885-022-09631-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Scavenger receptor class A member 3 (SCARA3) is decreased in prostate cancer and myeloma. However, functions of SCARA3 in various cancers remain unclear. In this study, we tried to evaluate the functional study of SCARA3 in lung cancer. METHODS The expression level of SCARA3 in the TCGA-database, lung cancer tissue microarray and lung cancer cells and the prognosis of lung cancer patients were measured. Lung cancer tissue microarray was analyzed pathologically using immunohistochemistry, and quantitative analysis of SCARA3 in normal lung cells and lung cancer cells was analyzed using western blot analysis. Survival curves for lung cancer patients were prepared with the Kaplan-Meier method. Migration and invasion of SCARA3 overexpressed lung cancer cells were determined using a Transwell chamber system. Proliferation of lung cancer cells was determined based on cell viability assay using cell culture in vitro and a tumorigenicity model of BALB/C nude mouse in vivo. RESULTS The expression of SCARA3 was abnormally reduced in TCGA-database, lung tissue microarray, and various lung cancer cells. However, overexpression of SCARA3 reduced the proliferation of lung cancer. The ability of SCARA3 to inhibit cancer cell proliferation was maintained even in vivo using a mouse xenograft model. In addition, overexpression of SCARA3 reduced migration and invasion ability of lung cancer cells and induced decreases of EMT markers such as β-catenin, vimentin, and MMP9. We aimed to prove the role of SCARA3 in the treatment of Lung cancer, and shown that the expression level of SCARA3 is important in cancer treatment using cisplatin. The enhancement of the effect of cisplatin according to SCARA3 overexpression is via the AKT and JNK pathways. CONCLUSIONS This study confirmed an abnormal decrease in SCARA3 in lung cancer. Overexpression of SCARA3 potently inhibited tumors in lung cancer and induced apoptosis by increasing sensitivity of lung cancer to cisplatin. These results suggest that SCARA3 is a major biomarker of lung cancer and that the induction of SCARA3 overexpression can indicate an effective treatment.
Collapse
Affiliation(s)
- Jeeho Kim
- Laboratory of Genomic Instability and Cancer therapeutics and Department of Pharmacology, Chosun University School of Medicine, 375 Seosuk-Dong, Gwangju, 501-759, South Korea.,Department of Pharmacology, Chosun University School of Medicine, 375 Seosuk-dong, Gwangju, 501-759, South Korea
| | - Ho Jin You
- Laboratory of Genomic Instability and Cancer therapeutics and Department of Pharmacology, Chosun University School of Medicine, 375 Seosuk-Dong, Gwangju, 501-759, South Korea. .,Department of Pharmacology, Chosun University School of Medicine, 375 Seosuk-dong, Gwangju, 501-759, South Korea.
| | - Chakyung Youn
- Department of Meridian & Acupoint∙Diagnosis College of Korean Medicine, Dongshin University 67, Dongsindae-gil, Naju-si, Jeollanam-do, Republic of Korea.
| |
Collapse
|
8
|
Zhou ZW, Long HZ, Xu SG, Li FJ, Cheng Y, Luo HY, Gao LC. Therapeutic Effects of Natural Products on Cervical Cancer: Based on Inflammatory Pathways. Front Pharmacol 2022; 13:899208. [PMID: 35645817 PMCID: PMC9136176 DOI: 10.3389/fphar.2022.899208] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/09/2022] Open
Abstract
Inflammation is a protective response of the body to an irritant. When an inflammatory response occurs, immune cells are recruited to the injury, eliminating the irritation. The excessive inflammatory response can cause harm to the organism. Inflammation has been found to contribute to cervical cancer if there is a problem with the regulation of inflammatory response. Cervical cancer is one of the most common malignant tumors globally, and the incidence tends to be younger. The harm of cervical cancer cannot be ignored. The standard treatments for cervical cancer include surgery, radiotherapy and chemotherapy. However, the prognosis for this treatment is poor, so it is urgent to find a safer and more effective treatment. Natural products are considered excellent candidates for the treatment of cervical cancer. In this review, we first describe the mechanisms by which inflammation induces cervical cancer. Subsequently, we highlight natural products that can treat cervical cancer through inflammatory pathways. We also introduce natural products for the treatment of cervical cancer in clinical trials. Finally, methods to improve the anticancer properties of natural products were added, and the development status of natural products was discussed.
Collapse
Affiliation(s)
- Zi-Wei Zhou
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Hui-Zhi Long
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Shuo-Guo Xu
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Feng-Jiao Li
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Yan Cheng
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Hong-Yu Luo
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Li-Chen Gao
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| |
Collapse
|
9
|
Massa S, Pagliarello R, Paolini F, Venuti A. Natural Bioactives: Back to the Future in the Fight against Human Papillomavirus? A Narrative Review. J Clin Med 2022; 11:jcm11051465. [PMID: 35268556 PMCID: PMC8911515 DOI: 10.3390/jcm11051465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Human papillomavirus (HPV) still represents an important threat to health worldwide. Better therapy in terms of further improvement of outcomes and attenuation of related side-effects is desirable. The pharmaceutical industry has always targeted natural substances-phytochemicals in particular-to identify lead compounds to be clinically validated and industrially produced as antiviral and anticancer drugs. In the field of HPV, numerous naturally occurring bioactives and dietary phytochemicals have been investigated as potentially valuable in vitro and in vivo. Interference with several pathways and improvement of the efficacy of chemotherapeutic agents have been demonstrated. Notably, some clinical trials have been conducted. Despite being endowed with general safety, these natural substances are in urgent need of further assessment to foresee their clinical exploitation. This review summarizes the basic research efforts conducted so far in the study of anti-HPV properties of bio-actives with insights into their mechanisms of action and highlights the variety of their natural origin in order to provide comprehensive mapping throughout the different sources. The clinical studies available are reported, as well, to highlight the need of uniformity and consistency of studies in the future to select those natural compounds that may be suited to clinical application.
Collapse
Affiliation(s)
- Silvia Massa
- Biotechnology Laboratory, Casaccia Research Center, Biotechnology and Agro-Industry Division, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy;
- Correspondence:
| | - Riccardo Pagliarello
- Biotechnology Laboratory, Casaccia Research Center, Biotechnology and Agro-Industry Division, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy;
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy
| | - Francesca Paolini
- HPV-Unit, Unità Operativa Semplice Dipartimentale (UOSD) Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (F.P.); (A.V.)
| | - Aldo Venuti
- HPV-Unit, Unità Operativa Semplice Dipartimentale (UOSD) Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (F.P.); (A.V.)
| |
Collapse
|
10
|
Jamialahmadi T, Guest PC, Afshari AR, Majeed M, Sahebkar A. Testing the Effect of Curcumin on Proliferative Capacity of Colorectal Cancer Cells. Methods Mol Biol 2022; 2343:287-298. [PMID: 34473331 DOI: 10.1007/978-1-0716-1558-4_20] [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] [Indexed: 02/12/2023]
Abstract
This chapter presents a protocol for studying the effects of curcumin in a colorectal cell line and a mouse model of colitis-associated colon carcinogenesis. The protocol using the CT26 cell line incorporates cell proliferation, migration, invasion, spheroid formation, cell cycle, polymerase chain reaction (PCR), and western blot analyses. For the mouse model, this involved a macroscopic and histological examination of the colon and assays for oxidative damage markers.
Collapse
Affiliation(s)
- Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
- Charlesworth House, Debden, Essex, UK
| | - Amir R Afshari
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Pellegrini E, Multari G, Gallo FR, Vecchiotti D, Zazzeroni F, Condello M, Meschini S. A natural product, voacamine, sensitizes paclitaxel-resistant human ovarian cancer cells. Toxicol Appl Pharmacol 2022; 434:115816. [PMID: 34856211 DOI: 10.1016/j.taap.2021.115816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/02/2021] [Accepted: 11/26/2021] [Indexed: 11/12/2022]
Abstract
Most women with ovarian cancer are treated with chemotherapy before or after surgery. Unfortunately, chemotherapy treatment can cause negative side effects and the onset of multidrug resistance (MDR). The aim of this study is to evaluate the chemosensitizing effect of a natural compound, voacamine (VOA), in ovarian (A2780 DX) and colon (LoVo DX) cancer drug-resistant cell lines which overexpress P-glycoprotein (P-gp), in combination with paclitaxel (PTX), or doxorubicin (DOX) or 5-fluorouracil (5-FU). VOA, a bisindole alkaloid extracted from Peschiera fuchsiaefolia, has already been shown to be effective in enhancing the effect of doxorubicin, because it interferes with the P-gp function. Ovarian cancer cytotoxicity test shows that single treatments with VOA, DOX and PTX do not modify cell viability, while pretreatment with VOA, and then PTX or DOX for 72 h, induces a decrease. In colon cancer, since 5-FU is not a-substrate for P-gp, VOA has no sensitizing effect while in VOA + DOX there is a decrease in viability. Annexin V/PI test, cell cycle analysis, activation of cleaved PARP1 confirm that VOA plus PTX induce apoptotic cell death. Confocal microscopy observations show the different localization of NF-kB after treatment with VOA + PTX, confirming the inhibition of nuclear translocation induced by VOA pretreatment. Our data show the specific effect of VOA which only works on drugs known to be substrates of P-gp.
