1
|
Pravin N, Jóźwiak K. PROTAC unleashed: Unveiling the synthetic approaches and potential therapeutic applications. Eur J Med Chem 2024; 279:116837. [PMID: 39305635 DOI: 10.1016/j.ejmech.2024.116837] [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: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 10/28/2024]
Abstract
Proteolysis-Targeting Chimeras (PROTACs) are a novel class of bifunctional small molecules that alter protein levels by targeted degradation. This innovative approach uses the ubiquitin-proteasome system to selectively eradicate disease-associated proteins, providing a novel therapeutic strategy for a wide spectrum of diseases. This review delineates detailed synthetic approaches involved in PROTAC building blocks, including the ligand and protein binding parts, linker attached structural components of PROTACs and the actual PROTAC molecules. Furthermore, the recent advancements in PROTAC-mediated degradation of specific oncogenic and other disease-associated proteins, such as those involved in neurodegenerative, antiviral, and autoimmune diseases, were also discussed. Additionally, we described the current landscape of PROTAC clinical trials and highlighted key studies that underscore the translational potential of this emerging therapeutic modality. These findings demonstrate the versatility of PROTACs in modulating the levels of key proteins involved in various severe diseases.
Collapse
Affiliation(s)
- Narayanaperumal Pravin
- Department of Biopharmacy, Medical University of Lublin, Ul.W.Chodzki 4a, 20-093 Lublin, Poland.
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, Ul.W.Chodzki 4a, 20-093 Lublin, Poland.
| |
Collapse
|
2
|
Mehdinejad S, Peymani M, Salehzadeh A, Zaefizadeh M. Genetic insights and therapeutic potential for colorectal cancer: mutation analysis of KRAS gene and efficacy of Oleuropein-conjugated iron oxide nanoparticles. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8771-8783. [PMID: 38837069 DOI: 10.1007/s00210-024-03182-9] [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: 01/12/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
This study aimed to address the challenges of treating advanced stages of colon cancer (CRC) by exploring potential therapeutic options. The research focused on the genetic aspects of CRC, specifically the mutation rate of the KRAS gene, along with other genes like TTN, APC, MUC16, and TP53, using the TCGA dataset. Additionally, the study investigated the efficacy of Oleuropein, a polyphenolic compound found in olives, in combating CRC by using iron oxide nanoparticles coated with glucose and conjugated with Oleuropein. The study characterized the physicochemical properties of the nanoparticles, and the cytotoxic effects of the nanoparticles were evaluated on CRC and normal fibroblast cell lines, demonstrating significantly higher cytotoxicity against CRC cells compared to normal cells. Furthermore, the study analyzed gene expression changes using the GSE124627 dataset to understand the influence of KRAS alterations. It identified numerous upregulated and downregulated genes in KRAS-overexpressing samples, suggesting their involvement in critical cancer-related pathways. These findings suggest that KRAS-influenced genes could serve as potential therapeutic targets for CRC treatment. The study also examined the expression levels of identified genes in CRC samples compared to normal samples. Among the upregulated genes, 22 showed significant increases in cancer samples, while 14 downregulated genes exhibited decreased expression in both KRAS-influenced and cancer samples. Cox regression analysis identified specific upregulated genes, including ANKZF1, SNAI1, PPFIA4, SIX4, and NOTUM, associated with poor prognosis. Kaplan-Meier analysis further confirmed the correlation between increased expression of these genes and higher patient mortality rates. In conclusion, this study provided valuable insights into the genetic aspects of CRC and potential therapeutic strategies. The use of Oleuropein-conjugated iron oxide nanoparticles showed promising cytotoxic effects on colon cancer cells. These findings contribute to advancing our understanding of CRC and offer potential targets for further investigation and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
3
|
El-Fakharany EM, El-Gendi H, Abdel-Wahhab K, Abu-Serie MM, El-Sahra DG, Ashry M. Therapeutic efficacy of α-lactalbumin coated oleic acid based liposomes against colorectal carcinoma in Caco-2 cells and DMH-treated albino rats. J Biomol Struct Dyn 2024; 42:9220-9234. [PMID: 37624964 DOI: 10.1080/07391102.2023.2250452] [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: 05/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Colorectal cancer (CRC) is a malignant tumor recognized as a major cause of morbidity and mortality throughout the world. Therefore, novel liposomes of oleic acid coated with camel α-lactalbumin (α-LA coated liposomes) were developed for their potential antitumor activity against CRC, both in vitro and in DMH-induced CRC-modeled animal. In vitro results indicated the high safety of α-LA coated liposomes towards normal human cells with potent antitumor activity against Caco-2 cells at an IC50 value of 57.01 ± 3.55 µM with selectivity index of 6.92 ± 0.48. This antitumor activity has been attributed to induction of the apoptotic mechanism, as demonstrated by nuclear condensation and arrest of Caco-2 cells in sub-G1 populations. α-LA coated liposomes also revealed a significant up-regulation of the p53 gene combined with a down-regulation of the Bcl2 gene. Moreover, in vivo results revealed that treatment of induced-CRC modeled animals with α-LA coated liposomes for six weeks markedly improved the CRC-disorders; this was achieved from the significant reduction in the values of AFP, CEA, CA19.9, TNF-α, IL-1β, MDA, and NO coupled with remarkable rise in SOD, GPx, GSH, CAT, and CD4+ levels. The histopathological findings asserted the therapeutic potential of α-LA coated liposomes in the treatment of CRC. Therefore, the present results proved the antitumor activity of α-LA coated liposomes against CRC through the restoration of impaired oxidative stress, improved immune response, and reduced inflammation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA- City), Alexandria, Egypt
| | - Hamada El-Gendi
- Bioprocess Development Department, GEBRI, SRTA-City, Alexandria, Egypt
| | | | - Marwa M Abu-Serie
- Medical Biotechnology Department, GEBRI, SRTA-City, Alexandria, Egypt
| | - Doaa Galal El-Sahra
- Medical Surgical Nursing Department, Faculty of Nursing, Modern University for Technology and Information, Cairo, Egypt
| | - Mahmoud Ashry
- Zoology Department, Faculty of Science, Al-Azhar University, Assuit, Egypt
| |
Collapse
|
4
|
Mahdavi Niyaki Z, Salehzadeh A, Peymani M, Zaefizadeh M. Exploring the Therapeutic Potential of Fe 3O 4@Glu-Oleuropein Nanoparticles in Targeting KRAS Pathway-Regulating lncRNAs in Colorectal Cancer Cells. Biol Trace Elem Res 2024; 202:3073-3085. [PMID: 37792268 DOI: 10.1007/s12011-023-03892-w] [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: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
Cancer, the leading cause of death worldwide, has witnessed significant advancements in treatment through targeted therapies. Among the proto-oncogenes prevalent in human cancers, KRAS stands out, and recent research has focused on long noncoding RNAs (lncRNAs) as regulators of miRNAs targeting the KRAS oncogene. This study specifically explores lncRNAs involved in the KRAS pathway in colorectal cancer (CRC). To investigate this, researchers employed iron oxide nanoparticles coated with glucose and conjugated with Oleuropein (Fe3O4@Glu-Oleuropein NPs) to evaluate their impact on candidate lncRNAs associated with KRAS pathway deregulation. The study utilized TCGA data to identify genes affected by KRAS mutation and lncRNAs linked to KRAS in CRC. Enrichr and MsigDB databases helped identify relevant pathways. Genes with a correlation coefficient above 0.5 and a P-value less than 0.01 with candidate lncRNAs were selected. MTT and flow cytometry assays determined the anti-proliferative and apoptotic effects of Fe3O4@Glu-Oleuropein NPs on CRC cells (SW480) and normal cells (HEK293). The findings showed that increased expression of FEZF1-AS1, GAS6-AS1, and LINC00920 correlated with mutated KRAS, and co-expressed genes were significantly involved in hypoxia, KRAS signaling, DNA repair, and IL-2/STAT5 signaling pathways. Fe3O4@Glu-Oleuropein NPs exhibited higher toxicity toward cancer cells, with IC50 values of 92 μg/ml for SW480 and 281 μg/ml for HEK293. Flow cytometry analysis revealed a substantial increase in necrotic and apoptotic cells when treated with Fe3O4@Glu-Oleuropein, along with down-regulation of GAS6-AS1, LINC00920, and FEZF1-AS1 lncRNAs in treated cells. In conclusion, this study highlights the therapeutic potential of Fe3O4@Glu-Oleuropein on colon cancer cells in vitro. The identification of lncRNAs involved in the KRAS pathway provides insights into the underlying mechanisms and offers avenues for further research in targeted cancer therapies.
