1
|
Wahnou H, El Kebbaj R, Liagre B, Sol V, Limami Y, Duval RE. Curcumin-Based Nanoparticles: Advancements and Challenges in Tumor Therapy. Pharmaceutics 2025; 17:114. [PMID: 39861761 PMCID: PMC11768525 DOI: 10.3390/pharmaceutics17010114] [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: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Curcumin, a bioactive compound derived from the rhizome of Curcuma longa L., has garnered significant attention for its potent anticancer properties. Despite its promising therapeutic potential, its poor bioavailability, rapid metabolism, and low water solubility hinder curcumin's clinical application. Nanotechnology offers a viable solution to these challenges by enabling the development of curcumin-based nanoparticles (CNPs) that enhance its bioavailability and therapeutic efficacy. This review provides a comprehensive overview of the recent advancements in the design and synthesis of CNPs for cancer therapy. We discuss various NP formulations, including polymeric, lipid-based, and inorganic nanoparticles, highlighting their role in improving curcumin's pharmacokinetic and pharmacodynamic profiles. The mechanisms by which CNPs exert anticancer effects, such as inducing apoptosis, inhibiting cell proliferation, and modulating signaling pathways, are explored in details. Furthermore, we examine the preclinical and clinical studies that have demonstrated the efficacy of CNPs in treating different types of tumors, including breast, colorectal, and pancreatic cancers. Finally, the review addresses the current challenges and future perspectives in the clinical translation of CNPs, emphasizing the need for further research to optimize their design for targeted delivery and to enhance their therapeutic outcomes. By synthesizing the latest research, this review underscores the potential of CNPs as a promising avenue for advancing cancer therapy.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P2693, Maarif, Casablanca 20100, Morocco;
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Vincent Sol
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Youness Limami
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| | | |
Collapse
|
2
|
Wang G, Pan L, Guo R. Restoration of miR-200 expression suppresses proliferation and mobility of pancreatic cancer cell. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03717-0. [PMID: 39754677 DOI: 10.1007/s00210-024-03717-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/08/2024] [Indexed: 01/06/2025]
Abstract
A number of various human malignancies have been associated with abnormal microRNAs (miRNA) expression. There are evidence that miR-200 operates as both tumor suppressor and an onco-miR in a variety of tumors. In this study, we evaluated the effects of miR-200 on the proliferation and migration of pancreatic cancer cells, as well as the underlying molecular pathways. Clinical tissue samples were used to investigate the expression of miR-200 in pancreatic cancer and normal tissues, and the gene expression omnibus (GEO) database provided bioinformatics confirmation. Using the pCMV vector, miR-200 was transfected into PANC-1 pancreatic cancer cells. After transfection, expression of cancer-related genes (at the mRNA and protein levels) was evaluated. The miR-200-transfected pancreatic cancer cells' survival, invasion, migration, and apoptosis were also investigated. According to the bioinformatics analysis, decreased miR-200 expression was associated with a worse prognosis in pancreatic cancer patients. Moreover, low levels of miR-200 in pancreatic cancer tissues were approved. After transfection, pancreatic cancer cells exhibit a sustained increase in expression of miR-200, which inhibits cell viability, invasion, and migration. Additional investigations revealed that increasing expression of miR-200 increases the proportion of pancreatic cancer cells that endure apoptosis. Changes in the mRNA and protein expression of apoptosis- and metastasis-related genes may account for these findings. Our results indicate that miR-200 functions as a tumor suppressor in pancreatic cancer cells and that upregulating miR-200 levels may be a useful therapeutic strategy for pancreatic cancer patients to halt the progression of the illness.
Collapse
Affiliation(s)
- Guiming Wang
- Department of General Surgery, NHC Key Laboratory of Hormones and Development and Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianji, 300134, China
| | - Lifeng Pan
- Department of General Surgery, NHC Key Laboratory of Hormones and Development and Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianji, 300134, China
| | - Rende Guo
- Department of General Surgery, Tianjin First Center Hospital, Tianji, 300384, China.
| |
Collapse
|
3
|
Keshavarz Shahbaz S, Koushki K, Izadi O, Penson PE, Sukhorukov VN, Kesharwani P, Sahebkar A. Advancements in curcumin-loaded PLGA nanoparticle delivery systems: progressive strategies in cancer therapy. J Drug Target 2024; 32:1207-1232. [PMID: 39106154 DOI: 10.1080/1061186x.2024.2389892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Cancer is a leading cause of death worldwide, and imposes a substantial socioeconomic burden with little impact especially on aggressive types of cancer. Conventional therapies have many serious side effects including generalised systemic toxicity which limits their long-term use. Tumour resistance and recurrence is another main problem associated with conventional therapy. Purified or extracted natural products have been investigated as cost-effective cancer chemoprotective agents with the potential to reverse or delaying carcinogenesis. Curcumin (CUR) as a natural polyphenolic component, exhibits many pharmacological activities such as anti-cancer, anti-inflammatory, anti-microbial, activity against neurodegenerative diseases including Alzheimer, antidiabetic activities (type II diabetes), anticoagulant properties, wound healing effects in both preclinical and clinical studies. Despite these effective protective properties, CUR has several limitations, including poor aqueous solubility, low bioavailability, chemical instability, rapid metabolism and a short half-life time. To overcome the pharmaceutical problems associated with free CUR, novel nanomedicine strategies (including polymeric nanoparticles (NPs) such as poly (lactic-co-glycolic acid) (PLGA) NPs have been developed. These formulations have the potential to improve the therapeutic efficacy of curcuminoids. In this review, we comprehensively summarise and discuss recent in vitro and in vivo studies to explore the pharmaceutical significance and clinical benefits of PLGA-NPs delivery system to improve the efficacy of CUR in the treatment of cancer.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Omid Izadi
- Department of Industrial Engineering, ACECR Institute of Higher Education Kermanshah, Kermanshah, Iran
| | - Peter E Penson
- Clinical Pharmacy and Therapeutics Research Group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Farhadi A, Poursheikhani A, Heidari MF, Rajaeinejad M, Mosaed R, Faridfar A, Khosroshahi MA, Aslani P, Khalil-Moghaddam S, Behroozi J. Effects of miR-330 restoration on pancreatic cancer cells oncogenesis. Pathol Res Pract 2024; 258:155337. [PMID: 38735276 DOI: 10.1016/j.prp.2024.155337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Inappropriate expressions of various miRNAs have reported in different human malignancies. Evidence suggested that miR-330 may play as both onco-miR and/or tumor suppressor-miR in different cancers. In the present study, we evaluated effects of miR-330 on proliferation and migration of pancreatic cancer (PC) cells as well as underlying molecular mechanisms. DESIGN The expression of miR-330 was evaluated in clinical tissue samples of patients with PC. Transfection of the PC cells (PANC-1) by miR-330 was conducted by pCMV vector. The cancer-related genes expression was investigated in mRNA and protein level following transfection of the PC cells. Furthermore, the PC cells viability, invasion, migration, mitochondrial membrane potential, apoptosis, autophagy, and cell cycle profile were investigated after transfection by miR-330. RESULTS The results indicated that expression of miR-330 downregulated in patients with PC. Stable increase of miR-330 expression after transfection in PC cells reduces viability, mitochondrial membrane potential, invasion, and migration. Further assessments demonstrated that upregulation of miR-330 increases apoptosis and autophagy percentage in the PC cells. Moreover, a cell cycle arrest was observed in G1, Sub-G1, and S phases following transfection of the PC cells. These findings can be explained by modified mRNA and protein expression of apoptosis- and metastasis-related genes. CONCLUSION Our study suggested that miR-330 acts as a tumor suppressor in PC cells, and revealed that upregulation of miR-330 may provide an effective therapeutic approach for overcoming progression and metastasis in patients with PC.