Collapse
Affiliation(s)
- Evelin Pellegrini
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy
| | - Giuseppina Multari
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy
| | - Francesca Romana Gallo
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Condello
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy..
| | - Stefania Meschini
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy..
| |
Collapse
|
12
|
Wroński P, Wroński S, Kurant M, Malinowski B, Wiciński M. Curcumin May Prevent Basement Membrane Disassembly by Matrix Metalloproteinases and Progression of the Bladder Cancer. Nutrients 2021; 14:32. [PMID: 35010907 PMCID: PMC8746354 DOI: 10.3390/nu14010032] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 12/25/2022] Open
Abstract
Authors present a review of crucial mechanisms contributing to the invasion of the basement membrane (BM) of the urothelium by cancer cells and to the progression of bladder cancer (BC). The breeching of the urothelial BM, facilitated by an aberrant activation of matrix metalloproteinases (MMP) is particularly perilous. Inhibition of activation of these proteinases constitutes a logic opportunity to restrain progression. Because of limited efficacy of current therapeutic methods, the search for the development of alternative approaches constitutes "the hot spot" of modern oncology. Recent studies revealed significant anticancer potential of natural phytochemicals. Especially, curcumin has emerged as a one of the most promising phytochemicals and showed its efficacy in several human malignancies. Therefore, this article addresses experimental and clinical data indicating multi-directional inhibitory effect of curcumin on the growth of bladder cancer. We particularly concentrate on the mechanisms, by which curcumin inhibits the MMP's activities, thereby securing BM integrity and alleviating the eventual cancer invasion into the bladder muscles. Authors review the recently accumulating data, that curcumin constitutes a potent factor contributing to the more effective treatment of the bladder cancer.
Collapse
Affiliation(s)
- Paweł Wroński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
- Department of Oncological Urology, The Franciszek Lukaszczyk Oncology Center, Romanowskiej 2, 85-796 Bydgoszcz, Poland
| | - Stanisław Wroński
- Department of Urology, Jan Biziel Memorial University Hospital, Ujejskiego 75, 85-168 Bydgoszcz, Poland;
| | - Marcin Kurant
- Department of Urology, District Hospital, 10 Lesna Street, 89-600 Chojnice, Poland;
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
| | - Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (P.W.); (B.M.)
| |
Collapse
|
13
|
Anirudhan TS, Varghese S, Manjusha V. Hyaluronic acid coated Pluronic F127/Pluronic P123 mixed micelle for targeted delivery of Paclitaxel and Curcumin. Int J Biol Macromol 2021; 192:950-957. [PMID: 34662655 DOI: 10.1016/j.ijbiomac.2021.10.061] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
The hydrophobicity of most of the anticancer drugs offers a great challenge in selecting a system for their effective transport. Here comes the importance of micelles that offers a hydrophobic core for incorporating these drugs. In this study, Hyaluronic Acid coated Pluronic mixed micelle loaded with Paclitaxel and Curcumin was designed and evaluated its anticancer activity in MCF-7 cells. Pluronic F127 (PF127) and Pluronic P123 (PP123) were taken for preparing the mixed micelles. The targeting ligand folic acid (FA) was conjugated to one end of PP123 forming FA-PP. The end hydroxyl groups of PF127 were oxidized to aldehyde groups resulted in PF-CHO. Mixed micelles were prepared from PF-CHO and FA-PP and the end aldehyde groups were used for coating the micelles with hyaluronic acid. The material was characterized using FTIR, H1NMR, DLS, FE-SEM and TEM. The coated micelles showed spherical shape with drug loading efficiency of 50.15 and 65.05% for Paclitaxel and Curcumin, respectively. In vitro drug release was studied at pH 5.5 and 7.4. Dual drug-loaded material showed higher in-vitro anticancer activity than free Paclitaxel and Curcumin. The results suggested that synthesized mixed micelle with dual drugs showed great potential for targeted delivery to MCF-7 cells.
Collapse
Affiliation(s)
- T S Anirudhan
- Department of Chemistry, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Trivandrum 695 581, India.
| | - Susan Varghese
- Department of Chemistry, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Trivandrum 695 581, India
| | - V Manjusha
- Department of Chemistry, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Trivandrum 695 581, India
| |
Collapse
|
14
|
Ke Y, Yang X, Luo D. miR-193a-3p Overexpression Inhibits Proliferation and Enhances Paclitaxel Chemosensitivity in Human Non-Small-Cell Lung Cancer Cells. INT J PHARMACOL 2021. [DOI: 10.3923/ijp.2021.541.548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Hanna DH, R. Saad G. Induction of mitochondria mediated apoptosis in human ovarian cancer cells by folic acid coated tin oxide nanoparticles. PLoS One 2021; 16:e0258115. [PMID: 34597348 PMCID: PMC8486119 DOI: 10.1371/journal.pone.0258115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE This study aims to prepare folic acid coated tin oxide nanoparticles (FA-SnO2 NPs) for specifically targeting human ovarian cancer cells with minimum side effects against normal cells. METHODS The prepared FA-SnO2 NPs were characterized by FT-IR, UV-vis spectroscopy, XRD, SEM and TEM. The inhibition effects of FA-SnO2 NPs against SKOV3 cancer cell were tested by MTT and LDH assay. Apoptosis induction in FA-SnO2 NPs treated SKOV3 cells were investigated using Annexin V/PI, AO/EB and Comet assays and the possible mechanisms of the cytotoxic action were studied by Flow cytometry, qRT-PCR, Immunohistochemistry, and Western blotting analyses. The effects of FA-SnO2 NPs on reactive oxygen species generation in SKOV3 cells were also examined. Additionally, the safety of utilization FA-SnO2 NPs were studied in vivo using Wister rats. RESULTS The obtained FA-SnO2 NPs displayed amorphous spherical morphology with an average diameter of 157 nm and a zeta potential value of -24 mV. Comparing to uncoated SnO2 NPs, FA-SnO2 NPs had a superior inhibition effect towards SKOV3 cell growth that was suggested to be mediated through higher reactive oxygen species generation. It was showed that FA-SnO2 NPs increased significantly the % of apoptotic cells in the sub- G1 and G2/M phases with a higher intensity comet nucleus in SKOV3 treated cells. Furthermore, FA-SnO2 NPs was significantly increased the expression levels of P53, Bax, and cleaved Caspase-3 and accompanied with a significant decrease of Bcl-2 in the treated SKOV3 cells. CONCLUSION Overall, the results suggested that an increase in cellular FA-SnO2 NPs internalization resulted in a significant induced cytotoxicity in SKOV3 cancer cells in dose-dependent mode through ROS-mediated cell apoptosis that may have occurred through mitochondrial pathway. Additionally, the results confirmed the safety of utilization FA-SnO2 NPs against living systems. So, FA-SnO2 NPs with a specific targeting moiety may be a promising therapeutic candidate for human ovarian cancer.
Collapse
Affiliation(s)
- Demiana H. Hanna
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | - Gamal R. Saad
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| |
Collapse
|
16
|
Sahin TK, Bilir B, Kucuk O. Modulation of inflammation by phytochemicals to enhance efficacy and reduce toxicity of cancer chemotherapy. Crit Rev Food Sci Nutr 2021; 63:2494-2508. [PMID: 34529530 DOI: 10.1080/10408398.2021.1976721] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Treatment of cancer with chemotherapeutic drugs is associated with numerous adverse effects as well as the eventual development of resistance to chemotherapy. There is a great need for complementary therapies such as botanicals and nutritional supplements with little or no side effects that prevent resistance to chemotherapy and reduce its adverse effects. Inflammation plays a major role in the development of chemoresistance and the adverse effects of chemotherapy. Phytochemicals have well-established anti-inflammatory effects; thus, they could be used as complementary therapies along with chemotherapy to increase its efficacy and reduce its toxicity. Botanical compounds inhibit the NF-κB signaling pathway, which plays an important role in the generation of inflammation, chemotherapy resistance, and modulation of cell survival and apoptosis. Botanicals have previously been studied extensively for their cancer chemopreventive activities and are generally considered safe for human consumption. The present review focuses on the modulation of inflammation by phytochemicals and their role in increasing the efficacy and reducing the toxicity of cancer chemotherapy.
Collapse
Affiliation(s)
- Taha Koray Sahin
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Birdal Bilir
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Haritha NH, Nawab A, Vijayakurup V, Anto NP, Liju VB, Alex VV, Amrutha AN, Aiswarya SU, Swetha M, Vinod BS, Sundaram S, Guijarro MV, Herlevich T, Krishna A, Nestory NK, Bava SV, Sadasivan C, Zajac-Kaye M, Anto RJ. Targeting Thymidylate Synthase Enhances the Chemosensitivity of Triple-Negative Breast Cancer Towards 5-FU-Based Combinatorial Therapy. Front Oncol 2021; 11:656804. [PMID: 34336653 PMCID: PMC8320437 DOI: 10.3389/fonc.2021.656804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/28/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The ongoing treatment modalities for breast cancer (BC) primarily rely on the expression status of ER, PR and HER-2 receptors in BC tissues. Our strategy of chemosensitization provides new insights to counter chemoresistance, a major obstacle that limits the benefits of chemotherapy of mammary cancers. METHODS By utilizing a murine breast cancer model employing NSG mice bearing orthotopic triple-negative breast cancer (TNBC) xenografts, we have evaluated the ability of phytochemical curcumin in chemosensitizing BC to 5-Fluorouracil (5-FU) chemotherapy and the differential modulations of cellular events in response to this strategy, independent of their receptor status. RESULTS A significant synergistic antitumor potential was observed in the murine model with a sub-optimal dose treatment of 5-FU plus curcumin, as evaluated by a reduction in the tumor-related parameters. We authenticated the pivotal role of thymidylate synthase (TS) in regulating the 5-FU-curcumin synergism using the TNBC pre-clinical model. Our study also confirmed the pharmacological safety of this chemotherapeutic plus phytoactive combination using acute and chronic toxicity studies in Swiss albino mice. Subsequently, the molecular docking analysis of curcumin binding to TS demonstrated the affinity of curcumin towards the cofactor-binding site of TS, rather than the substrate-binding site, where 5-FU binds. Our concomitant in vivo and in silico evidence substantiates the superior therapeutic index of this combination. CONCLUSION This is the first-ever pre-clinical study portraying TS as the critical target of combinatorial therapy for mammary carcinomas and therefore we recommend its clinical validation, especially in TNBC patients, who currently have limited therapeutic options.