Collapse
Affiliation(s)
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
5
|
Moghimipour E, Handali S. Functionalized liposomes as a potential drug delivery systems for colon cancer treatment: A systematic review. Int J Biol Macromol 2024; 269:132023. [PMID: 38697444 DOI: 10.1016/j.ijbiomac.2024.132023] [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: 07/14/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Colon cancer is one of the lethal diseases in the world with approximately 700,000 fatalities annually. Nowadays, due to the side effects of existing methods in the treatment of colon cancer such as radiotherapy and chemotherapy, the use of targeted nanocarriers in cancer treatment has received wide attention, and among them, especially liposomes have been studied a lot. Based on this, anti-tumor drugs hidden in targeted active liposomes can selectively act on cancer cells. In this systematic review, the use of various ligands such as folic acid, transferrin, aptamer, hyaluronic acid and cRGD for active targeting of liposomes to achieve improved drug delivery to colon cancer cells has been reviewed. The original articles published in English in the databases of Science Direct, PubMed and Google scholar from 2012 to 2022 were reviewed. From the total of 26,256 published articles, 19 studies met the inclusion criteria. The results of in vitro and in vivo studies have revealed that targeted liposomes lead to increasing the efficacy of anti-cancer agents on colon cancer cells with reducing side effects compared to free drugs and non-targeted liposomes. To the best of our knowledge, this is the first systematic review showing promising results for improvement treatment of colon cancer using targeted liposomes.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Bhat AA, Kukreti N, Afzal M, Goyal A, Thapa R, Ali H, Shahwan M, Almalki WH, Kazmi I, Alzarea SI, Singh SK, Dua K, Gupta G. Ferroptosis and circular RNAs: new horizons in cancer therapy. EXCLI JOURNAL 2024; 23:570-599. [PMID: 38887390 PMCID: PMC11180955 DOI: 10.17179/excli2024-7005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/09/2024] [Indexed: 06/20/2024]
Abstract
Cancer poses intricate challenges to treatment due to its complexity and diversity. Ferroptosis and circular RNAs (circRNAs) are emerging as innovative therapeutic avenues amid the evolving landscape of cancer therapy. Extensive investigations into circRNAs reveal their diverse roles, ranging from molecular regulators to pivotal influencers of ferroptosis in cancer cell lines. The results underscore the significance of circRNAs in modulating molecular pathways that impact crucial aspects of cancer development, including cell survival, proliferation, and metastasis. A detailed analysis delineates these pathways, shedding light on the molecular mechanisms through which circRNAs influence ferroptosis. Building upon recent experimental findings, the study evaluates the therapeutic potential of targeting circRNAs to induce ferroptosis. By identifying specific circRNAs associated with the etiology of cancer, this analysis paves the way for the development of targeted therapeutics that exploit vulnerabilities in cancer cells. This review consolidates the existing understanding of ferroptosis and circRNAs, emphasizing their role in cancer therapy and providing impetus for ongoing research in this dynamic field. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U. P., India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| |
Collapse
|
7
|
Zamanian MY, Golmohammadi M, Abdullaev B, García MO, Alazbjee AAA, Kumar A, Mohaamed SS, Hussien BM, Khalaj F, Hodaei SM, Shirsalimi N, Moriasi G. A narrative review on therapeutic potential of naringenin in colorectal cancer: Focusing on molecular and biochemical processes. Cell Biochem Funct 2024; 42:e4011. [PMID: 38583080 DOI: 10.1002/cbf.4011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Colorectal cancer (CRC) is a common and highly metastatic cancer affecting people worldwide. Drug resistance and unwanted side effects are some of the limitations of current treatments for CRC. Naringenin (NAR) is a naturally occurring compound found in abundance in various citrus fruits such as oranges, grapefruits, and tomatoes. It possesses a diverse range of pharmacological and biological properties that are beneficial for human health. Numerous studies have highlighted its antioxidant, anticancer, and anti-inflammatory activities, making it a subject of interest in scientific research. This review provides a comprehensive overview of the effects of NAR on CRC. The study's findings indicated that NAR: (1) interacts with estrogen receptors, (2) regulates the expression of genes related to the p53 signaling pathway, (3) promotes apoptosis by increasing the expression of proapoptotic genes (Bax, caspase9, and p53) and downregulation of the antiapoptotic gene Bcl2, (4) inhibits the activity of enzymes involved in cell survival and proliferation, (5) decreases cyclin D1 levels, (6) reduces the expression of cyclin-dependent kinases (Cdk4, Cdk6, and Cdk7) and antiapoptotic genes (Bcl2, x-IAP, and c-IAP-2) in CRC cells. In vitro CDK2 binding assay was also performed, showing that the NAR derivatives had better inhibitory activities on CDK2 than NAR. Based on the findings of this study, NAR is a potential therapeutic agent for CRC. Additional pharmacology and pharmacokinetics studies are required to fully elucidate the mechanisms of action of NAR and establish the most suitable dose for subsequent clinical investigations.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Bekhzod Abdullaev
- Central Asian Center of Development Studies, New Uzbekistan University, Tashkent, Uzbekistan
- School of Medicine, Central Asian University, Tashkent, Uzbekistan
- Department of Medical Oncology and Radiology, Samarkand State Medical University
| | - María Olalla García
- Universidad Estatal de Bolívar, Facultad de Ciencias de la Salud y del Ser Humano, Carrera de Enfermería, CP, Guaranda, Ecuador
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, Russia
| | - Sameer S Mohaamed
- Department of Pharmacy, Al Rafidain University College, Bagdad, Iraq
| | - Beneen M Hussien
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| | - Fattaneh Khalaj
- Digestive Diseases Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Niyousha Shirsalimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
8
|
Salek S, Moazamian E, Mohammadi Bardbori A, Shamsdin SA. The anticancer effect of potential probiotic L. fermentum and L. plantarum in combination with 5-fluorouracil on colorectal cancer cells. World J Microbiol Biotechnol 2024; 40:139. [PMID: 38514489 DOI: 10.1007/s11274-024-03929-9] [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: 10/25/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024]
Abstract
5-Fluorouracil (5-FU) is an effective chemotherapy drug in the treatment of colorectal cancer (CRC). However, auxiliary or alternative therapies must be sought due to its resistance and potential side effects. Certain probiotic metabolites exhibit anticancer properties. In this study evaluated the anticancer and potential therapeutic activities of cell extracts potential probiotic strains, Limosilactobacillus fermentum and Lactiplantibacillus plantarum isolated from the mule milk and the standard probiotic strain Lacticaseibacillus rhamnosus GG (LGG) against the human colon cancer cell line (HT-29) and the normal cell line (HEK-293) alone or in combination with 5-FU. In this study, L. plantarum and L. fermentum, which were isolated from mule milk, were identified using biochemical and molecular methods. Their probiotic properties were investigated in vitro and compared with the standard probiotic strain of the species L. rhamnosus GG. The MTT assay, acridine orange/ethidium bromide (AO/EB) fluorescent staining, and flow cytometry were employed to measure the viability of cell lines, cell apoptosis, and production rates of Th17 cytokines, respectively. The results demonstrated that the combination of lactobacilli cell extracts and 5-FU decreased cell viability and induced apoptosis in HT-29 cells. Furthermore, this combination protected HEK-293 cells from the cytotoxic effects of 5-FU, enhancing their viability and reducing apoptosis. Moreover, the combination treatment led to an increase in the levels of IL-17A, IFN-γ, and TNF-α, which can enhance anti-tumor immunity. In conclusion, the cell extracts of the lactobacilli strains probably can act as a potential complementary anticancer therapy.
Collapse
Affiliation(s)
- Sanaz Salek
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Elham Moazamian
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran.
| | - Afshin Mohammadi Bardbori
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedeh Azra Shamsdin
- Gasteroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Arjmand B, Alavi-Moghadam S, Faraji Z, Aghajanpoor-Pasha M, Jalaeikhoo H, Rajaeinejad M, Nikandish M, Faridfar A, Rezazadeh-Mafi A, Rezaei-Tavirani M, Irompour A. The Potential Role of Intestinal Stem Cells and Microbiota for the Treatment of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:115-128. [PMID: 38811486 DOI: 10.1007/5584_2024_803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Colorectal cancer is a global health concern with high incidence and mortality rates. Conventional treatments like surgery, chemotherapy, and radiation therapy have limitations in improving patient survival rates. Recent research highlights the role of gut microbiota and intestinal stem cells in maintaining intestinal health and their potential therapeutic applications in colorectal cancer treatment. The interaction between gut microbiota and stem cells influences epithelial self-renewal and overall intestinal homeostasis. Novel therapeutic approaches, including immunotherapy, targeted therapy, regenerative medicine using stem cells, and modulation of gut microbiota, are being explored to improve treatment outcomes. Accordingly, this chapter provides an overview of the potential therapeutic applications of gut microbiota and intestinal stem cells in treating colorectal cancer.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Faraji
- Iranian Cancer Control Center (MACSA), Tehran, Iran
| | | | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikandish
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Ahmad Rezazadeh-Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shaheed Beheshti Medical University, Tehran, Iran
| | | | - Arsalan Irompour
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Luo CY, Su WC, Jiang HF, Luo LT, Shen DY, Su GQ. DPY30 promotes colorectal carcinoma metastasis by upregulating ZEB1 transcriptional expression. Cancer Cell Int 2023; 23:333. [PMID: 38115111 PMCID: PMC10731791 DOI: 10.1186/s12935-023-03126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023] Open
Abstract
DPY30 belongs to the core subunit of components of the histone lysine methyltransferase complex, which is implicated in tumorigenesis, cell senescence, and other biological events. However, its contribution to colorectal carcinoma (CRC) progression and metastasis has yet to be elucidated. Therefore, this study aimed to investigate the biological function of DPY30 in CRC metastasis both in vitro and in vivo. Herein, our results revealed that DPY30 overexpression is significantly positively correlated with positive lymph nodes, epithelial-mesenchymal transition (EMT), and CRC metastasis. Moreover, DPY30 knockdown in HT29 and SW480 cells markedly decreased EMT progression, as well as the migratory and invasive abilities of CRC cells in vitro and lung tumor metastasis in vivo. Mechanistically, DPY30 increased histone H3K4me3 level and promoted EMT and CRC metastasis by upregulating the transcriptional expression of ZEB1. Taken together, our findings indicate that DPY30 may serve as a therapeutic target and prognostic marker for CRC.
Collapse
Affiliation(s)
- Chun-Ying Luo
- Medical College, Guangxi University, Nanning, 530004, Guangxi Province, People's Republic of China
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, People's Republic of China
| | - Wei-Chao Su
- Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen Xianyue Hospital, Xianyue Hospital Affiliated With Xiamen Medical College, No. 55 Zhenhai Road, Xiamen, 361003, Fujian Province, People's Republic of China
| | - Hai-Feng Jiang
- Department of Colorectal Tumor Surgery, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, No. 55 Zhenhai Road, Xiamen, 361003, Fujian Province, People's Republic of China
| | - Ling-Tao Luo
- Department of Colorectal Tumor Surgery, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, No. 55 Zhenhai Road, Xiamen, 361003, Fujian Province, People's Republic of China
| | - Dong-Yan Shen
- Xiamen Cell Therapy Research Center, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, Fujian Province, People's Republic of China.
| | - Guo-Qiang Su
- Medical College, Guangxi University, Nanning, 530004, Guangxi Province, People's Republic of China.
- Department of Colorectal Tumor Surgery, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, No. 55 Zhenhai Road, Xiamen, 361003, Fujian Province, People's Republic of China.
| |
Collapse
|
11
|
Omer AB, Fatima F, Ahmed MM, Aldawsari MF, Alalaiwe A, Anwer MK, Mohammed AA. Enhanced Apigenin Dissolution and Effectiveness Using Glycyrrhizin Spray-Dried Solid Dispersions Filled in 3D-Printed Tablets. Biomedicines 2023; 11:3341. [PMID: 38137562 PMCID: PMC10742019 DOI: 10.3390/biomedicines11123341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
This study aimed to prepare glycyrrhizin-apigenin spray-dried solid dispersions and develop PVA filament-based 3D printlets to enhance the dissolution and therapeutic effects of apigenin (APN); three formulations (APN1-APN3) were proportioned from 1:1 to 1:3. A physicochemical analysis was conducted, which revealed process yields of 80.5-91% and APN content within 98.0-102.0%. FTIR spectroscopy confirmed the structural preservation of APN, while Powder-XRD analysis and Differential Scanning Calorimetry indicated its transformation from a crystalline to an amorphous form. APN2 exhibited improved flow properties, a lower Angle of Repose, and Carr's Index, enhancing compressibility, with the Hausner Ratio confirming favorable flow properties for pharmaceutical applications. In vitro dissolution studies demonstrated superior performance with APN2, releasing up to 94.65% of the drug and revealing controlled release mechanisms with a lower mean dissolution time of 71.80 min and a higher dissolution efficiency of 19.2% compared to the marketed APN formulation. This signified enhanced dissolution and improved therapeutic onset. APN2 exhibited enhanced antioxidant activity; superior cytotoxicity against colon cancer cells (HCT-116), with a lower IC50 than APN pure; and increased antimicrobial activity. A stability study confirmed the consistency of APN2 after 90 days, as per ICH, with an f2 value of 70.59 for both test and reference formulations, ensuring reliable pharmaceutical development. This research underscores the potential of glycyrrhizin-apigenin solid dispersions for pharmaceutical and therapeutic applications, particularly highlighting the superior physicochemical properties, dissolution behavior, biological activities, and stability of APN2, while the development of a 3D printlet shell offers promise for enhanced drug delivery and therapeutic outcomes in colon cancer treatment, displaying advanced formulation and processing techniques.
Collapse
Affiliation(s)
- Asma B. Omer
- Department of Health Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Farhat Fatima
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; (M.M.A.); (M.F.A.)
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; (M.M.A.); (M.F.A.)
| | - Mohammed F. Aldawsari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; (M.M.A.); (M.F.A.)
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; (M.M.A.); (M.F.A.)
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia; (M.M.A.); (M.F.A.)
| | - Abdul Aleem Mohammed
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66433, Saudi Arabia
| |
Collapse
|
12
|
Tao C, Rouhi J. A biosensor based on graphene oxide nanocomposite for determination of carcinoembryonic antigen in colorectal cancer biomarker. ENVIRONMENTAL RESEARCH 2023; 238:117113. [PMID: 37696325 DOI: 10.1016/j.envres.2023.117113] [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: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/13/2023]
Abstract
Colorectal cancer is still a major global health concern, and early detection and accurate biomarker analyses are critical to its successful management. This paper describes the design and testing of a new biosensor based on a graphene oxide (GO) nanocomposite for the exact measurement of carcinoembryonic antigen (CEA), a well-known biomarker for colorectal cancer. The current study attempted to create a highly sensitive immunosensor for sensitive measurement of CEA based on a polypropylene-imine-dendrimer (PPI) and GO nanocomposite on GCE (PPI/GO/GCE). The PPI/GO nanocomposite served as an appropriate biocompatible nanostructure with a large surface area for immobilizing carcinoembryonic antigen (anti-CEA) and bovine serum albumin (BSA) molecules (BSA/anti-CEA/PPI/GO/GCE), thereby promoting the selectivity of electrochemical immunosensors, according to structural and electrochemical studies. Results showed that the BSA/anti-CEA/PPI/GO/GCE as a selective, sensitive, and stable immunosensor revealed a wide linear response from 0.001 to 2000 ng/mL, and a limit of detection of 0.3 pg/mL, which indicated comparable or better performance towards the CEA immunosensors in recent reports in the literature. This was due to the synergetic effect of the GO nanosheets and PPI with porous structure and more conductivity. Analytical results showed values of RSD (4.49%-5.04%) and recovery (90.00%-99.98%) are suitable for effective and accurate practical assessments in CEA in clinical samples. The capacity of the BSA/anti-CEA/PPI/GO/GCE to determine CEA in human blood was studied.