Collapse
Affiliation(s)
- Arezoo Farhadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Arash Poursheikhani
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Foad Heidari
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran; Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Rajaeinejad
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Peyman Aslani
- Department of Parasitology and Mycology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Javad Behroozi
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
5
|
Gervasi F, Pojero F. Use of Oleuropein and Hydroxytyrosol for Cancer Prevention and Treatment: Considerations about How Bioavailability and Metabolism Impact Their Adoption in Clinical Routine. Biomedicines 2024; 12:502. [PMID: 38540115 PMCID: PMC10968586 DOI: 10.3390/biomedicines12030502] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/12/2024] [Accepted: 02/18/2024] [Indexed: 11/11/2024] Open
Abstract
The fact that the Mediterranean diet could represent a source of natural compounds with cancer-preventive and therapeutic activity has been the object of great interest, especially with regard to the mechanisms of action of polyphenols found in olive oil and olive leaves. Secoiridoid oleuropein (OLE) and its derivative hydroxytyrosol (3,4-dihydroxyphenylethanol, HT) have demonstrated anti-proliferative properties against a variety of tumors and hematological malignancies both in vivo and in vitro, with measurable effects on cellular redox status, metabolism, and transcriptional activity. With this review, we aim to summarize the most up-to-date information on the potential use of OLE and HT for cancer treatment, making important considerations about OLE and HT bioavailability, OLE- and HT-mediated effects on drug metabolism, and OLE and HT dual activity as both pro- and antioxidants, likely hampering their use in clinical routine. Also, we focus on the details available on the effects of nutritionally relevant concentrations of OLE and HT on cell viability, redox homeostasis, and inflammation in order to evaluate if both compounds could be considered cancer-preventive agents or new potential chemotherapy drugs whenever their only source is represented by diet.
Collapse
Affiliation(s)
- Francesco Gervasi
- Specialistic Oncology Laboratory Unit, ARNAS Hospitals Civico Di Cristina e Benfratelli, 90127 Palermo, Italy;
| | - Fanny Pojero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| |
Collapse
|
6
|
Aghaei E, Soltanzadeh H, Kohan L, Heiat M. Apatinib increases anticancer potential of doxorubicin in breast cancer cells. Mol Biol Rep 2023; 50:10137-10145. [PMID: 37921980 DOI: 10.1007/s11033-023-08860-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 11/05/2023]
Abstract
BACKGROUND In recent years, drug resistance has become a most important challenge in chemotherapy of malignancies. Here, we investigated a novel approach to enhance therapeutic potential of doxorubicin (Dox as a common chemotherapeutic drug) by co-administration of apatinib (Apa as a monoclonal antibody) in breast cancer treatment. METHODS AND RESULTS Effects of Apa, Dox, and their combinations (Apa-Dox) were investigated on proliferation of MDA-MB-231 breast cancer cells by MTT assay. Moreover, migration and invasion of the treated and untreated control cancer cells were evaluated by scratch and transwell methods, respectively. Apoptosis percentage of the treated cancer cells was investigated by flow cytometry method. Finally, apoptosis-, metastasis-, and angiogenesis-related gene expression at mRNA and protein levels in the cancer cells were investigated by Real-Time PCR and western blotting methods, respectively. Our results indicated that treatments of cancer cells by Apa, Dox, and Apa-Dox significantly decrease proliferation, migration, and invasion of MDA-MB-231 breast cancer cells. Treatments of the breast cancer cells by Apa, Dox, and Apa-Dox significantly increase apoptosis percentage. We observed that anticancer effects of Apa, Dox, and Apa-Dox may due to modification of apoptosis-, metastasis-, and angiogenesis-related gene expression (at mRNA and protein level) in the breast cancer cells. However, anticancer potential of Apa-Dox combination was significantly more than Apa and Dox monotherapy. CONCLUSION We demonstrated that Apa significantly increases anticancer potential of Dox in MDA-MB-231 breast cells. However, further in-vitro, in-vivo, and clinical studies are required to confirm this result.
Collapse
Affiliation(s)
- Elnaz Aghaei
- Department of Biology, Faculty of Science, Arsanjan Branch, Islamic Azad University, Arsanjan, Iran
| | - Hossein Soltanzadeh
- Department of Genetics, Bonab Branch, Islamic Azad University, Bonab, Iran.
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Leila Kohan
- Department of Biology, Faculty of Science, Arsanjan Branch, Islamic Azad University, Arsanjan, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Disease (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Safavi R, Soltanzadeh H, Hojjati Bonab Z. Scrophularia amplexicaulis increases anti-cancer potential of doxorubicin in gastric cancer cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:2741-2750. [PMID: 37471627 DOI: 10.1002/tox.23909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/17/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Increased drug resistance has reduced efficiency of chemotherapic drugs such as Doxorubicin (Dox). Scrophularia amplexicaulis (Scr) is one of the most important medicinal plants in Iran that has anti-cancer activity. The aim of this study was to investigate a novel approach to enhance therapeutic efficacy of Dox (as a chemotherapeutic agent) by co-administration of Scr (as a bioactive herbal compound) in gastric cancer treatment. In the present study, effects of Dox, Scr, and their combinations (Scr-Dox) were evaluated on viability and proliferation of two gastric cancer cell lines (AGS and MKN28). Moreover, morphological changes, invasion, migration, colony formation, and apoptosis rate in the treated cancer cells were evaluated. Expression of BAX, BCL2, SAMC, SURVIVIN, CASP9, P53, MMP9, and MMP2 in the treated cancer cells and untreated controls were evaluated by Real-Time PCR method. Treatments of cancer cells by Scr, Dox, and Scr-Dox significantly decreased proliferation, invasion, migration, and colony formation of gastric cancer cells. Treatments of cancer cells by Scr, Dox, and Scr-Dox significantly increased apoptosis rate as well as decreased cells mobility through modification of apoptosis- and metastasis-related genes expression. However, anti-cancer activity of Scr-Dox combination was significantly more than Scr and Dox treatments alone. In general, we demonstrated that Scr-Dox combination therapy exerts more profound anti-cancer effects on AGS and MKN28 cell lines than Scr and Dox monotherapy.