Collapse
Affiliation(s)
- Nair Hariprasad Haritha
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Akbar Nawab
- Department of Anatomy and Cell Biology, Cancer and Genetics Research Complex, University of Florida, Gainesville, FL, United States
| | - Vinod Vijayakurup
- Department of Anatomy and Cell Biology, Cancer and Genetics Research Complex, University of Florida, Gainesville, FL, United States
| | - Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Vijayasteltar B. Liju
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Vijai V. Alex
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | | - Mundanattu Swetha
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Balachandran S. Vinod
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Kottayam, India
| | - Maria V. Guijarro
- Department of Anatomy and Cell Biology, Cancer and Genetics Research Complex, University of Florida, Gainesville, FL, United States
| | - Thomas Herlevich
- Department of Anatomy and Cell Biology, Cancer and Genetics Research Complex, University of Florida, Gainesville, FL, United States
| | - Archana Krishna
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Nesteena K. Nestory
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Smitha V. Bava
- Department of Biotechnology, University of Calicut, Malappuram, India
| | | | - Maria Zajac-Kaye
- Department of Anatomy and Cell Biology, Cancer and Genetics Research Complex, University of Florida, Gainesville, FL, United States
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
18
|
Nawara HM, Afify SM, Hassan G, Zahra MH, Seno A, Seno M. Paclitaxel-Based Chemotherapy Targeting Cancer Stem Cells from Mono- to Combination Therapy. Biomedicines 2021; 9:biomedicines9050500. [PMID: 34063205 PMCID: PMC8147479 DOI: 10.3390/biomedicines9050500] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutical agent commonly used to treat several kinds of cancer. PTX is known as a microtubule-targeting agent with a primary molecular mechanism that disrupts the dynamics of microtubules and induces mitotic arrest and cell death. Simultaneously, other mechanisms have been evaluated in many studies. Since the anticancer activity of PTX was discovered, it has been used to treat many cancer patients and has become one of the most extensively used anticancer drugs. Regrettably, the resistance of cancer to PTX is considered an extensive obstacle in clinical applications and is one of the major causes of death correlated with treatment failure. Therefore, the combination of PTX with other drugs could lead to efficient therapeutic strategies. Here, we summarize the mechanisms of PTX, and the current studies focusing on PTX and review promising combinations.
Collapse
Affiliation(s)
- Hend M. Nawara
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Said M. Afify
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Division of Biochemistry, Chemistry Department, Faculty of Science, Menoufia University, Menoufia 32511, Egypt
| | - Ghmkin Hassan
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Department of Microbiology and Biochemistry, Faculty of Pharmacy, Damascus University, Damascus 10769, Syria
| | - Maram H. Zahra
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Akimasa Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
| | - Masaharu Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (H.M.N.); (S.M.A.); (G.H.); (M.H.Z.); (A.S.)
- Correspondence: ; Tel.: +81-86-251-8216
| |
Collapse
|
19
|
Harikrishnan A, Khanna S, Veena V. Design of New Improved Curcumin Derivatives to Multi-targets of Cancer and Inflammation. Curr Drug Targets 2021; 22:573-589. [PMID: 32753008 DOI: 10.2174/1389450121666200804113745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Curcumin is a major active principle of Curcuma longa. There are more than 1700 citations in the Medline, reflecting various biological effects of curcumin. Most of these biological activities are associated with the antioxidant, anti-inflammatory and antitumor activity of the molecule. Several reports suggest various targets of natural curcumin that include growth factors, growth factor receptor, cytokines, enzymes and gene regulators of apoptosis. This review focuses on the improved curcumin derivatives that target the cancer and inflammation. METHODOLOGY In this present review, we explored the anticancer drugs with curcumin-based drugs under pre-clinical and clinical studies with critical examination. Based on the strong scientific reports of patentable and non-patented literature survey, we have investigated the mode of the interactions of curcumin-based molecules with the target molecules. RESULTS Advanced studies have added new dimensions of the molecular response of cancer cells to curcumin at the genomic level. However, poor bioavailability of the molecule seems to be the major limitation of the curcumin. Several researchers have been involved to improve the curcumin derivatives to overcome this limitation. Sufficient data of clinical trials to various cancers that include multiple myeloma, pancreatic cancer and colon cancer, have also been discussed. CONCLUSION The detailed analysis of the structure-activity relationship (SAR) and common synthesis of curcumin-based derivatives have been discussed in the review. Utilising the predictions of in silico coupled with validation reports of in vitro and in vivo studies have concluded many targets for curcumin. Among them, cancer-related inflammation genes regulating curcumin-based molecules are a very promising target to overcome hurdles in the multimodality therapy of cancer.
Collapse
Affiliation(s)
- A Harikrishnan
- Department of Chemistry, School of Arts and Sciences, Vinayaka Mission Research Foundation-Aarupadai Veedu (VMRF-AV) campus, Paiyanoor, Chennai-603104, Tamil Nadu, India
| | - Sunali Khanna
- Nair Hospital Dental College, Municipal Corporation of Greater Mumbai, Mumbai, 400 008, India
| | - V Veena
- Department of Biotechnology, School of Applied Sciences, REVA University, Rukmini knowledge park, Kattigenahalli, Yelahanka, Bengaluru - 5600 064. Karnataka State, India
| |
Collapse
|
20
|
Dai X, Hao J, Feng Y, Wang J, Li Q, Ma C, Wang X, Chang Z, Wang S, Wang Y. Revealing Changes in Curcumin Bioavailability using Vitamin C as an Enhancer by HPLC-MS/MS. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916666191220150039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Curcumin (CUR), a natural isolated compound from turmeric, helps in
fighting many diseases, but the broad application of curcumin has been limited ascribed to low bioavailability.
Objective:
The aim of this study is to pursue the enhancement of curcumin bioavailability through coadministration
of vitamin C.
Methods:
Such purpose was achieved through the analysis of curcumin pharmacokinetics by highperformance
liquid chromatography coupled with electrospray ionization - tandem mass spectrometry
(HPLC - ESI - MS/MS). The plasma was separated on a C18 reverse-phase column using acetonitrile
and ammonium formate solution (pH 6.5; 2.0 mM) at 0.8 mL/min. MS/MS detection was carried out in
negative mode using mass patterns of m/z 367.0 > 216.7 for curcumin and m/z 265.2 > 223.9 for internal
standard (honokiol).
Results:
Successful application of the proposed method in the pharmacokinetic study presented clear
changes in key pharmacokinetic parameters, including the growth of AUC (0-t) up to 2.4 times, a 2.2-
fold increase of Cmax, 2.2-fold loss of CL, and 1.5-fold diminishment of t1/2.
Conclusion:
An HPLC-ESI-MS/MS method for the determination of curcumin in rat plasma and validated
the improvement of bioavailability of curcumin through co-administration of vitamin C was determined.
These changes were reasoned to the inhibition of lipid peroxidation induced by the use of
vitamin C. Such a simple strategy is possible to become an alternative for enhancing curcumin efficiency
in practice.
Collapse
Affiliation(s)
- Xufen Dai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Jiaxue Hao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Ying Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Jing Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Qiannan Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Cuicui Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Xing Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Zhongman Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Shixiang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| | - Yuxin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi’an 710069,China
| |
Collapse
|
21
|
Involvement of p53-dependent apoptosis signal in antitumor effect of Colchicine on human papilloma virus (HPV)-positive human cervical cancer cells. Biosci Rep 2021; 40:222342. [PMID: 32163135 PMCID: PMC7098170 DOI: 10.1042/bsr20194065] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/28/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Colchicine, a plant-derived alkaloid with relatively low toxicity on normal human epidermal keratinocytes (HEKn), has selective inhibitory effect on the growth of CaSki (HPV16-positive) and HeLa (HPV18-positive) human cervical cancer cell lines via the induction of apoptosis. Colchicine (2.5, 5.0 and 10.0 ng/ml) significantly reduced the expression of human papilloma virus (HPV) 16 E6/E7 mRNA and protein in CaSki and HeLa cells. Moreover, reduced expression of E6 and E7 induced by Colchicine resulted in the up-regulation of tumor suppressor proteins, p53 and Rb, as well as down-regulation of phospho Rb (pRb) protein. In addition, Bax, cytosolic cytochrome c and cleaved caspase-3 protein were increased while Bcl-2 protein was decreased significantly by 48 h of Colchicine treatment. These results implied that Colchicine could be explored as a potent candidate agent for the treatment and prevention of HPV-associated cervical cancer without deleterious effects.