Collapse
Affiliation(s)
- Chenyu Tao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, Hebei, China.
| | - Jalal Rouhi
- Faculty of Physics, University of Tabriz, Tabriz, 51566, Iran.
| |
Collapse
|
13
|
Saeedifar AM, Ghazavi A, Mosayebi G, Ganji A. Synergistic apoptotic effects of ethanolic extracts of ginger and Ganoderma lucidum in a colorectal cancer cell line. Biotech Histochem 2023; 98:353-359. [PMID: 36970755 DOI: 10.1080/10520295.2023.2190620] [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: 03/29/2023] Open
Abstract
Current conventional therapy for colorectal cancer includes surgery, radiation and chemotherapy, all of which produce side effects. Herbal medicine can control the side effects of conventional treatments. We investigated the synergistic effect of a mixture of Zingiber officinale Roscoe (Ginger) and Ganoderma lucidum extracts on colorectal cancer cell apoptosis in vitro. We prepared ethanolic extracts of ginger (GEE) and G. lucidum (GLEE). Cytotoxicity was evaluated using MTT assay and the half-maximal inhibitory concentration (IC50) of each extract was calculated. The effect of these extracts on apoptosis in cancer cells was assessed using flow cytometry; Bax, Bcl2 and caspase-3 gene expression was evaluated using real-time PCR. GEE and GLEE decreased CT-26 cell viability significantly in a dose-dependent manner; however, the combined application of GEE + GLEE was most effective. Bax:Bcl-2 gene expression ratio, caspase-3 gene expression and the number of apoptotic cells were increased significantly in CT-26 cells treated at the IC50 level of each compound, especially in the GEE + GLEE treatment group. Combined ginger and Ganoderma lucidum extracts exhibited synergistic antiproliferative and apoptotic effects on colorectal cancer cells.
Collapse
Affiliation(s)
- Amir Mohammad Saeedifar
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ghazavi
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Traditional and Complementary Medicine Research Center (TCMRC), Arak University of Medical Sciences, Arak, Iran
| | - Ghasem Mosayebi
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ganji
- Department of Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
14
|
Daneshvar S, Zamanian MY, Ivraghi MS, Golmohammadi M, Modanloo M, Kamiab Z, Pourhosseini SME, Heidari M, Bazmandegan G. A comprehensive view on the apigenin impact on colorectal cancer: Focusing on cellular and molecular mechanisms. Food Sci Nutr 2023; 11:6789-6801. [PMID: 37970406 PMCID: PMC10630840 DOI: 10.1002/fsn3.3645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 11/17/2023] Open
Abstract
Colon cancer (CC) is one of the most common and deadly cancers worldwide. Oncologists are facing challenges such as development of drug resistance and lack of suitable drug options for CC treatment. Flavonoids are a group of natural compounds found in fruits, vegetables, and other plant-based foods. According to research, they have a potential role in the prevention and treatment of cancer. Apigenin is a flavonoid that is present in many fruits and vegetables. It has been used as a natural antioxidant for a long time and has been considered due to its anticancer effects and low toxicity. The results of this review study show that apigenin has potential anticancer effects on CC cells through various mechanisms. In this comprehensive review, we present the cellular targets and signaling pathways of apigenin indicated to date in in vivo and in vitro CC models. Among the most important modulated pathways, Wnt/β-catenin, PI3K/AKT/mTOR, MAPK/ERK, JNK, STAT3, Bcl-xL and Mcl-1, PKM2, and NF-kB have been described. Furthermore, apigenin suppresses the cell cycle in G2/M phase in CC cells. In CC cells, apigenin-induced apoptosis is increased by inhibiting the formation of autophagy. According to the results of this study, apigenin appears to have the potential to be a promising agent for CC therapy, but more research is required in the field of pharmacology and pharmacokinetics to establish the apigenin effects and its dosage for clinical studies.
Collapse
Affiliation(s)
- Siamak Daneshvar
- Department of General SurgerySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Yasin Zamanian
- Department of PhysiologySchool of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and ToxicologySchool of PharmacyHamadan University of Medical SciencesHamadanIran
| | | | | | - Mona Modanloo
- Pharmaceutical Sciences Research CenterMazandaran University of Medical SciencesSariIran
| | - Zahra Kamiab
- Clinical Research Development UnitAli‐Ibn Abi‐Talib HospitalRafsanjan University of Medical SciencesRafsanjanIran
- Department of Community MedicineSchool of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Seyed Mohammad Ebrahim Pourhosseini
- Non‐Communicable Diseases Research CenterRafsanjan University of Medical SciencesRafsanjanIran
- Department of Internal MedicineSchool of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mahsa Heidari
- Department of BiochemistryInstitute of Biochemistry and Biophysics (IBB)University of TehranTehranIran
| | - Gholamreza Bazmandegan
- Physiology‐Pharmacology Research CenterResearch Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
- Department of Physiology and PharmacologySchool of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
15
|
Postwala H, Shah Y, Parekh PS, Chorawala MR. Unveiling the genetic and epigenetic landscape of colorectal cancer: new insights into pathogenic pathways. Med Oncol 2023; 40:334. [PMID: 37855910 DOI: 10.1007/s12032-023-02201-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
Colorectal cancer (CRC) is a complex disease characterized by genetic and epigenetic alterations, playing a crucial role in its development and progression. This review aims to provide insights into the emerging landscape of these alterations in CRC pathogenesis to develop effective diagnostic tools and targeted therapies. Genetic alterations in signaling pathways such as Wnt/β-catenin, and PI3K/Akt/mTOR are pivotal in CRC development. Genetic profiling has identified distinct molecular subtypes, enabling personalized treatment strategies. Epigenetic modifications, including DNA methylation and histone modifications, also contribute to CRC pathogenesis by influencing critical cellular processes through gene silencing or activation. Non-coding RNAs have emerged as essential players in epigenetic regulation and CRC progression. Recent research highlights the interplay between genetic and epigenetic alterations in CRC. Genetic mutations can affect epigenetic modifications, leading to dysregulated gene expression and signaling cascades. Conversely, epigenetic changes can modulate genetic expression, amplifying or dampening the effects of genetic alterations. Advancements in understanding pathogenic pathways have potential clinical applications. Identifying genetic and epigenetic markers as diagnostic and prognostic biomarkers promises more accurate risk assessment and early detection. Challenges remain, including validating biomarkers and developing robust therapeutic strategies through extensive research and clinical trials. The dynamic nature of genetic and epigenetic alterations necessitates a comprehensive understanding of their temporal and spatial patterns during CRC progression. In conclusion, the genetic and epigenetic landscape of CRC is increasingly being unraveled, providing valuable insights into its pathogenesis. Integrating genetic and epigenetic knowledge holds great potential for improving diagnostics, prognostics, and personalized therapies in CRC. Continued research efforts are vital to translate these findings into clinical practice, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Humzah Postwala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Yesha Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, Florida, 32211, USA
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, 380009, Gujarat, India.
| |
Collapse
|
16
|
Deng X, Yang J, Zhang Y, Chen X, Wang C, Suo H, Song J. An Update on the Pivotal Roles of Probiotics, Their Components, and Metabolites in Preventing Colon Cancer. Foods 2023; 12:3706. [PMID: 37835359 PMCID: PMC10572180 DOI: 10.3390/foods12193706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/01/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023] Open
Abstract
Diet, lifestyle, and gut microbiota composition are key risk factors for the progression of colon cancer. Probiotics are living microorganisms that can offer health benefits to the parasitifer when ingested in competent quantities. Several in vivo, in vitro, and clinical studies have demonstrated that probiotics can prevent and mitigate the development of colon cancer. The anti-colon cancer mechanisms of probiotics include the suppression of cell proliferation and the promotion of cancer cell apoptosis, immunomodulation, the modulation of intestinal microorganisms and their metabolism, strengthening the intestinal barrier, and antioxidant effects. This article describes the pathogenesis of colon cancer and the available therapeutic options. In addition, this paper reviews the mechanisms by which probiotics mitigate colon cancer as well as the mitigating effects of probiotic components and metabolites on colon cancer.
Collapse
Affiliation(s)
- Xue Deng
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
| | - Jing Yang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing 400067, China;
| | - Yu Zhang
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
| | - Xiaoyong Chen
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
- National Citrus Engineering Research Center, Southwest University, Chongqing 400712, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, China; (X.D.); (Y.Z.); (X.C.); (C.W.); (H.S.)
| |
Collapse
|
17
|
Alrosan AZ, Alrosan K, Heilat GB, Alsharedeh R, Abudalo R, Oqal M, Alqudah A, Elmaghrabi YA. Potential roles of NEDD4 and NEDD4L and their utility as therapeutic targets in high‑incidence adult male cancers (Review). Mol Clin Oncol 2023; 19:68. [PMID: 37614371 PMCID: PMC10442760 DOI: 10.3892/mco.2023.2664] [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: 02/18/2023] [Accepted: 07/07/2023] [Indexed: 08/25/2023] Open
Abstract
The term 'cancer' refers to >100 disorders that progressively manifest over time and are characterized by uncontrolled cell division. Although malignant growth can occur in virtually any human tissue, the underlying mechanisms underlying all forms of cancer are consistent. The International Agency for Research on Cancer's annual GLOBOCAN 2020 report provided an update on the global cancer incidence and mortality. Excluding non-melanoma skin cancer, the report predicts that there will be 19.3 million new cancer cases and >10 million cancer-related fatalities in 2023. Lung, prostate, and colon cancers are the most prevalent and lethal cancers in males. It was recognized that post-translational modifications (PTMs) of proteins are necessary for almost all cellular biological processes, as well as in cancer development and metastasis to other bodily organs. Thus, PTMs have a considerable impact on how proteins behave. Various PTMs may have harmful roles by affecting the hallmarks of cancer, metabolism and the regulation of the tumor microenvironment. PTMs and genetic changes/mutations are essential in carcinogenesis and cancer development. A pivotal PTM mechanism is protein ubiquitination. Of note, the rate-limiting stage of the protein ubiquitination cascade is hypothesized to be E3-ligase-mediated ubiquitination. Numerous studies revealed that the neural precursor cell expressed developmentally downregulated protein 4 (NEDD4) E3 ligase is among the E3 ubiquitin ligases that have essential roles in cellular processes. It regulates protein degradation and substrate ubiquitination. In addition, it has been shown that NEDD4 primarily functions as an oncogene in various malignancies but can also act as a tumor suppressor in certain types of tumor. In the present review, the roles of NEDD4 as an anticancer protein in various high-incidence male malignancies and the significance of NEDD4 as a potential cancer therapeutic target are discussed. In addition, the targeting of NEDD4 as a therapeutic strategy for the treatment of human malignancies is explored.