Collapse
Affiliation(s)
- Reza Safavi
- Department of Genetics, Bonab Branch, Islamic Azad University, Bonab, Iran
| | - Hossein Soltanzadeh
- Department of Genetics, Bonab Branch, Islamic Azad University, Bonab, Iran
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zahra Hojjati Bonab
- Department of Microbiology, Bonab Branch, Islamic Azad University, Bonab, Iran
| |
Collapse
|
8
|
Isazadeh H, Oruji F, Shabani S, Behroozi J, Nasiri H, Isazadeh A, Akbari M. Advances in siRNA delivery approaches in cancer therapy: challenges and opportunities. Mol Biol Rep 2023; 50:9529-9543. [PMID: 37741808 DOI: 10.1007/s11033-023-08749-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 09/25/2023]
Abstract
Advancements in the clinical applications of small interfering RNA (siRNA) in cancer therapy have opened up new possibilities for precision medicine. siRNAs, as powerful genetic tools, have shown potential in targeting and suppressing the expression of specific genes associated with cancer progression. Their effectiveness has been further enhanced by incorporating them into nanoparticles, which protect siRNAs from degradation and enable targeted delivery. However, despite these promising developments, several challenges persist in the clinical translation of siRNA-based cancer therapy. This comprehensive review explores the progress and challenges associated with the clinical applications of siRNA in cancer therapy. This review highlights the use of siRNA-loaded nanoparticles as an effective delivery system for optimizing siRNA efficacy in various types of carcinomas and the potential of siRNA-based therapy as a genetic approach to overcome limitations associated with conventional chemotherapeutic agents, including severe drug toxicities and organ damage. Moreover, it emphasizes on the key challenges, including off-target effects, enzymatic degradation of siRNAs in serum, low tumor localization, stability issues, and rapid clearance from circulation that need to be addressed for successful clinical development of siRNA-based cancer therapy. Despite these challenges, the review identifies significant avenues for advancing siRNA technology from the laboratory to clinical settings. The ongoing progress in siRNA-loaded nanoparticles for cancer treatment demonstrates potential antitumor activities and safety profiles. By understanding the current state of siRNA-based therapy and addressing the existing challenges, we aim to pave the way for translating siRNA technology into effective oncologic clinics as an improved treatment options for cancer patients.
Collapse
Affiliation(s)
- Houman Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Farshid Oruji
- College of Medicine, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shima Shabani
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Javad Behroozi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran.
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Luo T, Guan H, Liu J, Wang J, Zhang Y. Curcumin inhibits esophageal squamous cell carcinoma progression through down-regulating the circNRIP1/miR-532-3p/AKT pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2705-2716. [PMID: 37471645 DOI: 10.1002/tox.23905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/20/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Curcumin shows an anti-cancer role in many kinds of tumors. However, the mechanism of its anti-tumor function in esophageal squamous cell carcinoma (ESCC) remains largely unknown. Herein, we explored the therapeutic potential of curcumin for esophageal cancer. Curcumin could time- and dose-dependently inhibit ESCC cells activity. Additionally, ESCC cells exposed to 20 μM of curcumin exhibited significantly decreased proliferative and invasive capacities, as well as enhanced cell apoptosis. ESCC tissues and cells exhibited significantly increased circNRIP1 expression when compared to their counterparts. circNRIP1 knockdown markedly impaired cell proliferation, clone formation, cell migration and invasion but promoted apoptosis. Exposure to 10-20 μM of curcumin inhibited circNRIP1 expression, however, overexpression of circNRIP1 could significantly restored the biological characteristics that were inhibited by curcumin exposure in vivo and in vitro. circNRIP1 promoted the malignancy of ESCC by combining miR-532-3p, and downstream AKT3. Curcumin inhibited AKT phosphorylation by up-regulating miR-532-3p expression, thereby inhibiting the activation of the AKT pathway. In summary, curcumin is a potent inhibitor of ESCC growth, which can be achieved through the regulation of the circNRIP1/miR-532-3p/AKT pathway. This research may provide new mechanisms for curcumin to inhibit the malignant development of ESCC.
Collapse
Affiliation(s)
- Tianxia Luo
- Department of Physiology, School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hongya Guan
- Trauma Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jia Liu
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jiang Wang
- Department of Gastrointestinal Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yueli Zhang
- Department of Clinical Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Esmaeili H, Nasrollahzadeh Sabet M, Mosaed R, Chamanara M, Hadi S, Hazrati E, Farhadi A, Heidari MF, Behroozi J. Oleanolic acid increases the anticancer potency of doxorubicin in pancreatic cancer cells. J Biochem Mol Toxicol 2023; 37:e23426. [PMID: 37345903 DOI: 10.1002/jbt.23426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/20/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
Combination therapy is a novel cancer therapy approach that combines two or more chemotherapy drugs. This treatment modality enhances the efficacy of chemotherapy by targeting key pathways in an additive or synergistic manner. Therefore, we investigated the efficacy of combination therapy by widely used chemotherapy drug doxorubicin (DOX) and oleanolic acid (OA) to induction of apoptosis for pancreatic cancer (PC) therapy. The effects of DOX, OA, and their combination (DOX-OA) were investigated on proliferation and viability of PC cell line (PANC-1) by MTT assay. Moreover, migration and invasion of the cancer cells were evaluated by trans-well migration assay and wound healing assay. Flow cytometry and DAPI (4',6-diamidino-2-phenylindole) staining were employed to investigate apoptosis quantification and qualification of the treated cancer cells. Finally, mRNA expression of apoptosis-related genes was assessed by quantitative real-time polymerase chain reaction. Our results demonstrated that the proliferation and metastasis potential of PC cells significantly decreased after treatment by DOX, OA, and DOX-OA. Moreover, we observed an increase in apoptosis percentage in the treated cancer cells. The apoptosis-related gene expression was modified to increase the apoptosis rate in all of the treatment groups. However, the anticancer potency of DOX-OA combination was significantly more than that of DOX and OA treatments alone. Our study suggested that DOX-OA combination exerts more profound anticancer effects against PC cell lines than DOX or OA monotherapy. This approach may increase the efficiency of chemotherapy and reduce unintended side effects by lowering the prescribed dose of DOX.