Collapse
|
22
|
Yardım A, Kandemir FM, Çomaklı S, Özdemir S, Caglayan C, Kucukler S, Çelik H. Protective Effects of Curcumin Against Paclitaxel-Induced Spinal Cord and Sciatic Nerve Injuries in Rats. Neurochem Res 2021; 46:379-395. [PMID: 33201400 DOI: 10.1007/s11064-020-03174-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 01/09/2023]
Abstract
Paclitaxel (PTX) is an antineoplastic agent commonly used in the treatment of solid tumors and is known to cause dose-limiting peripheral neurotoxicity. This study was performed to evaluate the protective effect of curcumin (CUR) against PTX-induced spinal cord and sciatic nerve injuries in rats. The rats were administered PTX (2 mg/kg, BW) intraperitoneally for the first 5 consecutive days followed by administration of CUR (100 and 200 mg/kg, BW daily in corn oil) orally for 10 days. Our results showed that CUR significantly reduced mRNA expression levels of NF-κB, TNF-α, IL-6, iNOS and GFAP whereas caused an increase in levels of Nrf2, HO-1 and NQO1 in the spinal cord and sciatic nerve of PTX-induced rats. In addition, CUR suppressed the activation of apoptotic and autophagic pathways by increasing Bcl-2 and Bcl-xL, and decreasing p53, caspase-3, Apaf-1, LC3A, LC3B and beclin-1 mRNA expression levels. The results showed that CUR also maintained the spinal cord and sciatic nerve histological architecture and integrity by both LFB staining and H&E staining. Immunohistochemical expressions of 8-OHdG, caspase-3 and LC3B in the PTX-induced spinal cord tissue were decreased after administration of CUR. Taken together, our findings demonstrated that CUR has protective effects on PTX-induced spinal cord and sciatic nerve injuries in rats.
Collapse
Affiliation(s)
- Ahmet Yardım
- Department of Neurosurgery, Private Buhara Hospital, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, 25240, Erzurum, Turkey.
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240, Erzurum, Turkey
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, 25240, Erzurum, Turkey
| | - Cuneyt Caglayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, 12000, Bingöl, Turkey.
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, 25240, Erzurum, Turkey
| | - Hamit Çelik
- Department of Neurology, Private Buhara Hospital, Erzurum, Turkey
| |
Collapse
|
23
|
Kato K, Nagane M, Aihara N, Kamiie J, Miyanabe M, Hiraki S, Luo X, Nakanishi I, Shoji Y, Matsumoto KI, Yamashita T. Lipid-soluble polyphenols from sweet potato exert antitumor activity and enhance chemosensitivity in breast cancer. J Clin Biochem Nutr 2021; 68:193-200. [PMID: 34025021 PMCID: PMC8129977 DOI: 10.3164/jcbn.20-73] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022] Open
Abstract
Polyphenols are abundant in vegetables and fruit. They have been shown to have various antitumor, antioxidant, and anti-inflammatory effects. Here, we extracted the lipid-soluble fraction of polyphenols from fermented sweet potato (Ipomoea batatas). These lipid-soluble polyphenols mainly contained caffeic acid derivatives with strong antioxidant ability, which we hypothesized to affect diseases for which oxidative stress is a factor, such as cancer. We therefore investigated the antitumor and chemo-sensitizing effects of lipid-soluble polyphenols on E0771 murine breast cancer cells. The lipid-soluble polyphenols accumulated in the cells’ cytoplasm due to its high lipophilicity, and reduced reactive oxygen species through its strong antioxidant activity. The lipid-soluble polyphenols also arrested the cell cycle at G0/G1 by suppressing Akt activity, and enhanced the cytotoxicity of anticancer agents. In this model, lipid-soluble polyphenols inhibited tumor growth and enhanced the efficacy of chemotherapy drugs. These results suggest the potential of lipid-soluble polyphenols as a functional food to support cancer therapy.
Collapse
Affiliation(s)
- Kazuhiro Kato
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, 1-17-71, Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, 1-17-71, Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Naoyuki Aihara
- Laboratory of Veterinary Pathology, School of Veterinary Medicine, Azabu University, 1-17-71, Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Junichi Kamiie
- Laboratory of Veterinary Pathology, School of Veterinary Medicine, Azabu University, 1-17-71, Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Masakatsu Miyanabe
- Genuine R&D Co., Ltd., 2-36-12, Takamidai, Higashi-ku, Fukuoka 811-0215, Japan
| | - Shinobu Hiraki
- Genuine R&D Co., Ltd., 2-36-12, Takamidai, Higashi-ku, Fukuoka 811-0215, Japan
| | - Xiaolin Luo
- Genuine R&D Co., Ltd., 2-36-12, Takamidai, Higashi-ku, Fukuoka 811-0215, Japan
| | - Ikuo Nakanishi
- Quantitative RedOx Sensing Group, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences (NIRS), Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yoshimi Shoji
- Quantitative RedOx Sensing Group, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences (NIRS), Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ken-Ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences (NIRS), Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, 1-17-71, Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| |
Collapse
|
24
|
Sun Y, Ren J, Wang F. [6]-Gingerol impedes 7,12-dimethylbenz(a)anthracene-induced inflammation and cell proliferation-associated hamster buccal pouch carcinogenesis through modulating Nrf2 signaling events. J Biochem Mol Toxicol 2020; 35:e22689. [PMID: 33347680 DOI: 10.1002/jbt.22689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/22/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
The present study examines the chemopreventive role of [6]-gingerol, an active component of ginger, on 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis models. The HBP has been developed with an addition of 0.5% of DMBA to the HBP area three times per week, up to the end of the 16th experimental week. At the end of the experiment, we noticed 100% tumor incidence and precancerous lesions, such as dysplasia, hyperplasia, keratosis, and well-differentiated squamous cell carcinoma, in DMBA-induced HBP. Furthermore, we observed that [6]-gingerol inhibited the increased thiobarbituric acid-reactive substances and decreased antioxidant levels in DMBA-induced hamsters. Moreover, [6]-gingerol inhibits DMBA-exposed over expression of inflammatory markers (inducible nitric oxide synthase, interleukin [IL]-1β, IL-6, cyclooxygenase-2, and tumor necrosis factor-α) and cell proliferation markers (cyclin D1, proliferating cell nuclear antigen); induces proapoptotic markers in HBP. Nuclear factor erythroid-2-related factor-2 (Nrf2) is a major antioxidant transcription factor, which regulates the antioxidant gene-dependent scavenge of tumor proliferation and apoptosis. Overexpression of Nrf2 signaling plays a pivotal role and can be a novel target in preventing carcinogenesis. In this study, [6]-gingerol restores the DMBA-induced depletion of Nrf2 signaling and thereby prevents buccal pouch carcinogenesis in hamsters. These results point out that [6]-gingerol impedes the responses of inflammatory and cell proliferation-associated progression of cancer through the action of Nrf2 signaling.
Collapse
Affiliation(s)
- Yugang Sun
- Oral and maxillofacial surgery, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Jinmin Ren
- Health Management Center, Binzhou Municipal Hospital of Traditional Chinese Medicine, Binzhou, Shandong, China
| | - Fang Wang
- Department of Oncology, The Second People Hospital of Dezhou, Dezhou, Shandong, China
| |
Collapse
|
25
|
Martinovich GG, Martinovich IV, Vcherashniaya AV, Zenkov NK, Menshchikova EB, Cherenkevich SN. Chemosensitization of Tumor Cells by Phenolic Antioxidants: The Role of the Nrf2 Transcription Factor. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s000635092006010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
26
|
Curcumin, a Multifaceted Hormetic Agent, Mediates an Intricate Crosstalk between Mitochondrial Turnover, Autophagy, and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3656419. [PMID: 32765806 PMCID: PMC7387956 DOI: 10.1155/2020/3656419] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/01/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023]
Abstract
Curcumin has extensive therapeutic potential because of its antioxidant, anti-inflammatory, and antiproliferative properties. Multiple preclinical studies in vitro and in vivo have proven curcumin to be effective against various cancers. These potent effects are driven by curcumin's ability to induce G2/M cell cycle arrest, induce autophagy, activate apoptosis, disrupt molecular signaling, inhibit invasion and metastasis, and increase the efficacy of current chemotherapeutics. Here, we focus on the hormetic behavior of curcumin. Frequently, low doses of natural chemical products activate an adaptive stress response, whereas high doses activate acute responses like autophagy and cell death. This phenomenon is often referred to as hormesis. Curcumin causes cell death and primarily initiates an autophagic step (mitophagy). At higher doses, cells undergo mitochondrial destabilization due to calcium release from the endoplasmic reticulum, and die. Herein, we address the complex crosstalk that involves mitochondrial biogenesis, mitochondrial destabilization accompanied by mitophagy, and cell death.