Collapse
Affiliation(s)
- Amjad Z. Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Khaled Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Ghaith B. Heilat
- Department of General Surgery and Urology, Faculty of Medicine, The Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Rawan Alsharedeh
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The Yarmouk University, Irbid 21163, Jordan
| | - Rawan Abudalo
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Muna Oqal
- Department of Pharmaceutical Technology, Faculty of Pharmacy, The Hashemite University, Zarqa 13133, Jordan
| | - Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | | |
Collapse
|
18
|
Ibrahim MAA, Abdeljawaad KAA, Abdelrahman AHM, Abdelhamid MMH, Naeem MA, Mekhemer GAH, Sidhom PA, Sayed SRM, Paré PW, Hegazy MEF. SuperNatural inhibitors to reverse multidrug resistance emerged by ABCB1 transporter: Database mining, lipid-mediated molecular dynamics, and pharmacokinetics study. PLoS One 2023; 18:e0288919. [PMID: 37494356 PMCID: PMC10370898 DOI: 10.1371/journal.pone.0288919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
An effective approach to reverse multidrug resistance (MDR) is P-glycoprotein (P-gp, ABCB1) transport inhibition. To identify such molecular regulators, the SuperNatural II database, which comprises > 326,000 compounds, was virtually screened for ABCB1 transporter inhibitors. The Lipinski rule was utilized to initially screen the SuperNatural II database, identifying 128,126 compounds. Those natural compounds were docked against the ABCB1 transporter, and those with docking scores less than zosuquidar (ZQU) inhibitor were subjected to molecular dynamics (MD) simulations. Based on MM-GBA binding energy (ΔGbinding) estimations, UMHSN00009999 and UMHSN00097206 demonstrated ΔGbinding values of -68.3 and -64.1 kcal/mol, respectively, compared to ZQU with a ΔGbinding value of -49.8 kcal/mol. For an investigation of stability, structural and energetic analyses for UMHSN00009999- and UMHSN00097206-ABCB1 complexes were performed and proved the high steadiness of these complexes throughout 100 ns MD simulations. Pharmacokinetic properties of the identified compounds were also predicted. To mimic the physiological conditions, MD simulations in POPC membrane surroundings were applied to the UMHSN00009999- and UMHSN00097206-ABCB1 complexes. These results demonstrated that UMHSN00009999 and UMHSN00097206 are promising ABCB1 inhibitors for reversing MDR in cancer and warrant additional in-vitro/in-vivo studies.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
- School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Mahmoud M H Abdelhamid
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Mohamed Ahmed Naeem
- Nutrition and Food Sciences, Ain Shams University Specialized Hospital, Ain Shams University, Cairo, Egypt
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Shaban R M Sayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Paul W Paré
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | | |
Collapse
|
19
|
Morishita A, Oura K, Tadokoro T, Shi T, Fujita K, Tani J, Atsukawa M, Masaki T. Galectin-9 in Gastroenterological Cancer. Int J Mol Sci 2023; 24:ijms24076174. [PMID: 37047155 PMCID: PMC10094448 DOI: 10.3390/ijms24076174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Immunochemotherapy has become popular in recent years. The detailed mechanisms of cancer immunity are being elucidated, and new developments are expected in the future. Apoptosis allows tissues to maintain their form, quantity, and function by eliminating excess or abnormal cells. When apoptosis is inhibited, the balance between cell division and death is disrupted and tissue homeostasis is impaired. This leads to dysfunction and the accumulation of genetically abnormal cells, which can contribute to carcinogenesis. Lectins are neither enzymes nor antibodies but proteins that bind sugar chains. Among soluble endogenous lectins, galectins interact with cell surface sugar chains outside the cell to regulate signal transduction and cell growth. On the other hand, intracellular lectins are present at the plasma membrane and regulate signal transduction by regulating receptor–ligand interactions. Galectin-9 expressed on the surface of thymocytes induces apoptosis of T lymphocytes and plays an essential role in immune self-tolerance by negative selection in the thymus. Furthermore, the administration of extracellular galectin-9 induces apoptosis of human cancer and immunodeficient cells. However, the detailed pharmacokinetics of galectin-9 in vivo have not been elucidated. In addition, the cell surface receptors involved in galectin-9-induced apoptosis of cancer cells have not been identified, and the intracellular pathways involved in apoptosis have not been fully investigated. We have previously reported that galectin-9 induces apoptosis in various gastrointestinal cancers and suppresses tumor growth. However, the mechanism of galectin-9 and apoptosis induction in gastrointestinal cancers and the detailed mechanisms involved in tumor growth inhibition remain unknown. In this article, we review the effects of galectin-9 on gastrointestinal cancers and its mechanisms.
Collapse
|
20
|
Manoharan JP, Nirmala Karunakaran K, Vidyalakshmi S, Dhananjayan K. Computational binding affinity and molecular dynamic characterization of annonaceous acetogenins at nucleotide binding domain (NBD) of multi-drug resistance ATP-binding cassette sub-family B member 1 (ABCB1). J Biomol Struct Dyn 2023; 41:821-832. [PMID: 34907862 DOI: 10.1080/07391102.2021.2013321] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multi drug resistance (MDR) in tumor might be caused leading to the overexpression of transporters, such as ATP-binding cassette sub-family B member 1 (ABCB1). A combination of non-toxic and potent ABC inhibitors along with conventional anti-cancer drugs is needed to reverse MDR in tumors. A variety of phytochemicals have been previously shown to reverse MDR. Annonaceous acetogenins (AAs) with C35/C37 long-chain fatty acids were reported for their anti-tumor activity, however, their effect on reversing MDR is not yet investigated. We aimed to investigate some selective AAs against the B1 subtype of ABC transporter using computational studies. Various modules of Maestro software were utilized for our in-silico analysis. Few well-characterized AAs were screened for their drug-likeness properties and tested for binding affinity at ATP and drug binding sites of ABCB1 through molecular docking. The stability of the ligand-protein complex (lowest docking score) was then determined by a molecular dynamic (MD) simulation study. Out of 24 AAs, Annonacin A (-8.10 kcal/mol) and Annohexocin (-10.49 kcal/mol) docked with a greater binding affinity at the ATP binding site than the first-generation inhibitor of ABCB1 (Verapamil: -3.86 kcal/mol). MD simulation of Annonacin A: ABCB1 complex for 100 ns also indicated that Annonacin A would stably bind to the ATP binding site. We report that Annonacin A binds at a greater affinity with ABCB1 and might act as a potential drug lead to reverse MDR in tumor cells. Communicated by Ramaswamy H. Sarma.
Collapse
|
21
|
3-Substituted-2,3-Dihydrothiazole as a promising scaffold to design EGFR inhibitors. Bioorg Chem 2022; 129:106172. [DOI: 10.1016/j.bioorg.2022.106172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/31/2022] [Accepted: 09/18/2022] [Indexed: 11/21/2022]
|
22
|
Zhang Q, Kandasamy K, Alyami NM, Alyami HM, Natarajan N, Elayappan PK. Influence of Padina gymnospora on Apoptotic Proteins of Oral Cancer Cells-a Proteome-Wide Analysis. Appl Biochem Biotechnol 2022; 194:5945-5962. [PMID: 35849254 DOI: 10.1007/s12010-022-04045-w] [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] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
Abstract
Oral carcinoma is one of the most vicious forms of cancer with a very low survival rate, as its patients often respond poorly to conventional chemotherapy. Presently several researchers are attempting to pursue an alternative to this therapy using natural products. Considering the promising strategy and induction of apoptosis to target the cancer cells, we evaluated the influence of a seaweed Padina gymnospora (15 µg/ml and 20 µg/ml) in enhancing apoptosis of oral cancer cells (KB-CHR-8-5) after 24-h incubation. The morphological changes indicating apoptosis were primarily assessed using a light microscope after which the apoptosis was confirmed by performing AO/EB staining method. Subsequently, MMP and ROS levels in the cells were assessed using Rh 123 and DCFH-DA staining procedures, respectively. All the above tests confirmed the ability of P. gymnospora to accelerate apoptosis in the oral cancer cells. As a next step, wide proteome analysis was performed where the proteins from P. gymnospora-treated cells were separated using the 2D electrophoresis technique and compared with that of control cells to isolate the differentially expressed proteins. This procedure resulted in the isolation of 10 proteins which were identified using MALDI-TOF/TOF MS, which established that most of the isolated proteins were part of the apoptotic process of the cell. The proteins identified are part of huge and complex pathways where it gets linked with many more genes which are also associated with apoptosis. Bioinformatics of these identified proteins was analyzed using STRING and PANTHER databases. These proteins contribute to cell apoptosis by affecting various functions, biological processes, and the synthesis of cellular components. PANTHER also demonstrated that these proteins belong to the classes of proteins that take part in several vital pathways of the cell among which the apoptotic pathway is the predominant one.
Collapse
Affiliation(s)
- Qian Zhang
- School of Stomatology, QiLu Medical University, No.1678, Renmin West Road, Zibo City, 255300, China
| | - Kavitha Kandasamy
- Department of Biochemistry, Vivekanandha College of Arts and Sciences for Women (Autonomous), Elayampalayam, Tiruchengode, Namakkal, 637205, India
| | - Nouf M Alyami
- Department of Zoology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Hanadi M Alyami
- Dental Administration, King Fahad Medical City, Riyadh, 11451, Saudi Arabia
| | - Nandakumar Natarajan
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Poorni Kaliyappan Elayappan
- Department of Biochemistry, Vivekanandha College of Arts and Sciences for Women (Autonomous), Elayampalayam, Tiruchengode, Namakkal, 637205, India.
| |
Collapse
|
23
|
Gu B, Wang B, Li X, Feng Z, Ma C, Gao L, Yu Y, Zhang J, Zheng P, Wang Y, Li H, Zhang T, Chen H. Photodynamic therapy improves the clinical efficacy of advanced colorectal cancer and recruits immune cells into the tumor immune microenvironment. Front Immunol 2022; 13:1050421. [PMID: 36466825 PMCID: PMC9716470 DOI: 10.3389/fimmu.2022.1050421] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/02/2022] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVE Although photodynamic therapy (PDT) has been proven effective in various tumors, it has not been widely used as a routine treatment for colorectal cancer (CRC), and the characteristics of changes in the tumor microenvironment (TME) after PDT have not been fully elucidated. This study evaluated the efficacy of PDT in patients with advanced CRC and the changes in systemic and local immune function after PDT. METHODS Patients with stage III-IV CRC diagnosed in our hospital from November 2020 to July 2021 were retrospectively analyzed to compare the survival outcomes among each group. Subsequently, short-term efficacy, systemic and local immune function changes, and adverse reactions were assessed in CRC patients treated with PDT. RESULTS A total of 52 CRC patients were enrolled in this retrospective study from November 2020 to July 2021, and the follow-up period ended in March 2022. The overall survival (OS) of the PDT group was significantly longer than that of the non-PDT group (p=0.006). The objective response rate (ORR) and disease control rate two months after PDT were 44.4% and 88.9%, respectively. Differentiation degree (p=0.020) and necrosis (p=0.039) are two crucial factors affecting the short-term efficacy of PDT. The systemic immune function of stage III patients after PDT decreased, whereas that of stage IV patients increased. Local infiltration of various immune cells such as CD3+ T cells, CD4+ T cells, CD8+ T cells, CD20+ B cells and macrophages in the tumor tissue were significantly increased. No severe adverse reactions associated with PDT were observed. CONCLUSION PDT is effective for CRC without significant side effects according to the available data. It alters the TME by recruiting immune cells into tumor tissues.
Collapse
Affiliation(s)
- Baohong Gu
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Bofang Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xuemei Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Zedong Feng
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Chenhui Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Lei Gao
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yang Yu
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Peng Zheng
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yunpeng Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Haiyuan Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Tao Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hao Chen
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou, Gansu, China
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
24
|
Kumar A, Singh AK, Singh H, Thareja S, Kumar P. Regulation of thymidylate synthase: an approach to overcome 5-FU resistance in colorectal cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:3. [PMID: 36308643 DOI: 10.1007/s12032-022-01864-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/29/2022] [Indexed: 01/17/2023]
Abstract
Thymidylate synthase is the rate-limiting enzyme required for DNA synthesis and overexpression of this enzyme causes resistance to cancer cells. Long treatments with 5-FU cause resistance to Thymidylate synthase targeting drugs. We have also compiled different mechanisms of drug resistance including autophagy and apoptosis, drug detoxification and ABC transporters, drug efflux, signaling pathways (AKT/PI3K, RAS-MAPK, WNT/β catenin, mTOR, NFKB, and Notch1 and FOXM1) and different genes associated with resistance in colorectal cancer. We can overcome 5-FU resistance in cancer cells by regulating thymidylate synthase by natural products (Coptidis rhizoma), HDAC inhibitors, mTOR inhibitors, Folate antagonists, and several other drugs which have been used in combination with TS inhibitors. This review is a compilation of different approaches reported for the regulation of thymidylate synthase to overcome resistance in colorectal cancer cells.