Collapse
Affiliation(s)
- Hosein Esmaeili
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mehrdad Nasrollahzadeh Sabet
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Saeid Hadi
- Department of Health, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ebrahim Hazrati
- Department of Anesthesiology and Critical Care, AJA University of Medical Sciences, Tehran, Iran
| | - Arezoo Farhadi
- Department of Genetics and Biotechnology, Faculty of Life Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Mohammad Foad Heidari
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Javad Behroozi
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
- Department of Genetics and Advanced Medical Technology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Vergaro V, Dell'Anna MM, Shahsavari HR, Baldassarre F, Migoni D, Mastrorilli P, Fanizzi FP, Ciccarella G. Synthesis of a light-responsive platinum curcumin complex, chemical and biological investigations and delivery to tumor cells by means of polymeric nanoparticles. NANOSCALE ADVANCES 2023; 5:5340-5351. [PMID: 37767039 PMCID: PMC10521244 DOI: 10.1039/d3na00200d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/04/2023] [Indexed: 09/29/2023]
Abstract
Platinum-based anticancer drugs are common in chemotherapy, but problems such as systemic toxicity and acquired resistance of some tumors hamper their clinical applications and therapeutic efficacy. It is necessary to synthesize Pt-based drugs and explore strategies to reduce side effects and improve pharmacokinetic profiles. Photo-responsive chemotherapeutics have emerged as an alternative strategy against several cancers, as photoactivation offers spatial selectivity and fewer side effects. Here, we combine chemical synthesis and nanotechnology to create a multifunctional platinum drug delivery system based on the novel metal complex [Pt(ppy)(curc)] (ppy = deprotonated 2-phenylpyridine, curc = deprotonated curcumin)] embodying the naturally occurring bioactive molecule, curcumin. The ultrasonication method coupled with the layer-by-layer technology was employed to produce nanocolloids, which demonstrated a good biocompatibility, higher solubility in aqueous solution, stability, large drug loading, and good biological activity in comparison with the free drug. In vitro release experiments revealed that the polymeric nanoformulation is relatively stable under physiological conditions (pH = 7.4 and 37 °C) but sensitive to acidic environments (pH = 5.6 and 37 °C) which would trigger the release of the loaded drug. Our approach modifies the bioavailability of this Pt-based drug increasing its therapeutic action in terms of both cytotoxic and anti-metastasis effects.
Collapse
Affiliation(s)
- Viviana Vergaro
- Biological and Environmental Sciences Department, UdR INSTM of Lecce University of Salento Via Monteroni 73100 Lecce Italy
- Institute of Nanotechnology, CNR NANOTEC, Consiglio Nazionale delle Ricerche Via Monteroni 73100 Lecce Italy
| | | | - Hamid R Shahsavari
- DICATECh, Politecnico di Bari via Orabona, 4 70125 Bari Italy
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Francesca Baldassarre
- Biological and Environmental Sciences Department, UdR INSTM of Lecce University of Salento Via Monteroni 73100 Lecce Italy
- Institute of Nanotechnology, CNR NANOTEC, Consiglio Nazionale delle Ricerche Via Monteroni 73100 Lecce Italy
| | - Danilo Migoni
- Biological and Environmental Sciences Department, UdR INSTM of Lecce University of Salento Via Monteroni 73100 Lecce Italy
| | | | - Francesco Paolo Fanizzi
- Biological and Environmental Sciences Department, UdR INSTM of Lecce University of Salento Via Monteroni 73100 Lecce Italy
| | - Giuseppe Ciccarella
- Biological and Environmental Sciences Department, UdR INSTM of Lecce University of Salento Via Monteroni 73100 Lecce Italy
- Institute of Nanotechnology, CNR NANOTEC, Consiglio Nazionale delle Ricerche Via Monteroni 73100 Lecce Italy
| |
Collapse
|
12
|
Heiat M, Rezaei E, Gharechahi J, Abbasi M, Behroozi J, Abyazi MA, Baradaran B. Knockdown of SIX4 inhibits pancreatic cancer cells via apoptosis induction. Med Oncol 2023; 40:287. [PMID: 37656231 DOI: 10.1007/s12032-023-02163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Sine oculis homeobox 4 (SIX4), a critical transcription factor modulating organ development, potentially participates in tumorigenesis through numerous pathways. Here, we investigated siRNA-mediated knockdown effects of SIX4 on pancreatic cancer cells and underlying molecular mechanisms. The expression of SIX4 in pancreatic cancer and adjacent tissues were investigated in clinical tissue samples and bioinformatically approved by gene expression omnibus (GEO) database. Appropriate siRNA transfected into PANC1 pancreatic cancer cells in order to SIX4 knockdown. The survival, migration, invasion, colony formation, mitochondrial membrane potential, apoptosis, autophagy, and cell cycle in the cancer cells were investigated after knockdown of SIX4. In addition, expression of genes involved in apoptosis and metastasis were assessed in the transfected cancer cells in mRNA and protein levels. High-throughput analysis using GEO database confirmed the overexpression of SIX4 in pancreatic cancer tissues by six independent pancreatic cancer microarrays. Knockdown of SIX4 by specific siRNA significantly decreased survival, colony formation, and mitochondrial membrane potential of the cancer cells. Further assessments demonstrated that knockdown of SIX4 increases the apoptosis and autophagy rates in the cancer cells through modifying the expression of related genes. Moreover, a significant decrease in migration and invasion rates were observed in SIX4 suppressed group. Furthermore, frequency of the cells transfected with SIX4 siRNA increased slightly in G1 and Sub-G1 phases of cell cycle. Our study suggested that siRNA-mediated knockdown of SIX4 increases the pancreatic cancer cells death and reduces the invasion and migration of the cancer cells through different molecular pathways.
Collapse
Affiliation(s)
- Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Rezaei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Javad Gharechahi
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoumeh Abbasi
- Department of Microbiology, Malekan Branch, Islamic Azad University, Malekan, Iran
| | - Javad Behroozi
- Department of Genetics and Biotechnology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Abyazi
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, 5166614766, Tabriz, Iran.
| |
Collapse
|
13
|
Hu Y, Zhang J, Dong L, Xu L, Chen E. DOX-loaded mesoporous hydroxyapatite modified by hyaluronic acid can achieve efficient targeted therapy for lung cancer. J Drug Target 2023; 31:612-622. [PMID: 37067080 DOI: 10.1080/1061186x.2023.2204411] [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: 08/04/2022] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 04/18/2023]
Abstract
It is a novel therapeutic strategy to suppress tumour growth and metastasis by regulating the interaction between bioactivity ions and the biological process of tumour cells. This study synthesised a mesoporous hydroxyapatite (MHAP)-based nanocarrier for targeted delivery of the anti-cancer drug doxorubicin (DOX). To further strengthen the targeting of DOX-loaded nanocarrier to tumour, HA that could specifically identify receptor on the surface of tumours was functionally modified. The drug release properties curve showed that the MHAP-HA@DOX complex showed pH-sensitive and sustained release properties. Also, the MHAP-HA@DOX complex represented high toxicity against lung cancer A549 cells. Besides, it displayed a significant inhibitory effect on tumour growth rate in tumour-bearing mice, while no evident toxicity for mice was observed. This nano-material is hoped to be an effective and novel nano-drug for lung cancer.