Collapse
|
27
|
Combination Therapy with Nanomicellar-Curcumin and Temozolomide for In Vitro Therapy of Glioblastoma Multiforme via Wnt Signaling Pathways. J Mol Neurosci 2020; 70:1471-1483. [PMID: 32666415 DOI: 10.1007/s12031-020-01639-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/16/2020] [Indexed: 01/22/2023]
Abstract
Glioblastoma (GBM) is the most serious brain tumor and shows a high rate of drug resistance. Wnt signaling is a very important pathway in GBM that can activate/inhibit other pathways, such as apoptosis and autophagy. In this study, we evaluated the efficacy of a combination of temozolomide (TMZ) plus curcumin or nanomicellar-curcumin on the inhibition of GBM growth in vitro, via effects on autophagy, apoptosis, and the Wnt signaling pathway. Two concentrations of curcumin and nanomicellar-curcumin (i.e., 20 μM and 50 μM) alone, and in combination with TMZ (50 μM) were used to induce cytotoxicity in the U87 GBM cell line. Wnt signaling-, autophagy-, and apoptosis-related genes were assessed by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blots. All treatments (except 20 μM curcumin alone) significantly decreased the viability of U87 cells compared to controls. Curcumin (50 μM), nanomicellar-curcumin alone and in combination with TMZ significantly decreased the invasion and migration of U87 cells. Autophagy-related proteins (Beclin 1, LC3-I, LC3-II) were significantly increased. Apoptosis-related proteins (Bcl-2 and caspase 8) were also significantly increased, while Bax protein was significantly decreased. The expression levels of Wnt pathway-associated genes (β-catenin, cyclin D1, Twist, and ZEB1) were significantly reduced.
Collapse
|
28
|
Khatoon E, Banik K, Harsha C, Sailo BL, Thakur KK, Khwairakpam AD, Vikkurthi R, Devi TB, Gupta SC, Kunnumakkara AB. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin Cancer Biol 2020; 80:306-339. [DOI: 10.1016/j.semcancer.2020.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
|
29
|
Synthesis and Investigation of the Curcumin-Loaded Magnetic Lipid Nanoparticles and Their Cytotoxicity Assessment on Human Breast Carcinoma Cell Line. Jundishapur J Nat Pharm Prod 2020. [DOI: 10.5812/jjnpp.91886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
30
|
Hanna DH, Saad GR. Nanocurcumin: preparation, characterization and cytotoxic effects towards human laryngeal cancer cells. RSC Adv 2020; 10:20724-20737. [PMID: 35517737 PMCID: PMC9054308 DOI: 10.1039/d0ra03719b] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 05/11/2020] [Indexed: 11/21/2022] Open
Abstract
The aim of the present study was to prepare curcumin nanoparticles (nanocurcumin) by a sol-oil method to improve curcumin absorption and bioavailability, and to investigate the therapeutic effects of the prepared nanoparticles on the inhibition mechanisms towards human Hep-2 cancer cells. The nanoparticles were characterized by Fourier transform infrared spectroscopy, transmission electron microscopy, X-ray diffraction, and zeta potential analysis. The prepared curcumin nanoparticles possessed a narrow particle size distribution with an average diameter of 28 nm. The inhibition effects on the growth of human Hep-2 cells were investigated using neutral red uptake and lactate dehydrogenase assays. The results indicated that the nanocurcumin has a selective effect in inhibiting Hep-2 cell growth in a dose- and time-dependent mode with the most effective IC50 value (17 ± 0.31 μg ml−1) obtained after 48 h of incubation without any cytotoxic effects on normal cells. This IC50 value of nanocurcumin revealed a significant increase of early and late apoptotic cells with an intense comet nucleus of Hep-2 cells as a marker of DNA damage. Flow cytometry analysis of the progression of apoptosis in nanocurcumin Hep-2 treated cells showed that arresting in the cell cycle in the G2/M phase with increasing apoptotic cells in the sub-G1 phase. At the same time, real-time PCR analysis indicated that the treatment of Hep-2 cells with nanocurcumin resulted in upregulation of P53, Bax, and Caspase-3, whereas there was downregulation of Bcl-XL. These findings gave insights into understanding that the inhibition mechanisms of nanocurcumin on the proliferation of Hep-2 cancer cells was through the G2/M cell cycle arrest and the induction of apoptosis was dependent on Caspase-3 and p53 activation. However, in vivo studies with an animal model are essential to validate these results. The aim of this study was to prepare curcumin nanoparticles using a sol–oil method to improve curcumin absorption and bioavailability, and to investigate the therapeutic effect of the prepared nanoparticles on the inhibition mechanisms toward human Hep-2 cancer cells.![]()
Collapse
Affiliation(s)
- Demiana H. Hanna
- Department of Chemistry
- Faculty of Science
- Cairo University
- Giza 12613
- Egypt
| | - Gamal R. Saad
- Department of Chemistry
- Faculty of Science
- Cairo University
- Giza 12613
- Egypt
| |
Collapse
|
31
|
Duan FG, Wang MF, Cao YB, Dan Li, Li RZ, Fan XX, Khan I, Lai HL, Zhang YZ, Hsiao WWL, Yao XJ, Wu QB, Liu L, Tang YJ, Leung ELH. MicroRNA-421 confers paclitaxel resistance by binding to the KEAP1 3'UTR and predicts poor survival in non-small cell lung cancer. Cell Death Dis 2019; 10:821. [PMID: 31659154 PMCID: PMC6817891 DOI: 10.1038/s41419-019-2031-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/11/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022]
Abstract
MicroRNAs regulate post-transcriptional gene expression and play important roles in multiple cellular processes. In this study, we found that miR-421 suppresses kelch-like ECH-associated protein 1(KEAP1) expression by targeting its 3'-untranslated region (3'UTR). A Q-PCR assay demonstrated that miR-421 is overexpressed in non-small cell lung cancer (NSCLC), especially in A549 cells. Consistently, the level of miR-421 was higher in clinical blood samples from lung cancer patients than in those from normal healthy donors, suggesting that miR-421 is an important lung cancer biomarker. Interestingly, overexpression of miR-421 reduced the level of KEAP1 expression, which further promoted lung cancer cell migration and invasion, as well as inhibited cell apoptosis both in vivo and in vitro. Furthermore, knockdown of miR-421 expression with an antisense morpholino oligonucleotide (AMO) increased ROS levels and treatment sensitivity to paclitaxel in vitro and in vivo, indicating that high miR-421 expression may at least partly account for paclitaxel tolerance in lung cancer patients. To find the upstream regulator of miR-421, one of the candidates, β-catenin, was knocked out via the CRISPR/Cas9 method in A549 cells. Our data showed that inhibiting β-catenin reduced miR-421 levels in A549 cells. In addition, β-catenin upregulation enhanced miR-421 expression, indicating that β-catenin regulates the expression of miR-421 in lung cancer. Taken together, our findings reveal the critical role of miR-421 in paclitaxel drug resistance and its upstream and downstream regulatory mechanisms. Therefore, miR-421 may serve as a potential molecular therapeutic target in lung cancer, and AMOs may be a potential treatment strategy.
Collapse
Affiliation(s)
- Fu-Gang Duan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Mei-Fang Wang
- Department of Respiratory and Critical Care, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ya-Bing Cao
- Department of Oncology, Kiang Wu Hospital, Macau, China
| | - Dan Li
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Run-Ze Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Huan-Ling Lai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Wendy Wen-Luan Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Qi-Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China.
| | - Yi-Jun Tang
- Department of Respiratory and Critical Care, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China. .,Department of Respiratory and Critical Care, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China. .,Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China.
| |
Collapse
|
32
|
Lindsay C, Kostiuk M, Conrad D, O’Connell DA, Harris J, Seikaly H, Biron VL. Antitumour effects of metformin and curcumin in human papillomavirus positive and negative head and neck cancer cells. Mol Carcinog 2019; 58:1946-1959. [DOI: 10.1002/mc.23087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Cameron Lindsay
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
- Otolaryngology‐Head and Neck Surgery Research Laboratory of AlbertaUniversity of Alberta Edmonton Alberta Canada
| | - Morris Kostiuk
- Otolaryngology‐Head and Neck Surgery Research Laboratory of AlbertaUniversity of Alberta Edmonton Alberta Canada
| | - Dustin Conrad
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
| | - Daniel A. O’Connell
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
| | - Jeffrey Harris
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
| | - Hadi Seikaly
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
- Otolaryngology‐Head and Neck Surgery Research Laboratory of AlbertaUniversity of Alberta Edmonton Alberta Canada
| | - Vincent L. Biron
- Division of Otolaryngology‐Head and Neck Surgery, Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
- Otolaryngology‐Head and Neck Surgery Research Laboratory of AlbertaUniversity of Alberta Edmonton Alberta Canada
| |
Collapse
|
33
|
Liu JJ, Ho JY, Lee HW, Baik MW, Kim O, Choi YJ, Hur SY. Inhibition of Phosphatidylinositol 3-kinase (PI3K) Signaling Synergistically Potentiates Antitumor Efficacy of Paclitaxel and Overcomes Paclitaxel-Mediated Resistance in Cervical Cancer. Int J Mol Sci 2019; 20:E3383. [PMID: 31295843 PMCID: PMC6679163 DOI: 10.3390/ijms20143383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/30/2019] [Accepted: 07/08/2019] [Indexed: 12/19/2022] Open
Abstract
Acquired paclitaxel (PTX) resistance limits its effectiveness and results in advanced cancer progression. This review investigated whether the inhibition of phosphatidylinositol 3-kinase (PI3K) signaling overcomes paclitaxel resistance in cervical cancer. It was established paclitaxel-resistant cell lines (PTX-R ME180/PTX-R HeLa) and determined the combination index for paclitaxel and PI3K inhibitors (BYL-719/ LY294002) by tetrazolium dye assay. Flow cytometry was used to detect the cell cycle and apoptosis. Migration and invasion were explored by wound healing and transwell assays. Genes related to multiple pathways were assessed by a western blot. It was found that the PI3K pathway was significantly activated in paclitaxel-resistant HeLa and ME180 cells compared to parental cells. PTX + PI3K inhibitor combined therapy showed a synergistic effect by strengthening paclitaxel-induced S and G2M arrest in PTX-R cell sublines by the inactivation of cyclin A1, cyclin B1, cyclin E, and Cdc2 expression. Moreover, combination therapy significantly enhanced drug sensitivity and apoptosis through the activation of Bax, and cleavage of poly-(ADP-ribose) polymerase compared with paclitaxel alone. In addition, PI3K inhibition also suppressed tumor migration and invasion by targeting β-catenin and matrix metalloproteinase-2/9. The authors suggest that the combination of a PI3K inhibitor with paclitaxel may enhance antitumor activity through a cascade of PI3K signaling events.