Collapse
Affiliation(s)
- Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India.
| |
Collapse
|
25
|
Beni FA, Kazemi M, Dianat-Moghadam H, Behjati M. MicroRNAs regulating Wnt signaling pathway in colorectal cancer: biological implications and clinical potentials. Funct Integr Genomics 2022; 22:1073-1088. [DOI: 10.1007/s10142-022-00908-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
|
26
|
In Vitro Cytotoxic Activity of Total Flavonoid from Equisetum Arvense Extract. Rep Biochem Mol Biol 2022; 11:487-492. [PMID: 36718311 PMCID: PMC9883023 DOI: 10.52547/rbmb.11.3.487] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 04/24/2022] [Indexed: 01/17/2023]
Abstract
Background Normally happening substances like flavonoids are regarded as active candidates for the treatment and prevention of cancer The purpose of this study was to see how Iraqi E. arvense total flavonoid affected cell lines biologically and human lung fibroblast normal cell line (WISH). Methods Plant powder was extracted by reflex apparatus, then thin-layer chromatography (TLC) was used to determine total flavonoids. Cytotoxicity assay (MTT) was used to determine the cytotoxic activity of the prepared plant against human breast cancer (MCF-7), cells human cervix cancer (HELA), human colon cancer (Caco-2) and human lung fibroblast normal cell line (WISH). Results The flavonoids Rutin, Quercetin, Kaempferol, and luteolin were detected using the Thin Layer Chromatography (TLC) technique. In contrast to the negative control, the extract inhibited cell growth to a highest of 82.158% for MCF-7 and 61.360% for Caco-2 at the concentration (100 µg/ml), and (54.880%) for Hela cell line at the concentration (100 µg/ml). In addition, the concentration (6.25 µg/ml) of total flavonoid extract produced a decrease in the growth of the normal WISH cell line to reach (1.094%). Conclusion Equisetum arvense contain high amounts of flavonoids, the qualification of some flavonoids compounds was detected using TLC. The total flavonoids showed significant cytotoxic activity against various types of cancer cell lines and normal cell line in vitro, the antitumor activity was highly efficient in a dose and cell type dependent manner.
Collapse
|
27
|
Kumar P, Mishra J, Kumar N. Mechanistic Role of Jak3 in Obesity-Associated Cognitive Impairments. Nutrients 2022; 14:nu14183715. [PMID: 36145091 PMCID: PMC9505565 DOI: 10.3390/nu14183715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aims: A compromise in intestinal mucosal functions is associated with several chronic inflammatory diseases. Previously, we reported that obese humans have a reduced expression of intestinal Janus kinase-3 (Jak3), a non-receptor tyrosine kinase, and a deficiency of Jak3 in mice led to predisposition to obesity-associated metabolic syndrome. Since meta-analyses show cognitive impairment as co-morbidity of obesity, the present study demonstrates the mechanistic role of Jak3 in obesity associated cognitive impairment. Our data show that high-fat diet (HFD) suppresses Jak3 expression both in intestinal mucosa and in the brain of wild-type mice. Methodology: Recapitulating these conditions using global (Jak3-KO) and intestinal epithelial cell-specific conditional (IEC-Jak3-KO) mice and using cognitive testing, western analysis, flow cytometry, immunofluorescence microscopy and 16s rRNA sequencing, we demonstrate that HFD-induced Jak3 deficiency is responsible for cognitive impairments in mice, and these are, in part, specifically due to intestinal epithelial deficiency of Jak3. Results: We reveal that Jak3 deficiency leads to gut dysbiosis, compromised TREM-2-functions-mediated activation of microglial cells, increased TLR-4 expression and HIF1-α-mediated inflammation in the brain. Together, these lead to compromised microglial-functions-mediated increased deposition of β-amyloid (Aβ) and hyperphosphorylated Tau (pTau), which are responsible for cognitive impairments. Collectively, these data illustrate how the drivers of obesity promote cognitive impairment and demonstrate the underlying mechanism where HFD-mediated impact on IEC-Jak3 deficiency is responsible for Jak3 deficiency in the brain, reduced microglial TREM2 expression, microglial activation and compromised clearance of Aβ and pTau as the mechanism during obesity-associated cognitive impairments. Conclusion: Thus, we not only demonstrate the mechanism of obesity-associated cognitive impairments but also characterize the tissue-specific role of Jak3 in such conditions through mucosal tolerance, gut–brain axis and regulation of microglial functions.
Collapse
|
28
|
Erdogan CS, Al Hassadi Y, Aru B, Yilmaz B, Gemici B. Combinatorial effects of melatonin and paclitaxel differ depending on the treatment scheme in colorectal cancer in vitro. Life Sci 2022; 308:120927. [PMID: 36063977 DOI: 10.1016/j.lfs.2022.120927] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
AIMS Colorectal carcinoma (CRC) is the third most prevalent cancer with high mortality. Besides regulating the circadian rhythm, melatonin (MTN) exerts anticancer activities. Paclitaxel (PTX) is successful against different malignancies, however, acquired resistance and variability in patient response restrict its use. mTOR and MAPK pathways are often deregulated in human cancers. We aimed to investigate whether MTN enhances or sensitizes the chemotherapeutic activity of PTX and if so, determine the underlying possible mechanisms in CRC in vitro. MAIN METHODS Antiproliferative and cytotoxic activities of PTX and MTN were assessed alone and in combination, as well as with different treatment regimens (renewal or replacement of the treatment after 24 h), up to 48 h. Apoptosis, viability and autophagy were assessed by flow cytometry. mTOR and MAPK pathway activities were investigated by immunoblotting. KEY FINDINGS Both drugs reduced cell viability in a dose-dependent manner at 24 and 48 h. Only the highest dose of MTN (500 μM) potentiated the cytotoxicity of PTX (50 nM). Replacement of PTX after 24 h with MTN was superior in reducing cell viability than vice versa via apoptosis induction. Renewal of MTN treatment every 24 h reduced autophagy compared to the control group, while other treatments did not alter the autophagic activity. A 24 h MTN treatment followed by 24 h PTX treatment increased S6 phosphorylation in a mTOR-independent manner and increased Erk1/2 phosphorylation. SIGNIFICANCE The present study suggests that sequential treatment with MTN and PTX distinctly affect apoptosis and cytotoxicity via regulating mTOR and MAPK pathways differentially in CRC.
Collapse
Affiliation(s)
- Cihan Suleyman Erdogan
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Yasmine Al Hassadi
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Basak Aru
- Yeditepe University, Faculty of Medicine, Department of Immunology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Bayram Yilmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Burcu Gemici
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey.
| |
Collapse
|
29
|
Sun M, Chen X, Yang Z. Single cell mass spectrometry studies reveal metabolomic features and potential mechanisms of drug-resistant cancer cell lines. Anal Chim Acta 2022; 1206:339761. [PMID: 35473873 PMCID: PMC9046687 DOI: 10.1016/j.aca.2022.339761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/28/2022]
Abstract
Irinotecan (Iri) is a key drug to treat metastatic colorectal cancer, but its clinical activity is often limited by de novo and acquired drug resistance. Studying the underlying mechanisms of drug resistance is necessary for developing novel therapeutic strategies. In this study, we used both regular and irinotecan-resistant (Iri-resistant) colorectal cell lines as models, and performed single cell mass spectrometry (SCMS) metabolomics studies combined with analyses from cytotoxicity assay, western blot, flow cytometry, quantitative real-time polymerase chain reaction (qPCR), and reactive oxygen species (ROS). Our SCMS results indicate that Iri-resistant cancer cells possess higher levels of unsaturated lipids compared with the regular cancer cells. In addition, multiple protein biomarkers and their corresponding mRNAs of colon cancer stem cells are overexpressed in Iri-resistance cells. Particularly, stearoyl-CoA desaturase 1 (SCD1) is upregulated with the development of drug resistance in Iri-resistant cells, whereas inhibiting the activity of SCD1 efficiently increase their sensitivity to Iri treatment. In addition, we demonstrated that SCD1 directly regulates the expression of ALDH1A1, which contributes to the cancer stemness and ROS level in Iri-resistant cell lines.
Collapse
|
30
|
Sevimli M, Bayram D, Özgöçmen M, Armağan I, Semerci Sevimli T. Boric acid suppresses cell proliferation by TNF signaling pathway mediated apoptosis in SW-480 human colon cancer line. J Trace Elem Med Biol 2022; 71:126958. [PMID: 35219976 DOI: 10.1016/j.jtemb.2022.126958] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/18/2022] [Indexed: 12/26/2022]
Abstract
BACKGROUND/AIM Colon cancer is one of the most common cancers. Treatment success and survival rates are not high enough with current approaches. Therefore, there is a need to develop new agents and treatment methods. Boric acid is the most frequently observed form of boron. Some epidemiological data suggest that environmental exposure to boric acid reduces the incidence of prostate cancer in men, cervical and lung cancers in women. Experimental studies show, boric acid reduces cell proliferation and stimulates apoptosis in some prostate, melanoma, breast cancer cell lines. In this study, it was investigated whether boric acid could be a new candidate molecule that could be used in the treatment of colon cancer. MATERIALS AND METHODS The effects of boric acid on human colon adenocarcinoma cell line SW-480 were investigated with BrdU, TUNEL, Caspase-3, and AIF immunohistochemical studies in both 2D and 3D culture systems. In addition, a qRT-PCR study was carried out to determine the expression changes in key genes that take part in apoptosis. RESULTS We observed that boric acid suppresses cell proliferation and induces apoptosis both in 2D and 3D culture conditions. In addition, as a result of qRt-PCR studies, it was revealed that the observed apoptotic process was related to the TNF signaling pathway. CONCLUSION Boric acid can be considered as a potential anti-cancer agent candidate for colon cancer treatment. DATA AVAILABILITY All data generated or analyzed during this study are included in this published article.
Collapse
Affiliation(s)
- Murat Sevimli
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey.
| | - Dilek Bayram
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey
| | - Meltem Özgöçmen
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey
| | - Ilkay Armağan
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey
| | - Tuğba Semerci Sevimli
- Eskisehir Osmangazi University, Institue of Health Sciences, Department of Stem Cell, Eskisehir, Turkey; Eskisehir Osmangazi University, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir, Turkey
| |
Collapse
|
31
|
Yangnok K, Innajak S, Sawasjirakij R, Mahabusarakam W, Watanapokasin R. Effects of Artonin E on Cell Growth Inhibition and Apoptosis Induction in Colon Cancer LoVo and HCT116 Cells. Molecules 2022; 27:molecules27072095. [PMID: 35408492 PMCID: PMC9000836 DOI: 10.3390/molecules27072095] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Today, colon cancer is the leading cause of cancer death. In Thailand, colon cancer is the third most common cancer in men and the second in women. Currently, the treatments for colon cancer include surgery, chemotherapy, radiation therapy, immunotherapy, hormone therapy, targeted drug therapy, and stem cell therapy. However, some treatments have side effects for cancer patients, causing unwanted symptoms. In addition, targeted therapy comes with a high cost for patients. Therefore, bioactive compounds might be a good choice for colon cancer treatment. In this study, we investigated the effect of artonin E on apoptosis induction in colon cancer LoVo and HCT116 cells. The concentration ranges of artonin E at 3, 5, 10, and 30 µg/mL in LoVo cells and 1, 1.5, 2, and 3 µg/mL in HCT116 cells were examined. The results implied that artonin E decreased cell viability and increased apoptotic cells in a dose-dependent manner. In addition, artonin E stimulated mitochondrial membrane potential (ΔΨm) changes associated with apoptosis by increasing the sub-G1 population analyzed by flow cytometry. Western blotting showed that artonin E increased the proapoptotic protein, Bax, and decreased anti-apoptotic proteins’ (Bcl-2 and Bcl-x) expression. Moreover, artonin E also increased cleaved caspase-7 and cleaved-PARP expression in both LoVo and HCT116 cells. Interestingly, artonin E induced apoptosis through p-ERK1/2, p-p38/p38, and p-c-Jun expression in both cells. Our results suggested that artonin E induced apoptosis via caspase activation associated with the MAPKs signaling pathway. Therefore, artonin E might be used as a potential anticancer drug for colon cancer in the future.
Collapse
Affiliation(s)
- Kanyaluck Yangnok
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (K.Y.); (S.I.)
| | - Sukanda Innajak
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (K.Y.); (S.I.)
| | - Ratchawin Sawasjirakij
- Faculty of Medicine, Medical University of Lublin, Aleje Racławickie 1, 20-059 Lublin, Poland;
| | - Wilawan Mahabusarakam
- Department of Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai 90112, Thailand;
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (K.Y.); (S.I.)