Collapse
Affiliation(s)
- Yanjie Hu
- Department of Respiratory and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jisong Zhang
- Department of Respiratory and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liangliang Dong
- Department of Respiratory and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Xu
- Department of Respiratory and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Enguo Chen
- Department of Respiratory and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Avcı CB, Sogutlu F, Pinar Ozates N, Shademan B, Gunduz C. Enhanced Anti-cancer Potency Using a Combination of Oleanolic Acid and Maslinic Acid to Control Treatment Resistance in Breast Cancer. Adv Pharm Bull 2023; 13:611-620. [PMID: 37646060 PMCID: PMC10460813 DOI: 10.34172/apb.2023.057] [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: 02/26/2022] [Revised: 07/11/2022] [Accepted: 09/09/2022] [Indexed: 09/01/2023] Open
Abstract
Purpose The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/ mTOR) pathway is a complex intracellular metabolic pathway that leads to cell growth and tumor proliferation and plays a key role in drug resistance in breast cancer. Therefore, the anti-cancer effects of oleanolic acid (OA), maslinic acid (MA), and their combination were investigated to improve the performance of the treatment strategy. Methods We investigated the effect of OA and MA on cell viability using the WST-1 method. The synergistic effect of the combination was analyzed by isobologram analysis. In addition, the effects of the two compounds, individually and in combination, on apoptosis, autophagy, and the cell cycle were investigated in MCF7 cells. In addition, changes in the expression of PI3K/AKT/mTOR genes involved in apoptosis, cell cycle and metabolism were determined by quantitative RT-PCR. Results MA, OA, and a combination of both caused G0/G1 arrest. Apoptosis also increased in all treated groups. The autophagosomal LC3-II formation was induced 1.74-fold in the MA-treated group and 3.25-fold in the MA-OA-treated group. The combination treatment resulted in increased expression of genes such as GSK3B, PTEN, CDKN1B and FOXO3 and decreased expression of IGF1, PRKCB and AKT3 genes. Conclusion The results showed that the combination of these two substances showed the highest synergistic effect at the lowest dose and using MA-OA caused cancer cells to undergo apoptosis. The use of combination drugs may reduce the resistance of cancer cells to treatment.
Collapse
Affiliation(s)
- Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | | | | | | | | |
Collapse
|
15
|
Zygouri P, Athinodorou AM, Spyrou K, Simos YV, Subrati M, Asimakopoulos G, Vasilopoulos KC, Vezyraki P, Peschos D, Tsamis K, Gournis DP. Oxidized-Multiwalled Carbon Nanotubes as Non-Toxic Nanocarriers for Hydroxytyrosol Delivery in Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:714. [PMID: 36839082 PMCID: PMC9965370 DOI: 10.3390/nano13040714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Carbon nanotubes (CNTs) possess excellent physicochemical and structural properties alongside their nano dimensions, constituting a medical platform for the delivery of different therapeutic molecules and drug systems. Hydroxytyrosol (HT) is a molecule with potent antioxidant properties that, however, is rapidly metabolized in the organism. HT immobilized on functionalized CNTs could improve its oral absorption and protect it against rapid degradation and elimination. This study investigated the effects of cellular oxidized multiwall carbon nanotubes (oxMWCNTs) as biocompatible carriers of HT. The oxidation of MWCNTs via H2SO4 and HNO3 has a double effect since it leads to increased hydrophilicity, while the introduced oxygen functionalities can contribute to the delivery of the drug. The in vitro effects of HT, oxMWCNTS, and oxMWCNTS functionalized with HT (oxMWCNTS_HT) were studied against two different cell lines (NIH/3T3 and Tg/Tg). We evaluated the toxicity (MTT and clonogenic assay), cell cycle arrest, and reactive oxygen species (ROS) formation. Both cell lines coped with oxMWCNTs even at high doses. oxMWCNTS_HT acted as pro-oxidants in Tg/Tg cells and as antioxidants in NIH/3T3 cells. These findings suggest that oxMWCNTs could evolve into a promising nanocarrier suitable for targeted drug delivery in the future.
Collapse
Affiliation(s)
- Panagiota Zygouri
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
| | - Antrea M. Athinodorou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantinos Spyrou
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
| | - Yannis V. Simos
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Mohammed Subrati
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | - Georgios Asimakopoulos
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | | | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Peschos
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantinos Tsamis
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios P. Gournis
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
16
|
Zhang X, Cui Y, Song X, Jin X, Sheng X, Xu X, Li T, Chen H, Gao L. Curcumin alleviates ketamine-induced oxidative stress and apoptosis via Nrf2 signaling pathway in rats' cerebral cortex and hippocampus. ENVIRONMENTAL TOXICOLOGY 2023; 38:300-311. [PMID: 36305173 DOI: 10.1002/tox.23697] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
AIMS To investigate curcumin's protective effect on nerve damage caused by ketamine anesthesia via the Nrf2 signaling pathway. Rats and PC12 cells were used in this experiment to investigate the mechanism of nerve injury caused by ketamine anesthesia. Furthermore, our findings suggest that curcumin may affect oxidative stress and apoptosis by targeting the Nrf2 pathway, thereby alleviating the nerve injury caused by ketamine. METHODS The rat cerebral cortex and hippocampus were stained with Nissl and immunohistochemistry to determine the number of neurons and the expression of Caspase-3, Bcl-2, and Bax. CCK-8 assay was used to determine the optimal concentration of ketamine, curcumin, and H2 O2 in PC12 cells. Flow cytometry was used to detect changes in reactive oxygen species and the rate of apoptosis in each group. To determine whether Nrf2 entered the nucleus, immunofluorescence was used. Both tissues and cells were subjected to RT-PCR and Western blotting detection at the same time. The levels of oxidative stress were determined using a malondialdehyde (MDA) and superoxide dismutase (SOD) assay kit. RESULTS Ketamine reduced the number of neurons in the cortex and hippocampus of rats. The proteins Bax and Caspase-3 were upregulated, while Bcl-2 was down-regulated in the cortex and hippocampus. The viability of PC12 cells has decreased. MDA content increased while SOD activity decreased in cortex, hippocampus, and PC12 cells. Ketamine had an effect on the expression of some genes in the Nrf2 signaling pathway as well as apoptosis. Curcumin pretreatment may be able to prevent ketamine-induced damage. CONCLUSIONS The oxidative stress and apoptosis caused by ketamine during growth of the cerebral cortex, hippocampus, and PC12 cells may be decreased by curcumin's activation of the Nrf2 signaling pathway. Our research provides a potential strategy for the secure administration of anesthetics in medical settings.