Collapse
Affiliation(s)
- Jing Jing Liu
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
- Cancer Research Institute, Department of Medical Life Science, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jung Yoon Ho
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
- Cancer Research Institute, Department of Medical Life Science, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Hye Won Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06591, Korea
| | - Min Wha Baik
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Oyoung Kim
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Youn Jin Choi
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea.
- Cancer Research Institute, Department of Medical Life Science, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Soo Young Hur
- Department of Gynecology and Obstetrics, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea.
- Cancer Research Institute, Department of Medical Life Science, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| |
Collapse
|
34
|
Alwadei M, Kazi M, Alanazi FK. Novel oral dosage regimen based on self-nanoemulsifying drug delivery systems for codelivery of phytochemicals - Curcumin and thymoquinone. Saudi Pharm J 2019; 27:866-876. [PMID: 31516329 PMCID: PMC6734017 DOI: 10.1016/j.jsps.2019.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/24/2019] [Indexed: 01/23/2023] Open
Abstract
Background Curcumin and Thymoquinone are very well-known phytochemicals for their potent anti-inflammatory and anticancer properties. The major challenges for curcumin is its poor aqueous solubility and erratic oral bioavailability. Objective To develop a novel liquid self-nanoemulsifying drug delivery system (SNEDDS) containing curcumin and thymoquinone and further converted into a solid dosage form using adsorbents Syloid® and Neusilin® as the solid carrier. Methods The characterization of the liquid and solid SNEDDS was performed by particle size & zeta potential analysis, scanning electron microscopy, differential scanning calorimetry, fourier transform infrared spectroscopy and X-ray powder diffraction. The drug loading, and in vitro release studies were carried out to investigate the efficiency of curcumin release from SNEDDS. Results The liquid SNEDDS containing black seed oil showed excellent self-emulsification performance with transparent appearance. The results of characterization studies showed that solidification using 50% (w/w) Syloid® and Neusilin® in the liquid formulation yield free flowing powder with no agglomeration but Neusilin® produced smooth granules than Syloid® and kept the drugs stable in amorphous state. In vitro dissolution studies indicated that liquid SNEDDS formulations of F4 and its solid SNEDDS using Neusilin® provided high dissolution efficiency and reproducibility for curcumin and thymoquinone. However, Neusilin® showed higher rate of dissolution (more than 65%, p < 0.05) compared to Syloid® for curcumin. Conclusions Curcumin loaded-SNEDDS formulation containing thymoquinone in liquid & solid dosage forms were successfully developed with an increased drug loading and dissolution rate, which could be the potential combined delivery system for various anti-inflammatory and anti-cancer treatments.
Collapse
Affiliation(s)
- Majed Alwadei
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fars Kaed Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
35
|
Hesari A, Rezaei M, Rezaei M, Dashtiahangar M, Fathi M, Rad JG, Momeni F, Avan A, Ghasemi F. Effect of curcumin on glioblastoma cells. J Cell Physiol 2018; 234:10281-10288. [DOI: 10.1002/jcp.27933] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022]
Affiliation(s)
- AmiReza Hesari
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine Arak University of Medical Sciences Arak Iran
| | - Marzieh Rezaei
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Maryam Rezaei
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Maryam Dashtiahangar
- Department of Biology, Faculty of Science Ferdowsi University of Mashhad Mashhad Iran
| | - Mozhgan Fathi
- Medical Genetics Research Center, Department of Medical Genetics, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Jeyran Ganji Rad
- Department of Biology Islamic Azad University of Science Researchs Gorgan Iran
| | - Fatemeh Momeni
- Thalassemia & Hemoglobinopathy Research Center Health Research Institute, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Amir Avan
- Department of Modern Sciences and Technologies School of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Faezeh Ghasemi
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine Arak University of Medical Sciences Arak Iran
- Blood Transfusion Research Center High Institute for Research and Education in Transfusion Medicine Tehran Iran
| |
Collapse
|
36
|
Silva GÁF, Nunes RAL, Morale MG, Boccardo E, Aguayo F, Termini L. Oxidative stress: therapeutic approaches for cervical cancer treatment. Clinics (Sao Paulo) 2018; 73:e548s. [PMID: 30540121 PMCID: PMC6257060 DOI: 10.6061/clinics/2018/e548s] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress results from an imbalance between the generation and elimination of oxidant species. This condition may result in DNA, RNA and protein damage, leading to the accumulation of genetic alterations that can favor malignant transformation. Persistent infection with high-risk human papillomavirus types is associated with inflammatory responses and reactive oxygen species production. In this context, oxidative stress, chronic inflammation and high-risk human papillomavirus can act in a synergistic manner. To counteract the harmful effects of oxidant species, protective molecules, known as antioxidant defenses, are produced by cells to maintain redox homeostasis. In recent years, the use of natural antioxidants as therapeutic strategies for cancer treatment has attracted the attention of the scientific community. This review discusses specific molecules and mechanisms that can act against or together with oxidative stress, presenting alternatives for cervical cancer prevention and treatment.
Collapse
Affiliation(s)
- Gabriela Ávila Fernandes Silva
- Instituto do Cancer do Estado de Sao Paulo ICESP, Centro de Investigacao Translacional em Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Radiologia e Oncologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, BR
| | - Rafaella Almeida Lima Nunes
- Instituto do Cancer do Estado de Sao Paulo ICESP, Centro de Investigacao Translacional em Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Radiologia e Oncologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, BR
| | - Mirian Galliote Morale
- Instituto do Cancer do Estado de Sao Paulo ICESP, Centro de Investigacao Translacional em Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Departamento de Radiologia e Oncologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, BR
| | - Enrique Boccardo
- Laboratorio de Oncovirologia, Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Francisco Aguayo
- Centro Avanzado de Enfermedades Cronicas (ACCDiS), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Oncologia Basico Clinica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lara Termini
- Instituto do Cancer do Estado de Sao Paulo ICESP, Centro de Investigacao Translacional em Oncologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| |
Collapse
|
37
|
Zhang C, Hao Y, Wu L, Dong X, Jiang N, Cong B, Liu J, Zhang W, Tang D, De Perrot M, Zhao X. Curcumin induces apoptosis and inhibits angiogenesis in murine malignant mesothelioma. Int J Oncol 2018; 53:2531-2541. [PMID: 30272283 PMCID: PMC6203149 DOI: 10.3892/ijo.2018.4569] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/21/2018] [Indexed: 01/08/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare form of cancer that is associated with asbestos exposure. Unfortunately, current therapies have limited efficacy. Previous studies have indicated that curcumin exerts antiproliferative and antitumor effects, and has low toxicity. The present study aimed to evaluate the anticancer effects of curcumin on the RN5 MPM cell line. The inhibitory effects of curcumin on cell viability were determined using the sulforhodamine B assay. In addition, cell cycle progression was analyzed by propidium iodide (PI) staining and flow cytometry, and curcumin‑induced apoptosis was measured by Annexin V/PI double staining. The translocation of apoptosis-inducing factor (AIF) was assessed by western blotting and immunofluorescence, and the expression levels of the phosphoinositide 3-kinase (PI3K)-AKT serine/threonine kinase (Akt)‑mammalian target of rapamycin (mTOR) signaling pathway proteins and mitochondria-associated proteins were evaluated by western blotting. In vivo antitumor effects were evaluated in a subcutaneous murine model. Briefly, tumors were harvested from the mice, and immunohistochemistry was conducted to evaluate cell proliferation, apoptosis and angiogenesis. The results indicated that curcumin inhibited RN5 cell viability and induced apoptotic cell death. In addition the findings suggested that curcumin-induced cell apoptosis occurred via the mitochondrial pathway, and caspase‑independent and AIF-dependent pathways. Further analysis revealed that curcumin may act as a PI3K-Akt-mTOR signaling pathway inhibitor by downregulating PI3K, p-Akt, p-mTOR and p-p70 ribosomal protein S6 kinase. Furthermore, curcumin inhibited tumor angiogenesis in vivo. In conclusion, curcumin may be potent enough to be developed as a novel therapeutic agent for the treatment of MPM.