- Correspondence: ; Tel.: +66-082-479-7824
| |
Collapse
|
32
|
Park YL, Kim HP, Ock CY, Min DW, Kang JK, Lim YJ, Song SH, Han SW, Kim TY. EMT-mediated regulation of CXCL1/5 for resistance to anti-EGFR therapy in colorectal cancer. Oncogene 2022; 41:2026-2038. [PMID: 35173310 DOI: 10.1038/s41388-021-01920-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 05/08/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The emergence of RAS/RAF mutant clone is the main feature of EGFR inhibitor resistance in KRAS wild-type colon cancer. However, its molecular mechanism is thought to be multifactorial, mainly due to cellular heterogeneity. In order to better understand the resistance mechanism in a single clone level, we successfully isolated nine cells with cetuximab-resistant (CR) clonality from in vitro system. All CR cells harbored either KRAS or BRAF mutations. Characteristically, these cells showed a higher EMT (Epithelial to mesenchymal transition) signature, showing increased EMT markers such as SNAI2. Moreover, the expression level of CXCL1/5, a secreted protein, was significantly higher in CR cells compared to the parental cells. In these CR cells, CXCL1/5 expression was coordinately regulated by SNAI2/NFKB and transactivated EGFR through CXCR/MMPI/EGF axis via autocrine singling. We also observed that combined cetuximab/MEK inhibitor not only showed growth inhibition but also reduced the secreted amounts of CXCL1/5. We further found that serum CXCL1/5 level was positively correlated with the presence of RAS/RAF mutation in colon cancer patients during cetuximab therapy, suggesting its role as a biomarker. These data indicated that the application of serum CXCL1/5 could be a potential serologic biomarker for predicting resistance to EGFR therapy in colorectal cancer.
Collapse
Affiliation(s)
- Ye-Lim Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea. .,Cancer Research Institute, Seoul National University, Seoul, Korea.
| | - Hwang-Phill Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea.,IMBDx Inc, Seoul, Korea
| | - Chan-Young Ock
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong-Wook Min
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jun Kyu Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Yoo Joo Lim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sang-Hyun Song
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Sae-Won Han
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tae-You Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea. .,Cancer Research Institute, Seoul National University, Seoul, Korea. .,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
33
|
Enhancing the anticancer effect of paclitaxel by using polymeric nanoparticles decorated with colorectal cancer targeting CPKSNNGVC-peptide. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
34
|
Rizk NI, Abulsoud AI, Kamal MM, Kassem DH, Hamdy NM. Exosomal-long non-coding RNAs journey in colorectal cancer: Evil and goodness faces of key players. Life Sci 2022; 292:120325. [PMID: 35031258 DOI: 10.1016/j.lfs.2022.120325] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023]
Abstract
Exosomes are nano-vesicles (NVs) secreted by cells and take part in cell-cell communications. Lately, these exosomes were proved to have dual faces in cancer. Actually, they can contribute to carcinogenesis through epithelial-mesenchymal transition (EMT), angiogenesis, metastasis and tumor microenvironment (TME) of various cancers, including colorectal cancer (CRC). On the other hand, they can be potential targets for cancer treatment. CRC is one of the most frequent tumors worldwide, with incidence rates rising in the recent decades. In its early stage, CRC is asymptomatic with poor treatment outcomes. Therefore, finding a non-invasive, early diagnostic biomarker tool and/or suitable defender to combat CRC is mandatory. Exosomes provide enrichment and safe setting for their cargos non-coding RNAs (ncRNAs) and proteins, whose expression levels can be upregulated ordown-regulated in cancer. Hence, exosomes can be used as diagnostic and/or prognostic tools for cancer. Moreover, exosomes can provide a novel potential therapeutic modality for tumors via loading with specific chemotherapeutic agents, with the advantage of possible tumor targeting. In this review, we will try to collect and address recent studies concerned with exosomes and their cargos' implications for CRC diagnosis and/or hopefully, treatment.
Collapse
Affiliation(s)
- Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy (Boys Branch), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohamed M Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt; The Centre for Drug Research and Development, Faculty of Pharmacy, BUE, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
35
|
Samprasit W, Opanasopit P, Chamsai B. Alpha-mangostin and resveratrol, dual-drugs-loaded mucoadhesive thiolated chitosan-based nanoparticles for synergistic activity against colon cancer cells. J Biomed Mater Res B Appl Biomater 2021; 110:1221-1233. [PMID: 34919783 DOI: 10.1002/jbm.b.34992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 10/25/2021] [Accepted: 12/05/2021] [Indexed: 02/04/2023]
Abstract
Alpha-mangostin (M) and resveratrol (R), dual-drugs-loaded mucoadhesive thiolated chitosan-based nanoparticles (NPs) coated by Eudragit® S100 (S) were developed for colon-specific delivery and synergistic activity against colon cancer cells. The NPs were prepared by the ionotropic gelation method and coated with S. The particle size and zeta potential of NPs before and after the coating process were observed. The M and R loading efficiency, mucoadhesive properties, as well as release patterns were examined. Moreover, the activity against colon cancer cells of M, R, and NPs were studied for their synergistic activity. M and R-loaded NPs (MR-TNPs) were spherical in shape with sizes of around 540 nm and zeta potential of +39 mV. The S coating of MR-TNPs provided larger particle sizes which offered lower zeta potential. However, it created an increase in M and R loading, prevented M and R release at the upper gastrointestinal tract, and enhanced M and R reaching the colon. S dissolved at pH > 7.0 while thiolated chitosan formed the mucoadhesion, resulting in M and R remaining in the colon and allowing them to enter the colon cancer cells. The half-maximal inhibitory concentration values of NPs was dramatically decreased when M and R were dually loaded into the NPs, which indicated significantly higher activity against colon cancer cells. Moreover, M and R loading at this ratio applied synergistic efficiency. The results illustrated that NPs successfully loaded drugs and achieved synergistic efficiency. This system could be promising in facilitating targeted nanomedicines for the treatment of colon cancer.
Collapse
Affiliation(s)
- Wipada Samprasit
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Praneet Opanasopit
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Benchawan Chamsai
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| |
Collapse
|
36
|
Winifred Nompumelelo Simelane N, Abrahamse H. Nanoparticle-Mediated Delivery Systems in Photodynamic Therapy of Colorectal Cancer. Int J Mol Sci 2021; 22:12405. [PMID: 34830287 PMCID: PMC8622021 DOI: 10.3390/ijms222212405] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) involving a malignant tumour remains one of the greatest contributing causes of fatal mortality and has become the third globally ranked malignancy in terms of cancer-associated deaths. Conventional CRC treatment approaches such as surgery, radiation, and chemotherapy are the most utilized approaches to treat this disease. However, they are limited by low selectivity and systemic toxicity, so they cannot completely eradicate this disease. Photodynamic therapy (PDT) is an emerging therapeutic modality that exerts selective cytotoxicity to cancerous cells through the activation of photosensitizers (PSs) under light irradiation to produce cytotoxic reactive oxygen species (ROS), which then cause cancer cell death. Cumulative research findings have highlighted the significant role of traditional PDT in CRC treatment; however, the therapeutic efficacy of the classical PDT strategy is restricted due to skin photosensitivity, poor cancerous tissue specificity, and limited penetration of light. The application of nanoparticles in PDT can mitigate some of these shortcomings and enhance the targeting ability of PS in order to effectively use PDT against CRC as well as to reduce systemic side effects. Although 2D culture models are widely used in cancer research, they have some limitations. Therefore, 3D models in CRC PDT, particularly multicellular tumour spheroids (MCTS), have attracted researchers. This review summarizes several photosensitizers that are currently used in CRC PDT and gives an overview of recent advances in nanoparticle application for enhanced CRC PDT. In addition, the progress of 3D-model applications in CRC PDT is discussed.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| |
Collapse
|
37
|
Combination of resveratrol and BIBR1532 inhibits proliferation of colon cancer cells by repressing expression of LncRNAs. Med Oncol 2021; 39:12. [PMID: 34779924 DOI: 10.1007/s12032-021-01611-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The development of tumor drug resistance is observed in the treatment of CRC. Combinations of anticancer agents are attracting considerable interest in order to overcome drug resistance in CRC. This study aims to investigate the effect of resveratrol and BIBR1532, either alone or in combination, on the cell viability as well as on expression of long non-coding RNAs (LncRNAs) for HT-29 colon adenocarcinoma cells. The cytotoxic effects of resveratrol and BIBR1532 on HT-29 cells were determined using WST-1 test. Flow cytometry was used to determine apoptotic cell death after treatments. Real-Time PCR was used to identify expression of LncRNAs after treatments. LncExpDB and GEPIA2 were used to evaluate expression profiles of LncRNAs, whose expression levels were decreased in HT-29 cells after treatments, in normal tissues and colon adenocarcinoma tumors. IC50 concentrations of BIBR1532 and resveratrol were found to be 50.81 μM at 48 h and 86.23 μM at 72 h, respectively. Combination index value was 1.07617. BIBR1532, resveratrol, or their combination reduced the cell viability of HT-29 cells. CCAT1, CRNDE, HOTAIR, PCAT1, PVT1, SNHG16 were down-regulated after treatments. In silico analysis revealed that LncRNAs whose expression levels were decreased after treatments were associated with CRC. Resveratrol, BIBR1532, or their combination may have anti-proliferative effect on colorectal cancer cells through repressing expression of LncRNAs that are involved in progression of CRC.
Collapse
|
38
|
Zeya B, Nafees S, Imtiyaz K, Uroog L, Fakhri KU, Rizvi MMA. Diosmin in combination with naringenin enhances apoptosis in colon cancer cells. Oncol Rep 2021; 47:4. [PMID: 34738632 DOI: 10.3892/or.2021.8215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/09/2021] [Indexed: 11/05/2022] Open
Abstract
Colon cancer is one of the most commonly diagnosed malignancies, which begins as a polyp and grows to become cancer. Diosmin (DS) and naringenin (NR) are naturally occurring flavonoids that exhibit various pharmacological activities. Although several studies have illustrated the effectiveness of these flavonoids as anti‑cancerous agents individually, the combinatorial impact of these compounds has not been explored. In the present study, the combined effect of DS and NR (DiNar) in colon cancer cell lines HCT116 and SW480 were assessed by targeting apoptosis and inflammatory pathways. The MTT assay was used to evaluate the effect of DiNar on cell proliferation, while Chou‑Talalay analysis was employed to determine the combination index of DS and NR. Moreover, flow cytometry was used to monitor cell cycle arrest and population study. The onset of apoptosis was assessed by DAPI staining, DNA fragmentation, and Annexin V‑fluorescein isothiocyanate/propidium iodide (Annexin V‑FITC/PI). The expression levels of apoptotic pathway markers, Bcl‑2, Bax, caspase3, caspase8, caspase9 and p53, and inflammatory markers, NF‑κβ, IKK‑α and IKK‑β, were assessed using western blotting and reverse transcription‑quantitative PCR. These results suggested that DiNar treatment acts synergistically and induces cytotoxicity with a concomitant increase in chromatin condensation, DNA fragmentation and cell cycle arrest in the G0/G1 phase. Annexin V‑FITC/PI apoptosis assay also showed increased number of cells undergoing apoptosis in the DiNar treatment group. Furthermore, the expression of apoptosis and inflammatory markers was also more effectively regulated under the DiNar treatment. Thereby, these findings demonstrated that DiNar treatment could be a potential novel chemotherapeutic alternative in colon cancer.