Collapse
Affiliation(s)
- Xintong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaopeng Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaodi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuanbo Sheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyu Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ting Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hong Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin, China
| |
Collapse
|
17
|
De Leo V, Maurelli AM, Giotta L, Daniello V, Di Gioia S, Conese M, Ingrosso C, Ciriaco F, Catucci L. Polymer Encapsulated Liposomes for Oral Co-Delivery of Curcumin and Hydroxytyrosol. Int J Mol Sci 2023; 24:ijms24010790. [PMID: 36614233 PMCID: PMC9821336 DOI: 10.3390/ijms24010790] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Curcumin (Cur) is a hydrophobic polyphenol from the rhizome of Curcuma spp., while hydroxytyrosol (HT) is a water-soluble polyphenol from Olea europaea. Both show outstanding antioxidant properties but suffer from scarce bioavailability and low stability in biological fluids. In this work, the co-encapsulation of Cur and HT into liposomes was realized, and the liposomal formulation was improved using polymers to increase their survival in the gastrointestinal tract. Liposomes with different compositions were formulated: Type 1, composed of phospholipids and cholesterol; Type 2, also with a PEG coating; and Type 3 providing an additional shell of Eudragit® S100, a gastro-resistant polymer. Samples were characterized in terms of size, morphology, ζ-potential, encapsulation efficiency, and loading capacity. All samples were subjected to a simulated in vitro digestion and their stability was investigated. The Eudragit®S100 coating demonstrated prevention of early releases of HT in the mouth and gastric phases, while the PEG shell reduced bile salts and pancreatin effects during the intestinal digestion. In vitro antioxidant activity showed a cumulative effect for Cur and HT loaded in vesicles. Finally, liposomes with HT concentrations up to 40 μM and Cur up to 4.7 μM, alone or in combination, did not show cytotoxicity against Caco-2 cells.
Collapse
Affiliation(s)
- Vincenzo De Leo
- Department of Chemistry, University of Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
- Correspondence: (V.D.L.); (L.C.)
| | - Anna Maria Maurelli
- Department of Chemistry, University of Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Livia Giotta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, S.P. Lecce-Monteroni, 73100 Lecce, Italy
| | - Valeria Daniello
- Department of Medical and Surgical Sciences, University of Foggia, Viale L. Pinto 1, 71122 Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Viale L. Pinto 1, 71122 Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Viale L. Pinto 1, 71122 Foggia, Italy
| | - Chiara Ingrosso
- CNR-IPCF S.S. Bari, c/o Department of Chemistry, University of Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Fulvio Ciriaco
- Department of Chemistry, University of Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Lucia Catucci
- Department of Chemistry, University of Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
- Correspondence: (V.D.L.); (L.C.)
| |
Collapse
|
18
|
Phenylboronic Acid-Grafted Chitosan Nanocapsules for Effective Delivery and Controllable Release of Natural Antioxidants: Olive Oil and Hydroxytyrosol. Pharmaceutics 2022; 15:pharmaceutics15010081. [PMID: 36678711 PMCID: PMC9867062 DOI: 10.3390/pharmaceutics15010081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Olives and virgin olive oil (VOO) are a staple of Mediterranean diets and are rich in several beneficial phenolic compounds, including hydroxytyrosol (HT). Therefore, VOO was extracted from Koroneiki olive fruits, and its volatile as well as phenolic components were identified. Meanwhile, in order to upgrade the pharmaceutical capabilities of VOO and HT, a new conjugate phenylboronic acid-chitosan nanoparticles (PBA-CSNPs, NF-1) was fabricated and applied as nanocapsules for implanting high loading and efficient delivery of VOO and HT nanoformulations (NF-2 and NF-3). Due to the H-bonding interactions and boronate ester formation between the hydroxyl groups of the phenolic content of VOO or HT and the PBA groups in the nanocapsules (NF-1), VOO and HT were successfully loaded into the PBA-CSNPs nanocapsules with high loading contents and encapsulation efficacies. The NF-2 and NF-3 nanoformulations demonstrated physicochemical stability, as revealed by their respective zeta potential values, and pH-triggered drug release characteristics. The in vitro studies demonstrated that the nascent nanocapsules were almost completely nontoxic to both healthy and cancer cells, whereas VOO-loaded (NF-2) and HT-loaded nanocapsules (NF-3) showed efficient anti-breast cancer efficiencies. In addition, the antimicrobial and antioxidant potentials of VOO and HT were significantly improved after nanoencapsulation.
Collapse
|
19
|
Mostofi S, Shanehbandi D, Rahmani SA, Asadi M. Anti-migratory effect of curcumin on A-549 lung cancer cells via epigenetic reprogramming of RECK/ matrix metalloproteinase axis. Horm Mol Biol Clin Investig 2022; 43:455-461. [PMID: 35993840 DOI: 10.1515/hmbci-2021-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/11/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the effects of curcumin on the viability, migration, and apoptosis of A549 lung cancer cells. Furthermore, RECK/MMPs axis as a probable regulator of cancer cell migration was assessed. METHODS In this study, effect of curcumin on viability changes, cell migration, and percentage of apoptosis of A549 non-small cell lung carcinoma was examined. The methylation status of RECK gene was investigated using MS-HRM technique. Moreover, expression changes of genes involved in apoptosis and migration (including CASP3, CASP8, CASP9, BAX, BCL2, MMP9, MMP2, and RECK) were investigated by quantitative Real-Time PCR. RESULTS The results of MTT assay showed that the cytotoxic effect of curcumin was in a dose dependent manner. Flow cytometry results demonstrated a significant increase in the percentage of apoptotic cells in curcumin treated group. In addition, curcumin inhibited migration rate in lung cancer cells. qRT-PCR revealed that expression of the candidate genes was in line with suppressed growth and migration. This could be due to, decreased methylation of the RECK gene promoter after curcumin treatment. CONCLUSIONS Curcumin inhibited lung cancer cells through various molecular pathways. RECK/MMPs axis as a regulator of cancer cell migration was modulated after curcumin treatment and invasion of lung cancer cells was decreased.