Collapse
Affiliation(s)
- Chengke Zhang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yingtao Hao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Licun Wu
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiaopeng Dong
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ning Jiang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Bo Cong
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Jiang Liu
- Gene and Immunotherapy Center, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Wen Zhang
- Gene and Immunotherapy Center, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Dongqi Tang
- Gene and Immunotherapy Center, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Marc De Perrot
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
38
|
de Oliveira Júnior RG, Christiane Adrielly AF, da Silva Almeida JRG, Grougnet R, Thiéry V, Picot L. Sensitization of tumor cells to chemotherapy by natural products: A systematic review of preclinical data and molecular mechanisms. Fitoterapia 2018; 129:383-400. [DOI: 10.1016/j.fitote.2018.02.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022]
|
39
|
Marjaneh RM, Rahmani F, Hassanian SM, Rezaei N, Hashemzehi M, Bahrami A, Ariakia F, Fiuji H, Sahebkar A, Avan A, Khazaei M. Phytosomal curcumin inhibits tumor growth in colitis‐associated colorectal cancer. J Cell Physiol 2018; 233:6785-6798. [DOI: 10.1002/jcp.26538] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 02/06/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Reyhaneh Moradi Marjaneh
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Metabolic syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Nastaran Rezaei
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Milad Hashemzehi
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center Birjand University of Medical Sciences Birjnad Iran
| | - Fatemeh Ariakia
- Medical Toxicology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Hamid Fiuji
- Metabolic syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhosein Sahebkar
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Amir Avan
- Metabolic syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Cancer Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Metabolic syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
40
|
Curcumin in Advancing Treatment for Gynecological Cancers with Developed Drug- and Radiotherapy-Associated Resistance. Rev Physiol Biochem Pharmacol 2018; 176:107-129. [DOI: 10.1007/112_2018_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
41
|
Huang CY, Ju DT, Chang CF, Muralidhar Reddy P, Velmurugan BK. A review on the effects of current chemotherapy drugs and natural agents in treating non-small cell lung cancer. Biomedicine (Taipei) 2017; 7:23. [PMID: 29130448 PMCID: PMC5682982 DOI: 10.1051/bmdcn/2017070423] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, and this makes it an attractive disease to review and possibly improve therapeutic treatment options. Surgery, radiation, chemotherapy, targeted treatments, and immunotherapy separate or in combination are commonly used to treat lung cancer. However, these treatment types may cause different side effects, and chemotherapy-based regimens appear to have reached a therapeutic plateau. Hence, effective, better-tolerated treatments are needed to address and hopefully overcome this conundrum. Recent advances have enabled biologists to better investigate the potential use of natural compounds for the treatment or control of various cancerous diseases. For the past 30 years, natural compounds have been the pillar of chemotherapy. However, only a few compounds have been tested in cancerous patients and only partial evidence is available regarding their clinical effectiveness. Herein, we review the research on using current chemotherapy drugs and natural compounds (Wortmannin and Roscovitine, Cordyceps militaris, Resveratrol, OSU03013, Myricetin, Berberine, Antroquinonol) and the beneficial effects they have on various types of cancers including non-small cell lung cancer. Based on this literature review, we propose the use of these compounds along with chemotherapy drugs in patients with advanced and/or refractory solid tumours.
Collapse
Affiliation(s)
- Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan - Graduate Institute of Chinese Medical Science, China Medical University, Taichung 404, Taiwan - Department of Biological Science and Technology, Asia University, Taichung 413, Taiwan
| | - Da-Tong Ju
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Fen Chang
- Department of Internal Medicine, Division of Cardiology, Armed Forces Taichung General Hospital, Taichung 406, Taiwan
| | - P Muralidhar Reddy
- Department of Chemistry, Nizam College, Osmania University, Hyderabad-500001, India
| | - Bharath Kumar Velmurugan
- Faculty of Applied Sciences, Ton Duc Thang University, Tan Phong Ward, District 7, 700000 Ho Chi Minh City, Vietnam
| |
Collapse
|
42
|
Thulasidasan AKT, Retnakumari AP, Shankar M, Vijayakurup V, Anwar S, Thankachan S, Pillai KS, Pillai JJ, Nandan CD, Alex VV, Chirayil TJ, Sundaram S, Kumar GSV, Anto RJ. Folic acid conjugation improves the bioavailability and chemosensitizing efficacy of curcumin-encapsulated PLGA-PEG nanoparticles towards paclitaxel chemotherapy. Oncotarget 2017; 8:107374-107389. [PMID: 29296172 PMCID: PMC5746074 DOI: 10.18632/oncotarget.22376] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
Nanoencapsulation has emerged as a novel strategy to enhance the pharmacokinetic and therapeutic potential of conventional drugs. Recent studies from our lab have established the efficacy of curcumin in sensitizing cervical cancer cells and breast cancer cells towards paclitaxel and 5-FU chemotherapy respectively. Factors that hinder the clinical use of curcumin as a sensitizer or therapeutic agent include its poor bioavailability and retention time. Earlier reports of improvement in bioavailability and retention of drugs upon nanoencapsulation have motivated us in developing various nanoformulations of curcumin, which were found to exhibit significant enhancement in bioavailability and retention time as assessed by our previous in vitro studies. Among the various formulations tested, curcumin-entrapped in PLGA-PEG nanoparticles conjugated to folic acid (PPF-curcumin) displayed maximum cell death. In the present study, we have demonstrated the efficacy of this formulation in augmenting the bioavailability and retention time of curcumin, in vivo, in Swiss albino mice. Further, the acute and chronic toxicity studies proved that the formulation is pharmacologically safe. We have also evaluated its potential in chemosensitizing cervical cancer cells to paclitaxel and have verified the results using cervical cancer xenograft model in NOD-SCID mice. Folic acid conjugation significantly enhanced the efficacy of curcumin in down-regulating various survival signals induced by paclitaxel in cervical cancer cells and have considerably improved its potential in inhibiting the tumor growth of cervical cancer xenografts. The non-toxic nature coupled with improved chemosensitization potential makes PPF-curcumin a promising candidate formulation for clinical trials.
Collapse
Affiliation(s)
- Arun Kumar T Thulasidasan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Archana P Retnakumari
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Mohan Shankar
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, Manipal University, Manipal, Karnataka, India
| | - Vinod Vijayakurup
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Shabna Anwar
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Sanu Thankachan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Kavya S Pillai
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Jisha J Pillai
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - C Devika Nandan
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Vijai V Alex
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Teena Jacob Chirayil
- Division of Chemical Biology-Nano Drug Delivery Systems, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Research Scholar, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Kottayam, Kerala, India
| | | | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
43
|
Heat Shock Protein-Inducing Property of Diarylheptanoid Containing Chalcone Moiety from Alpinia katsumadai. Molecules 2017; 22:molecules22101750. [PMID: 29039794 PMCID: PMC6151646 DOI: 10.3390/molecules22101750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/14/2017] [Accepted: 10/16/2017] [Indexed: 01/10/2023] Open
Abstract
A new diarylheptanoid containing a chalcone moiety, katsumain H (1), was isolated from the seeds of Alpinia katsumadai. The structure was elucidated using a combination of 1D/2D NMR spectroscopy and mass spectrometry data analysis. The absolute configurations of C-3, C-5, and C-7 in 1 were assigned based on its optical rotation and after comparing its NMR chemical shifts with those of its diastereoisomers, katsumain E and katsumain F, which were previously isolated from this plant and characterized. In this study, the stimulatory effects of compounds 1 and 2 were evaluated on heat shock factor 1 (HSF1), heat shock protein 27 (HSP27), and HSP70. Compounds 1 and 2 increased the expression of HSF1 (1.056- and 1.200-fold, respectively), HSP27 (1.312- and 1.242-fold, respectively), and HSP70 (1.234- and 1.271-fold, respectively), without increased cytotoxicity.
Collapse
|
44
|
Parveen I, Ahmed N, Idrees D, Khan P, Hassan MI. Synthesis, estrogen receptor binding affinity and molecular docking of pyrimidine-piperazine-chromene and -quinoline conjugates. Bioorg Med Chem Lett 2017; 27:4493-4499. [DOI: 10.1016/j.bmcl.2017.07.077] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/22/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
|
45
|
Huang S, Wang D, Zhang S, Huang X, Wang D, Ijaz M, Shi Y. Tunicamycin potentiates paclitaxel-induced apoptosis through inhibition of PI3K/AKT and MAPK pathways in breast cancer. Cancer Chemother Pharmacol 2017; 80:685-696. [DOI: 10.1007/s00280-017-3393-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
|
46
|
Yao Q, Gutierrez DC, Hoang NH, Kim D, Wang R, Hobbs C, Zhu L. Efficient Codelivery of Paclitaxel and Curcumin by Novel Bottlebrush Copolymer-based Micelles. Mol Pharm 2017; 14:2378-2389. [PMID: 28605595 DOI: 10.1021/acs.molpharmaceut.7b00278] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The novel self-assembling bottlebrush polyethylene glycol-polynorbornene-thiocresol block copolymers (PEG-PNB-TC) were synthesized by the ring opening metathesis polymerization (ROMP), followed by functionalization of the polymer backbone via the thio-bromo "click" postpolymerization strategy. The PEG-PNB-TC copolymers could easily self-assemble into the nanoscale core-shell polymeric micelles. The hydrophobic anticancer drugs, such as paclitaxel (PTX), could be loaded into their hydrophobic core to form a stable drug-loaded micelle with a superior drug loading capacity of up to ∼35% (w/w). The sustained drug release behavior of the PEG-PNB-TC micelles was observed under a simulated "sink condition". Compared with commercial PTX formulation (Taxol), the PTX-loaded PEG-PNB-TC micelles showed the enhanced in vitro cellular uptake and comparable cytotoxicity in the drug-sensitive cancer cells, while the copolymers were much safer than Cremophor EL, the surfactant used in Taxol. Furthermore, curcumin (CUR), a natural chemotherapy drug sensitizer, was successfully coloaded with PTX into the PEG-PNB-TC micelles. High drug loading capacity of the PEG-PNB-TC micelles allowed for easy adjustment of drug doses and the ratio of the coloaded drugs. The combination of PTX and CUR showed synergistic anticancer effect in both the drug mixture and drug coloaded micelles at high CUR/PTX ratio, while low CRU/PTX ratio only exhibited additive effects. The combinatorial effects remarkably circumvented the PTX resistance in the multidrug resistant (MDR) cancer cells. Due to the easy polymerization and functionalization, excellent self-assembly capability, high drug loading capability, and great stability, the PEG-PNB-TC copolymers might be a promising nanomaterial for delivery of the hydrophobic anticancer drugs, especially for combination drug therapy.