Collapse
Affiliation(s)
- Bushra Zeya
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| | - Sana Nafees
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| | - Khalid Imtiyaz
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| | - Laraib Uroog
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| | - Khalid Umar Fakhri
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| | - M Moshahid A Rizvi
- The Genome Biology Laboratory, Department of Biosciences, Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New Delhi, Delhi 110025, India
| |
Collapse
|
39
|
Yang L, Bi T, Zhou S, Lan Y, Zhang R. CircRASSF2 facilitates the proliferation and metastasis of colorectal cancer by mediating the activity of Wnt/β-catenin signaling pathway by regulating the miR-195-5p/FZD4 axis. Anticancer Drugs 2021; 32:919-929. [PMID: 33929991 DOI: 10.1097/cad.0000000000001084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Circular RNAs (circRNA) are a key regulator of cancer progression, including colorectal cancer (CRC). Nevertheless, the role of circRASSF2 in CRC remains unclear. Quantitative real-time PCR was used to measure the expression of circRASSF2 and miR-195-5p. Cell counting kit 8 assay, colony formation assay, flow cytometry and transwell assay were used to determine the proliferation, apoptosis, migration and invasion of cells, respectively. The levels of proliferation, metastasis and Wnt/β-catenin signaling pathway-related proteins, as well as Frizzled 4 (FZD4) protein, were determined using western blot analysis. Furthermore, a dual-luciferase reporter assay, RNA immunoprecipitation assay and RNA pull-down assay were used to illumine the mechanism of circRASSF2. Animal experiments were used to determine the role of circRASSF2 in the tumor growth of CRC in vivo. Our study reported that circRASSF2 was upregulated in CRC tissues and cells, and its high expression was related to the poor prognosis of CRC patients. CircRASSF2 knockdown could inhibit proliferation, migration, invasion, and enhance apoptosis in CRC cells, and its overexpression had the opposite effect. Besides, our data revealed that circRASSF2 could sponge miR-195-5p, and miR-195-5p could target FZD4. The rescue experiments indicated that both miR-195-5p inhibitor and FZD4 overexpression could reverse the negative regulation of circRASSF2 silencing on CRC progression. Moreover, circRASSF2 could positively regulate the activity of Wnt/β-catenin signaling pathway by the miR-195-5p/FZD4 axis. In addition, circRASSF2 knockdown restrained the tumor growth of CRC in vivo. Our findings suggested that circRASSF2 might function as a tumor promoter to accelerate the progression of CRC via regulating the miR-195-5p/FZD4/Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Leilei Yang
- Department of Gastrointestinal Surgery, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai City, Zhejiang Province, China
| | | | | | | | | |
Collapse
|
40
|
Yang J, Tong Q, Zhang Y, Yuan S, Gao Y, Deng K, Wang Y, Lu J, Xie X, Zhang Z, Zhang J. Overexpression of Nicotinamide N-methyltransferase mainly covers stroma of colorectal cancer and correlates with unfavorable survival by its product 1-MNA. J Cancer 2021; 12:6170-6181. [PMID: 34539890 PMCID: PMC8425209 DOI: 10.7150/jca.56419] [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: 11/25/2020] [Accepted: 08/14/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Accumulating evidence indicates that Nicotinamide N-methyltransferase (NNMT) is abnormally expressed in tumor tissues of several cancers including colorectal cancer (CRC) and associated with cancer progression. However, the distribution characteristics and the clinical value of each part of NNMT expression in CRC are still not fully understood. The purpose of this study is to determine the distribution of NNMT expression and its association with survival in CRC. Methods: By using the cancer genome atlas (TCGA) and clinical proteomic tumor analysis consortium (CPTAC), we firstly analyzed the difference of gene and protein levels of NNMT between CRC and normal colorectal tissue. Then, NNMT protein expressions were detected in 18 intraepithelial neoplastic samples and 177 CRC tumor samples through immunohistochemistry in our study cohort. Furthermore, the relationship between NNMT expression and clinicopathological characteristics, overall survival (OS) and disease-free survival (DFS) of CRC patients were analyzed by Pearson χ2 test and log-rank test, respectively, in public datasets and our study cohort. Lastly, the function of NNMT and its product 1-methyl-nicotinamide (1-MNA) on migration and invasion in colorectal cancer cells was analyzed by wound healing assay and transwell assay. Results: We determined that higher NNMT expression in CRC tissues than normal tissues in both gene and protein level in TCGA and CPTAC datasets (all p < 0.05). In addition, the strong relationships of NNMT expression with stromal cells were found in the TCGA cohort. Fortunately, our cohort could validate that the expression of NNMT in tumor stroma cell was significantly higher than that in tumor cell (p < 0.0001), and both of them were significantly higher than that in adjacent normal tissue (ANT) (p < 0.0001 and p < 0.0001, respectively). Furthermore, the positive NNMT expression in tumor cell and stromal cell were associated with series of unfavorable clinical characteristics, including advanced TNM stage, lymph node metastasis, distant metastasis (all p < 0.05). Also, higher NNMT was associated with unfavorable survival both in our study and public datasets, including TCGA and two Gene Expression Omnibus (GEO) datasets (GSE33113 and GSE17538). Moreover, the functional experiments showed that stromal cells with high NNMT expression can secret 1-MAN to promote migration and invasion of CRC cells in vitro. Conclusions: In CRC, NNMT is overexpressed in tumor cells and stroma cells, and then mainly expressed in tumor stroma cells. Overexpression of NNMT in tumor cell and stroma cell both are associated with metastasis and unfavorable survival. Besides, stromal cells with high NNMT expression secrets 1-MAN to promote migration and invasion of CRC cells. Therefore, NNMT may be a potential prognostic indicator in CRC patients.
Collapse
Affiliation(s)
- Jun Yang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Ningbo Diagnostic Pathology Center, 685 North Huancheng Road, Ningbo 315010, Zhejiang, People's Republic of China.,Department of Pathology, Ningbo Medical Center Lihuili Hospital, 57 Xingning Road, Ningbo 315040, Zhejiang, People's Republic of China
| | - Qingchao Tong
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Ying Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Shijin Yuan
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Ke Deng
- Department of colorectal surgery, Ningbo Medical Center Lihuili Hospital, 57 Xingning Road, Ningbo 315040, Zhejiang, People's Republic of China
| | - Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Jie Lu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| | - Zhe Zhang
- Ningbo Diagnostic Pathology Center, 685 North Huancheng Road, Ningbo 315010, Zhejiang, People's Republic of China
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China.,Key Laboratory of Biotherapy of Zhejiang Province, 3 East Qingchun Road, Hangzhou 310016, Zhejiang, People's Republic of China
| |
Collapse
|
41
|
Aboyewa JA, Sibuyi NRS, Meyer M, Oguntibeju OO. Green Synthesis of Metallic Nanoparticles Using Some Selected Medicinal Plants from Southern Africa and Their Biological Applications. PLANTS (BASEL, SWITZERLAND) 2021; 10:1929. [PMID: 34579460 PMCID: PMC8472917 DOI: 10.3390/plants10091929] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023]
Abstract
The application of metallic nanoparticles (MNPs), especially that of silver, gold, cobalt, and zinc as antimicrobial, anticancer, drug delivery, contrast, and bioimaging agents has transformed the field of medicine. Their functions, which are attributed to their physicochemical properties, have gained prominence in various technological fields. Although MNPs can be produced via rigorous physical and chemical techniques, in recent years, a biological approach utilizing natural materials has been developed. With the increasing enthusiasm for safe and efficient nanomaterials, the biological method incorporating microorganisms and plants is preferred over physical and chemical methods of nanoparticle synthesis. Of these bio-entities, plants have received great attention owing to their capability to reduce and stabilize MNPs in a single one-pot protocol. South Africa is home to ~10% of the world's plant species, making it a major contributor to the world's ecological scenery. Despite the documented contribution of South African plants, particularly in herbal medicine, very few of these plants have been explored for the synthesis of the noble MNPs. This paper provides a review of some important South African medicinal plants that have been utilized for the synthesis of MNPs. The enhanced biological properties of the biogenic MNPs attest to their relevance in medicine. In this endeavour, more of the African plant biodiversity must be explored for the synthesis of MNPs and be validated for their potential to be translated into future nanomedicine.
Collapse
Affiliation(s)
- Jumoke A. Aboyewa
- Oxidative Stress Research Centre, Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Nicole R. S. Sibuyi
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Bellville 7530, South Africa;
| | - Mervin Meyer
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Bellville 7530, South Africa;
| | - Oluwafemi O. Oguntibeju
- Oxidative Stress Research Centre, Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| |
Collapse
|
42
|
Guo K, Zhang Y, Liu L, Meng H. LncRNA SNHG12 promotes the development and progression of colon cancer by regulating the miR-15a/PDK4 axis. Am J Transl Res 2021; 13:10233-10247. [PMID: 34650693 PMCID: PMC8507043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/20/2021] [Indexed: 06/13/2023]
Abstract
Colon cancer is a common gastrointestinal tumor with complex pathological process. Recently, the relationship between long non-coding RNA (lncRNA) and colon cancer has attracted more and more attention, whereas the underlying molecular mechanism is still poorly understood. Here, we found that the expression of lncRNA small nucleolar RNA host gene 12 (SNHG12) was markedly upregulated in colon cancer samples compared to normal adjacent tissues. Notably, patients with low expression of SNHG12 displayed higher survival rate than those with high expression of SNHG12. Further researches revealed that knockdown of SNHG12 suppressed the malignant phenotype of colon cancer cells. Interestingly, SNHG12 could function as a sponge to specifically bind to microRNA-15a (miR-15a). Moreover, we confirmed that pyruvate dehydrogenase kinase 4 (PDK4) is a direct target gene of miR-15a. Finally, inhibiting miR-15a expression largely abolished the effect of SNHG12 silencing on colon cancer cells. In conclusion, our data uncovered the critical role of SNHG12 in the development and progression of colon cancer through regulating the miR-15a/PDK4 axis, therefore providing a promising target for treating this disease.
Collapse
Affiliation(s)
- Kun Guo
- Department of Gastroenterology, Caoxian People’s HospitalHeze 274400, Shandong Province, China
| | - Yun Zhang
- Department of Gastroenterology, Caoxian People’s HospitalHeze 274400, Shandong Province, China
| | - Lizhi Liu
- Department of General Surgery, Linyi People’s HospitalDezhou 251500, Shandong Province, China
| | - Hua Meng
- Department of Gastroenterology, Caoxian People’s HospitalHeze 274400, Shandong Province, China
| |
Collapse
|
43
|
Chadha S, Kumar A, Srivastava SA, Behl T, Ranjan R. Inulin as a Delivery Vehicle for Targeting Colon-Specific Cancer. Curr Drug Deliv 2021; 17:651-674. [PMID: 32459607 DOI: 10.2174/1567201817666200527133719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/11/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
Natural polysaccharides, as well as biopolymers, are now days widely developed for targeting colon cancer using various drug delivery systems. Currently, healing conformations are being explored that can efficiently play a multipurpose role. Owing to the capability of extravagance colonic diseases with the least adverse effects, biopolymers for site specific colon delivery have developed an increased curiosity over the past decades. Inulin (INU) was explored for its probable application as an entrapment material concerning its degradation by enzymes in the colonic microflora and its drug release behavior in a sustained and controlled manner. INU is a polysaccharide and it consists of 2 to 1 linkage having an extensive array of beneficial uses such as a carrier for delivery of therapeutic agents as an indicative/investigative utensil or as a dietary fiber with added well-being aids. In the main, limited research, as well as information, is available on the delivery of therapeutic agents using inulin specifically for colon cancer because of its capability to subsist in the stomach's acidic medium. This exceptional steadiness and robustness properties are exploited in numerous patterns to target drugs securely for the management of colonic cancer, where they effectively act and kills colonic tumor cells easily. In this review article, recent efforts and inulin-based nano-technological approaches for colon cancer targeting are presented and discussed.
Collapse
Affiliation(s)
- Swati Chadha
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Tapan Behl
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rishu Ranjan
- Department of Pharmaceutics, Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
44
|
Ghasemi T, Khalaj-Kondori M, Hosseinpour Feizi MA, Asadi P. Long non-coding RNA AGAP2-AS1 is up regulated in colorectal cancer. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2021; 40:829-844. [PMID: 34308771 DOI: 10.1080/15257770.2021.1956530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Accumulating evidence has indicated that, aberrant lncRNA expression plays essential roles in the colorectal cancer (CRC) tumorigenesis. AGAP2-AS1 is upregulated in some cancers, however, its involvement in the CRC tumorigenesis in the population of North-West of Iran has remained unknown. In this study, we evaluated its deregulation in CRC microarray datasets, colon cell lines, CRC tumor, adenomatous colorectal polyps and their paired normal tissues. The results showed that AGAP2-AS1 is upregulated in CRC and might be considered as a potential biomarker for CRC development. Moreover, our results suggest AGAP2-AS1 promoted CRC progression by sponging the hsa-miR-15/16 family and upregulation of their targets.