Collapse
Affiliation(s)
- Shabnam Mostofi
- Department of Genetics, Faculty of Basic Sciences, Islamic Azad University, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, 5166614766, Tabriz, Iran
| | - Seyyed Ali Rahmani
- Department of Genetics, Faculty of Basic Sciences, Islamic Azad University, Ahar, Iran
| | - Milad Asadi
- Institute of Health Sciences, Department of Basic Oncology, Ege University, Izmir, Turkey
| |
Collapse
|
20
|
Shademan B, Masjedi S, Karamad V, Isazadeh A, Sogutlu F, Rad MHS, Nourazarian A. CRISPR Technology in Cancer Diagnosis and Treatment: Opportunities and Challenges. Biochem Genet 2022; 60:1446-1470. [PMID: 35092559 DOI: 10.1007/s10528-022-10193-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022]
Abstract
A novel gene editing tool, the Cas system, associated with the CRISPR system, is emerging as a potential method for genome modification. This simple method, based on the adaptive immune defense system of prokaryotes, has been developed and used in human cancer research. These technologies have tremendous therapeutic potential, especially in gene therapy, where a patient-specific mutation is genetically corrected to cure diseases that cannot be cured with conventional treatments. However, translating CRISPR/Cas9 into the clinic will be challenging, as we still need to improve the efficiency, specificity, and application of the technology. In this review, we will explain how CRISPR-Cas9 technology can treat cancer at the molecular level, focusing on ordination and the epigenome. We will also focus on the promise and shortcomings of this system to ensure its application in the treatment and prevention of cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Sepideh Masjedi
- Department of Cellular and Molecular Biology Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatma Sogutlu
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
21
|
Farhoudi Sefidan Jadid M, Jahangirzadehd G, Behroozi J. Anti-proliferation effects of Apatinib in combination with Curcumin in breast cancer cells. Horm Mol Biol Clin Investig 2022; 44:27-32. [PMID: 36056785 DOI: 10.1515/hmbci-2022-0036] [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: 04/09/2022] [Accepted: 07/20/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Despite remarkable development of new therapeutic strategies to improve survival rates and treatment of patients with cancer, there are still many limitations in management of patients with distant metastasis breast cancer. Therefore, the aim of this study was to investigate a novel method to enhance therapeutic efficacy of Apatinib (as a chemotherapeutic agent) by co-administration of Curcumin (as a bioactive herbal compound) in breast cancer treatment. METHODS Effects of Apatinib, Curcumin, and their combinations (Apa-Cur) was evaluated on viability and proliferation of breast cell line (MCF7) by MTT assay. Moreover, effects of Apatinib, Curcumin, and Apa-Cur was investigated on apoptosis rate in the cancer cells. Expression levels of apoptosis-related genes (BAX, SMAC, BCL2, and SURVIVIN) in treated cancer cells and untreated controls were evaluated using the Real-Time PCR method. RESULTS The obtained results showed that all treatments of Apatinib, Curcumin, and Apa-Cur significantly decreased viability and proliferation of the breast cancer cells in a concentration- and time-dependent manner. However, anti-proliferation activity of Apa-Cur combination was significantly higher than Apatinib and Curcumin treatment alone. In addition, Apatinib, Curcumin, and Apa-Cur increased apoptosis percentage in the treated cancer cells through regulation of apoptosis-related genes expression. CONCLUSIONS In general, Apa-Cur combination therapy exerts more profound anti-proliferation effects on breast cancer cell than Apatinib or Curcumin monotherapy. However, further studies are required to identify other possible signaling pathways and mechanisms involved in the anticancer effects of Apatinib, Curcumin, and Apa-Cur.
Collapse
Affiliation(s)
| | | | - Javad Behroozi
- Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran.,Department of Genetics and Advanced Medical Technology, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Food-Derived Bioactive Molecules from Mediterranean Diet: Nanotechnological Approaches and Waste Valorization as Strategies to Improve Human Wellness. Polymers (Basel) 2022; 14:polym14091726. [PMID: 35566894 PMCID: PMC9103748 DOI: 10.3390/polym14091726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
The beneficial effects of the Mediterranean diet (MedDiet), the most widely followed healthy diet in the world, are principally due to the presence in the foods of secondary metabolites, mainly polyphenols, whose healthy characteristics are widely recognized. However, one of the biggest problems associated with the consumption of polyphenols as nutraceutical adjuvant concerns their bioavailability. During the last decades, different nanotechnological approaches have been developed to enhance polyphenol bioavailability, avoiding the metabolic modifications that lead to low absorption, and improving their retention time inside the organisms. This review focuses on the most recent findings regarding the encapsulation and delivery of the bioactive molecules present in the foods daily consumed in the MedDiet such as olive oil, wine, nuts, spice, and herbs. In addition, the possibility of recovering the polyphenols from food waste was also explored, taking into account the increased market demand of functional foods and the necessity to obtain valuable biomolecules at low cost and in high quantity. This circular economy strategy, therefore, represents an excellent approach to respond to both the growing demand of consumers for the maintenance of human wellness and the economic and ecological exigencies of our society.
Collapse
|
23
|
Caligiuri R, Di Maio G, Godbert N, Scarpelli F, Candreva A, Rimoldi I, Facchetti G, Lupo MG, Sicilia E, Mazzone G, Ponte F, Romeo I, La Deda M, Crispini A, De Rose R, Aiello I. Curcumin-based ionic Pt( ii) complexes: antioxidant and antimicrobial activity. Dalton Trans 2022; 51:16545-16556. [DOI: 10.1039/d2dt01653b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Four new Pt(ii) ionic complexes assembled from N-donor ligands and curcumin display interesting antioxidant and antimicrobial properties.
Collapse
Affiliation(s)
- Rossella Caligiuri
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Giuseppe Di Maio
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Nicolas Godbert
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Francesca Scarpelli
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Angela Candreva
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
- CNR NANOTEC-Istituto di Nanotecnologia UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy
| | - Isabella Rimoldi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milan, Italy
| | - Giorgio Facchetti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maria Giovanna Lupo
- Dipartimento di Medicina, Università degli Studi di Padova, 35128 Padova, Italy
| | - Emilia Sicilia
- Dipartimento di Chimica e Tecnologie Chimiche, 87036 Arcavacata di Rende, CS, Italy
| | - Gloria Mazzone
- Dipartimento di Chimica e Tecnologie Chimiche, 87036 Arcavacata di Rende, CS, Italy
| | - Fortuna Ponte
- Dipartimento di Chimica e Tecnologie Chimiche, 87036 Arcavacata di Rende, CS, Italy
| | - Isabella Romeo
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Massimo La Deda
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
- CNR NANOTEC-Istituto di Nanotecnologia UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy
| | - Alessandra Crispini
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Renata De Rose
- LAB CF-INABEC, Dipartimento di Chimica e Tecnologie Chimiche, 87036 Arcavacata di Rende, CS, Italy
| | - Iolinda Aiello
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, 87036 Arcavacata di Rende, CS, Italy
- CNR NANOTEC-Istituto di Nanotecnologia UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy
| |
Collapse
|
24
|
Maleki MP, Soltanzade H, Tanomand A, Shahsavari G. The layered double hydroxide (LDH) nanosheets decrease anticancer potential of Satureja khuzestanica in HepG2 hepatocellular carcinoma cells. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