Collapse
Affiliation(s)
- Qing Yao
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States.,Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, 110016 Liaoning, People's Republic of China
| | - David C Gutierrez
- Department of Chemistry, Texas A&M University-Kingsville , Kingsville, Texas 78363, United States
| | - Ngoc Ha Hoang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States.,Nanobiopharmaceutics laboratory, College of Pharmacy, Chung-Ang University , Seoul, South Korea.,Department of Pharmaceutics, Hanoi University of Pharmacy , Ha Noi, Vietnam
| | - Dongin Kim
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, Ohio State University , Columbus, Ohio 43025-2696, United States
| | - Christopher Hobbs
- Department of Chemistry, Texas A&M University-Kingsville , Kingsville, Texas 78363, United States
| | - Lin Zhu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States
| |
Collapse
|
47
|
Wei Y, Pu X, Zhao L. Preclinical studies for the combination of paclitaxel and curcumin in cancer therapy (Review). Oncol Rep 2017; 37:3159-3166. [PMID: 28440434 DOI: 10.3892/or.2017.5593] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 04/10/2017] [Indexed: 11/05/2022] Open
Abstract
Cancer is one of the most common causes of death and remains the first in China and the second in the US. The common treatments for cancer include surgery, radiation, chemotherapy, targeted therapy and immunotherapy, while chemotherapy remains one of the most important treatments. However, the efficacy of chemotherapy is limited due to drug induced-toxicities and resistance, particularly multiple drug resistance (MDR). Therefore, discovery and development of novel therapeutic drugs and/or combination therapy are urgently needed to reduce toxicity and improve efficacy. Paclitaxel has been widely used to treat various cancers including cervical, breast, ovarian, brain, bladder, prostate, liver and lung cancers. However, its therapeutic efficacy is limited and MDR is a major obstacle. Recently, numerous preclinical studies have shown that the combination of paclitaxel and curcumin may be an ideal strategy to reverse MDR and synergistically improve their therapeutic efficacy in cancer therapy. This review mainly focuses on the current development and progress of the combination of paclitaxel and curcumin in cancer therapy preclinically.
Collapse
Affiliation(s)
- Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646099, P.R. China
| | - Xinlin Pu
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646099, P.R. China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646099, P.R. China
| |
Collapse
|
48
|
Rikkunshito prevents paclitaxel-induced peripheral neuropathy through the suppression of the nuclear factor kappa B (NFκB) phosphorylation in spinal cord of mice. PLoS One 2017; 12:e0171819. [PMID: 28182729 PMCID: PMC5300261 DOI: 10.1371/journal.pone.0171819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/26/2017] [Indexed: 12/15/2022] Open
Abstract
Peripheral neuropathy is the major side effect caused by paclitaxel, a microtubule-binding antineoplastic drug. Paclitaxel-induced peripheral neuropathy causes a long-term negative impact on the patient's quality of life. However, the mechanism underlying paclitaxel-induced peripheral neuropathy is still unknown, and there is no established treatment. Ghrelin is known to attenuate thermal hyperalgesia and mechanical allodynia in chronic constriction injury of the sciatic nerve, and inhibit the activation of nuclear factor kappa B (NFκB) in the spinal dorsal horn. Rikkunshito (RKT), a kampo medicine, increases the secretion of ghrelin in rodents and humans. Thus, RKT may attenuate paclitaxel-induced peripheral neuropathy by inhibiting phosphorylated NFκB (pNFκB) in the spinal cord. We found that paclitaxel dose-dependently induced mechanical hyperalgesia in mice. Paclitaxel increased the protein levels of spinal pNFκB, but not those of spinal NFκB. NFκB inhibitor attenuated paclitaxel-induced mechanical hyperalgesia suggesting that the activation of NFκB mediates paclitaxel-induced hyperalgesia. RKT dose-dependently attenuated paclitaxel-induced mechanical hyperalgesia. Ghrelin receptor antagonist reversed the RKT-induced attenuation of paclitaxel-induced mechanical hyperalgesia. RKT inhibited the paclitaxel-induced increase in the protein levels of spinal pNFκB. Taken together, the present study indicates that RKT exerts an antihyperalgesic effect in paclitaxel-induced neuropathic pain by suppressing the activation of spinal NFκB.
Collapse
|
49
|
Jameel E, Naz H, Khan P, Tarique M, Kumar J, Mumtazuddin S, Ahamad S, Islam A, Ahmad F, Hoda N, Hassan MI. Design, synthesis, and biological evaluation of pyrimidine derivatives as potential inhibitors of human calcium/calmodulin-dependent protein kinase IV. Chem Biol Drug Des 2016; 89:741-754. [PMID: 27809417 DOI: 10.1111/cbdd.12898] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/17/2016] [Accepted: 10/22/2016] [Indexed: 01/18/2023]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CAMKIV) is a multifunctional Ser/Thr kinase, associated with cerebral hypoxia, cancer, and neurodegenerative diseases. Here, we report design, synthesis, and biological evaluation of seven pyrimidine-substituted novel inhibitors of CAMKIV. We successfully synthesized and extensively characterized (ESI-MS, 1 H NMR, and 13 C NMR studies) seven compounds that are showing appreciable binding affinity to the CAMKIV. Molecular docking and fluorescence binding studies revealed that compound 1 is showing very high binding free energy (ΔG = -11.52 kcal/mol) and binding affinity (K = 9.2 × 1010 m-1 ) to the CAMKIV. We further performed MTT assay to check the cytotoxicity and anticancer activity of these compounds. An appreciable IC50 (39 μm) value of compound 1 was observed on human hepatoma cell line and nontoxic till the 400 μm on human embryonic kidney cells. To ensure anticancer activity of all these compounds, we further performed propidium iodide assay to evaluate cell viability and DNA content during the cell cycle. We found that compound 1 is again showing a better anticancer activity on both human hepatoma and human embryonic kidney cell lines.
Collapse
Affiliation(s)
- Ehtesham Jameel
- Department of Chemistry, B.R. Ambedkar Bihar University, Muzaffarpur, Bihar, India
| | - Huma Naz
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Parvez Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd Tarique
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Jitendra Kumar
- Department of Chemistry, Jamia Millia Islamia, New Delhi, India
| | - Syed Mumtazuddin
- Department of Chemistry, B.R. Ambedkar Bihar University, Muzaffarpur, Bihar, India
| | - Shahzaib Ahamad
- Department of Biotechnology, College of Engineering and Technology, IFTM, Moradabad, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Faizan Ahmad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Nasimul Hoda
- Department of Chemistry, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
50
|
Nath LR, Gorantla JN, Thulasidasan AKT, Vijayakurup V, Shah S, Anwer S, Joseph SM, Antony J, Veena KS, Sundaram S, Marelli UK, Lankalapalli RS, Anto RJ. Evaluation of uttroside B, a saponin from Solanum nigrum Linn, as a promising chemotherapeutic agent against hepatocellular carcinoma. Sci Rep 2016; 6:36318. [PMID: 27808117 PMCID: PMC5093766 DOI: 10.1038/srep36318] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/11/2016] [Indexed: 02/08/2023] Open
Abstract
We report, for the first time, the remarkable efficacy of uttroside B, a potent saponin from Solanum nigrum Linn, against liver cancer. The compound has been isolated and characterized from the leaves of Solanum nigrum Linn, a plant widely used in traditional medicine and is a rich resource of several anticancer molecules. Uttroside B, that comprises of β-D-glucopyranosyl unit at C-26 of the furostanol and β-lycotetraosyl unit at C-3, is ten times more cytotoxic to the liver cancer cell line, HepG2 (IC50: 0.5 μM) than sorafenib (IC50: 5.8 μM), the only FDA-approved drug for liver cancer. Moreover, it induces cytotoxicity in all liver cancer cell lines, irrespective of their HBV status, while being non-toxic to normal immortalized hepatocytes. It induces apoptosis in HepG2 cells by down-regulating mainly the activation of MAPK and mTOR pathways. The drastic reduction in HepG2-xenograft tumor size achieved by uttroside B in NOD-SCID mice and substantiation of its biological safety through both acute and chronic toxicity studies in Swiss albino mice warrants clinical validation of the molecule against hepatic cancer, for which, the chemotherapeutic armamentarium currently has limited weapons.
Collapse
Affiliation(s)
- Lekshmi R. Nath
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Jaggaiah N. Gorantla
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram-695019, Kerala, India
| | - Arun Kumar T. Thulasidasan
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Vinod Vijayakurup
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Shabna Shah
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Shabna Anwer
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Sophia M. Joseph
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Jayesh Antony
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| | - Kollery Suresh Veena
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram-695019, Kerala, India
| | - Sankar Sundaram
- Department of Pathology, Government Medical College, Thiruvananthapuram-695011, Kerala, India
| | - Udaya K. Marelli
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune-411008, India
| | - Ravi S. Lankalapalli
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram-695019, Kerala, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram-695014, Kerala, India
| |
Collapse
|