25
|
Abbasi S, Bazyar R, Saremi MA, Alishiri G, Seyyedsani N, Farhoudi Sefidan Jadid M, Khorrami A, Golmarz PE, Jahangirzadeh G, Bedoustan AB, Isazadeh A, Hajazimian S, Amoodizaj FF. Wharton jelly stem cells inhibits AGS gastric cancer cells through induction of apoptosis and modification of MAPK and NF-κB signaling pathways. Tissue Cell 2021; 73:101597. [PMID: 34358919 DOI: 10.1016/j.tice.2021.101597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/14/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Gastric cancer) GC) is one of the most common cancer with high mortality worldwide. The human Wharton's jelly stem cells (hWJSCs) can inhibit several cancer cells through several molecular pathways. Therefore, the present study aimed to investigate anticancer effects of hWJSCs conditioned medium (hWJSC-CM) and cell-free lysate (hWJSC-CL) against of GC cell line AGS and underlying signaling pathways. METHODS In this study, we evaluated the effects of hWJSC-CM and hWJSC-CL on viability, proliferation, migration, invasion, apoptosis, and MAPK and NF-κB signaling pathways in AGS cells. Moreover, mRNA expression of genes involved in apoptosis (BAX, BCL2, SMAC, and SURVIVIN), as well as expression of proteins involved in NF-κB and MAPK signaling pathways were evaluated. RESULTS The obtained results showed that the hWJSC-CM and hWJSC-CL decreased viability, migration, and invasion of GC cell line AGS in a concentration and time dependent manner. We observed that the hWJSC-CM and hWJSC-CL induced apoptosis pathway through regulation of apoptosis involved genes mRNA expression. In addition, the hWJSC-CM and hWJSC-CL suppressed NF-κB signaling pathways as well as promoted MAPK signaling pathways. CONCLUSION In general, our study suggested that the hWJSC-CM and hWJSC-CL inhibits proliferation and viability of GC cell line AGS through induction of apoptosis, as well as modification of NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Samaneh Abbasi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Reza Bazyar
- Personalized Medicine Research Center of AmitisGen, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Saremi
- Personalized Medicine Research Center of AmitisGen, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gholamhoseen Alishiri
- Department of Rheumatology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nasrin Seyyedsani
- Department of Genetics, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Afshin Khorrami
- Young Researchers and Elit Club, Varamin-Pishva Branch, Islamic Azad University, Varamin, Pishva, Iran
| | | | | | | | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
26
|
Witika BA, Makoni PA, Matafwali SK, Mweetwa LL, Shandele GC, Walker RB. Enhancement of Biological and Pharmacological Properties of an Encapsulated Polyphenol: Curcumin. Molecules 2021; 26:4244. [PMID: 34299519 PMCID: PMC8303961 DOI: 10.3390/molecules26144244] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
There is a dearth of natural remedies available for the treatment of an increasing number of diseases facing mankind. Natural products may provide an opportunity to produce formulations and therapeutic solutions to address this shortage. Curcumin (CUR), diferuloylmethane; I,7-bis-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione is the major pigment in turmeric powder which has been reported to exhibit a number of health benefits including, antibacterial, antiviral, anti-cancer, anti-inflammatory and anti-oxidant properties. In this review, the authors attempt to highlight the biological and pharmacological properties of CUR in addition to emphasizing aspects relating to the biosynthesis, encapsulation and therapeutic effects of the compound. The information contained in this review was generated by considering published information in which evidence of enhanced biological and pharmacological properties of nano-encapsulated CUR was reported. CUR has contributed to a significant improvement in melanoma, breast, lung, gastro-intestinal, and genito-urinary cancer therapy. We highlight the impact of nano-encapsulated CUR for efficient inhibition of cell proliferation, even at low concentrations compared to the free CUR when considering anti-proliferation. Furthermore nano-encapsulated CUR exhibited bioactive properties, exerted cytotoxic and anti-oxidant effects by acting on endogenous and cholinergic anti-oxidant systems. CUR was reported to block Hepatitis C virus (HCV) entry into hepatic cells, inhibit MRSA proliferation, enhance wound healing and reduce bacterial load. Nano-encapsulated CUR has also shown bioactive properties when acting on antioxidant systems (endogenous and cholinergic). Future research is necessary and must focus on investigation of encapsulated CUR nano-particles in different models of human pathology.
Collapse
Affiliation(s)
- Bwalya Angel Witika
- ApotheCom|A MEDiSTRAVA Company (Medical Division of Huntsworth), London WC2A 1AN, UK;
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa
| | - Pedzisai Anotida Makoni
- Division of Pharmacology, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa;
| | - Scott Kaba Matafwali
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, LSHTM, London WC1E 7HT, UK;
| | - Larry Lawrence Mweetwa
- Department of Chemistry, Marine Biodiscovery Centre, University of Aberdeen, Aberdeen AB24 3FX, UK;
| | - Ginnethon Chaamba Shandele
- Department of Biochemistry, Institute of Basic and Biomedical Sciences, Levy Mwanawasa Medical University, P.O. Box 33991, Lusaka 10101, Zambia;
| | - Roderick Bryan Walker
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa
| |
Collapse
|
27
|
Isazadeh A, Hajazimian S, Shadman B, Safaei S, Babazadeh Bedoustani A, Chavoshi R, Shanehbandi D, Mashayekhi M, Nahaei M, Baradaran B. Anti-Cancer Effects of Probiotic Lactobacillus acidophilus for Colorectal Cancer Cell Line Caco-2 through Apoptosis Induction. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2020.52] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Colorectal cancer is one of the most common cancers worldwide. Probiotics are useful and non-pathogenic microorganisms in the gastrointestinal tract, which can show anticancer activity through the induction of apoptosis. This study aimed to evaluate the antiproliferative effects of Lactobacillus acidophilus probiotic on the Caco-2 colorectal cancer cell line. Methods: The supernatant (secreted metabolites) and bacterial extract of L. acidophilus probiotics were prepared and used as an anti-proliferative agent on the colorectal cancer cell line, Caco-2 in vitro. The effects of supernatant and extract of L. acidophilus were evaluated on the viability and proliferation of cancer cells using MTT assay. Moreover, morphological alterations of cancer cells treated with supernatant and extract of L. acidophilus were evaluated by an inverted phase contrast microscope. The mRNA expression levels of apoptosis-related genes (SURVIVIN and SMAC) in treated cancer cells and untreated controls were evaluated using the Real-Time PCR method. Results: The results showed that the supernatant and extract of L. acidophilus inhibited the viability and proliferation of cancer cells in a dose and time-dependent manner. Moreover, various morphological alterations were observed in the treated cancer cells, which are indicators of apoptosis induction. The mRNA expression of SURVIVIN and SMAC genes were significantly up-regulated and downregulated in the treated cancer cells, respectively. Conclusion: The results of the present study suggested that the supernatant and extract of L.acidophilus could inhibit the viability and proliferation of colorectal cancer cell line, Caco-2through induction of apoptosis, increase the survival rate of colon cancer patients.
Collapse
Affiliation(s)
- Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Genetics, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrouz Shadman
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Chavoshi
- Department of Genetics, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammadreza Nahaei
- Department of Microbiology and Laboratory Sciences, School of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|