Collapse
Affiliation(s)
- Tayyebeh Ghasemi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Parviz Asadi
- Medical Science Division, Imam Sajjad Hospital, Islamic Azad university, Tabriz, Iran
| |
Collapse
|
45
|
Probiotics: A Promising Candidate for Management of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13133178. [PMID: 34202265 PMCID: PMC8268640 DOI: 10.3390/cancers13133178] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the World's third most frequently diagnosed cancer type. It accounted for about 9.4% mortality out of the total incidences of cancer in the year 2020. According to estimated facts by World Health Organization (WHO), by 2030, 27 million new CRC cases, 17 million deaths, and around 75 million people living with the disease will appear. The facts and evidence that establish a link between the intestinal microflora and the occurrence of CRC are quite intuitive. Current shortcomings of chemo- and radiotherapies and the unavailability of appropriate treatment strategies for CRC are becoming the driving force to search for an alternative approach for the prevention, therapy, and management of CRC. Probiotics have been used for a long time due to their beneficial health effects, and now, it has become a popular candidate for the preventive and therapeutic treatment of CRC. The probiotics adopt different strategies such as the improvement of the intestinal barrier function, balancing of natural gut microflora, secretion of anticancer compounds, and degradation of carcinogenic compounds, which are useful in the prophylactic treatment of CRC. The pro-apoptotic ability of probiotics against cancerous cells makes them a potential therapeutic candidate against cancer diseases. Moreover, the immunomodulatory properties of probiotics have created interest among researchers to explore the therapeutic strategy by activating the immune system against cancerous cells. The present review discusses in detail different strategies and mechanisms of probiotics towards the prevention and treatment of CRC.
Collapse
|
46
|
Nkune NW, Kruger CA, Abrahamse H. Possible Enhancement of Photodynamic Therapy (PDT) Colorectal Cancer Treatment when Combined with Cannabidiol. Anticancer Agents Med Chem 2021; 21:137-148. [PMID: 32294046 DOI: 10.2174/1871520620666200415102321] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/23/2019] [Accepted: 02/04/2020] [Indexed: 11/22/2022]
Abstract
Colorectal Cancer (CRC) has a high mortality rate and is one of the most difficult diseases to manage due to tumour resistance and metastasis. The treatment of choice for CRC is reliant on the phase and time of diagnosis. Despite several conventional treatments available to treat CRC (surgical excision, chemo-, radiationand immune-therapy), resistance is a major challenge, especially if it has metastasized. Additionally, these treatments often cause unwanted adverse side effects and so it remains imperative to investigate alternative combination therapies. Photodynamic Therapy (PDT) is a promising treatment modality for the primary treatment of CRC, since it is non-invasive, has few side effects and selectively damages only cancerous tissues, leaving adjacent healthy structures intact. PDT involves three fundamentals: a Photosensitizer (PS) drug localized in tumour tissues, oxygen, and light. Upon PS excitation using a specific wavelength of light, an energy transfer cascade occurs, that ultimately yields cytotoxic species, which in turn induces cell death. Cannabidiol (CBD) is a cannabinoid compound derived from the Cannabis sativa plant, which has shown to exert anticancer effects on CRC through different pathways, inducing apoptosis and so inhibiting tumour metastasis and secondary spread. This review paper highlights current conventional treatment modalities for CRC and their limitations, as well as discusses the necessitation for further investigation into unconventional active nanoparticle targeting PDT treatments for enhanced primary CRC treatment. This can be administered in combination with CBD, to prevent CRC secondary spread and enhance the synergistic efficacy of CRC treatment outcomes, with less side effects.
Collapse
Affiliation(s)
- Nkune W Nkune
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Cherie A Kruger
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
47
|
Laka K, Mapheto K, Mbita Z. Selective in vitro cytotoxicity effect of Drimia calcarata bulb extracts against p53 mutant HT-29 and p53 wild-type Caco-2 colorectal cancer cells through STAT5B regulation. Toxicol Rep 2021; 8:1265-1279. [PMID: 34195018 PMCID: PMC8233163 DOI: 10.1016/j.toxrep.2021.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer is the fourth leading cause of oncological-related deaths and the third most diagnosed malignancy, worldwide. The emergence of chemoresistance is a fundamental drawback of colorectal cancer therapies and there is an urgent need for novel plant-derived therapeutics. In this regard, other compounds are needed to improve the efficacy of treatment against colorectal cancer. Medicinal plants have been effectively used by traditional doctors for decades to treat various ailments with little to no side effects. Drimia calcarata (D. calcarata) is one of the plants used by Pedi people in South Africa to treat a plethora of ailments. However, the anticancer therapeutic use of D. calcarata is less understood. Thus, this study was aimed at evaluating the potential anticancer activities of D. calcarata extracts against human colorectal cancer cells. The phytochemical analysis and antioxidant activity were analysed using LC-MS, DPPH, and FRAP. The inhibitory effects and IC50 values of D. calcarata extracts were determined using the MTT assay. Induction of cellular apoptosis was assessed using fluorescence microscopy, the Muse® Cell Analyser, and gene expression analysis by Polymerase Chain Reaction (PCR). Water extract (WE) demonstrated high phenolic, tannin, and flavonoid contents than the methanol extract (ME). LC-MS data demonstrated strong differences between the ME and WE. Moreover, WE showed the best antioxidant activity than ME. The MTT data showed that both ME and WE had no significant activity against human embryonic kidney Hek 293 cell line that served as non-cancer control cells. Caco-2 cells demonstrated high sensitivity to the ME and demonstrated resistance toward the WE, while HT-29 cells exhibited sensitivity to both D. calcarata extracts. The expression of apoptosis regulatory genes assessed by PCR revealed an upregulation of p53 by ME, accompanied by downregulation of Bcl-2 and high expression of Bax after treatment with curcumin. The Bax gene was undetected in HT-29 cells. The methanol extract induced mitochondrial-mediated apoptosis in colorectal Caco-2 and HT-29 cells and WE induced the extrinsic apoptotic pathway in HT-29 cells. ME downregulated STAT1, 3, and 5B in HT-29 cells. The D. calcarata bulb extracts, therefore, contain potential anticancer agents that can be further targeted for cancer therapeutics.
Collapse
Affiliation(s)
- K. Laka
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga, 0727, Polokwane, South Africa
| | - K.B.F. Mapheto
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga, 0727, Polokwane, South Africa
| | - Z. Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga, 0727, Polokwane, South Africa
| |
Collapse
|
48
|
Wang H, Xu Y, Sun J, Sui Z. The Novel Curcumin Derivative 1g Induces Mitochondrial and ER-Stress-Dependent Apoptosis in Colon Cancer Cells by Induction of ROS Production. Front Oncol 2021; 11:644197. [PMID: 34195069 PMCID: PMC8236884 DOI: 10.3389/fonc.2021.644197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Reactive oxygen species (ROS) play an important role in cellular metabolism. Many chemotherapeutic drugs are known to promote apoptosis through the production of ROS. In the present study, the novel curcumin derivative, 1g, was found to inhibit tumor growth in colon cancer cells both in vitro and in vivo. Bioinformatics was used to analyze the differentially expressed mRNAs. The mechanism of this effect was a change in mitochondrial membrane potential caused by 1g that increased its pro-apoptotic activity. In addition, 1g produced ROS, induced G1 checkpoint blockade, and enhanced endoplasmic reticulum (ER)-stress in colon cancer cells. Conversely, pretreatment with the ROS scavenging agent N-acetyl-l-cysteine (NAC) inhibited the mitochondrial dysfunction caused by 1g and reversed ER-stress, cell cycle stagnation, and apoptosis. Additionally, pretreatment with the p-PERK inhibitor GSK2606414 significantly reduced ER-stress and reversed the apoptosis induced by colon cancer cells. In summary, the production of ROS plays an important role in the destruction of colon cancer cells by 1g and demonstrates that targeted strategies based on ROS represent a promising approach to inhibit colon cancer proliferation. These findings reveal that the novel curcumin derivative 1g represents a potential candidate therapeutics for the treatment of colon cancer cells, via apoptosis caused by mitochondrial dysfunction and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Hao Wang
- Department of Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Medicine, Qingdao University, Qingdao, China
| | - Yingxing Xu
- Department of Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Medicine, Qingdao University, Qingdao, China
| | - Jialin Sun
- Department of Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Medicine, Qingdao University, Qingdao, China
| | - Zhongguo Sui
- Department of Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Lee J, Kwon J, Kim D, Park M, Kim K, Bae I, Kim H, Kong J, Kim Y, Shin U, Kim E. Gene Expression Profiles Associated with Radio-Responsiveness in Locally Advanced Rectal Cancer. BIOLOGY 2021; 10:biology10060500. [PMID: 34205090 PMCID: PMC8226560 DOI: 10.3390/biology10060500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/14/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Standard treatment of locally advanced rectal cancer (LARC) consists of chemotherapy, radiotherapy, and surgery. Identification of radio-resistant (RR) and radio-sensitive (RS) LARC has been a major hurdle for patient-specific treatment. The development of biomarkers that can discriminate radio-responsiveness before surgery could improve standard treatment and minimize unwanted side effects. Abstract LARC patients were sorted according to their radio-responsiveness and patient-derived organoids were established from the respective cancer tissues. Expression profiles for each group were obtained using RNA-seq. Biological and bioinformatic analysis approaches were used in deciphering genes and pathways that participate in the radio-resistance of LARC. Thirty candidate genes encoding proteins involved in radio-responsiveness–related pathways, including the immune system, DNA repair and cell-cycle control, were identified. Interestingly, one of the candidate genes, cathepsin E (CTSE), exhibited differential methylation at the promoter region that was inversely correlated with the radio-resistance of patient-derived organoids, suggesting that methylation status could contribute to radio-responsiveness. On the basis of these results, we plan to pursue development of a gene chip for diagnosing the radio-responsiveness of LARC patients, with the hope that our efforts will ultimately improve the prognosis of LARC patients.
Collapse
Affiliation(s)
- Jeeyong Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.L.); (D.K.); (K.K.); (I.B.)
| | - Junhye Kwon
- Department of Radiological & Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.K.); (M.P.); (H.K.); (Y.K.)
| | - DaYeon Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.L.); (D.K.); (K.K.); (I.B.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
| | - Misun Park
- Department of Radiological & Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.K.); (M.P.); (H.K.); (Y.K.)
| | - KwangSeok Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.L.); (D.K.); (K.K.); (I.B.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
| | - InHwa Bae
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.L.); (D.K.); (K.K.); (I.B.)
| | - Hyunkyung Kim
- Department of Radiological & Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.K.); (M.P.); (H.K.); (Y.K.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
| | - JoonSeog Kong
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Younjoo Kim
- Department of Radiological & Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.K.); (M.P.); (H.K.); (Y.K.)
- Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - UiSup Shin
- Department of Radiological & Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.K.); (M.P.); (H.K.); (Y.K.)
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
- Correspondence: (U.S.); (E.K.)
| | - EunJu Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.L.); (D.K.); (K.K.); (I.B.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (U.S.); (E.K.)
| |
Collapse
|
50
|
Multiple strategies with the synergistic approach for addressing colorectal cancer. Biomed Pharmacother 2021; 140:111704. [PMID: 34082400 DOI: 10.1016/j.biopha.2021.111704] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer treatment is improving widely over time, but finding a proper defender to beat them seems like a distant dream. The quest for identification and discovery of drugs with an effective action is still a vital work. The role of a membrane protein called P-glycoprotein, which functions as garbage chute that efflux the waste, xenobiotics, and toxins out of the cancer cells acts as a major reason behind the therapeutic failure of most chemotherapeutic drugs. In this review, we mainly focused on a multiple strategies by employing 5-Fluorouracil, curcumin, and lipids in Nano formulation for the possible treatment of colorectal cancer and its metastasis. Eventually, multidrug resistance and angiogenesis can be altered and it would be helpful in colorectal cancer targeting.We have depicted the possible way for the depletion of colorectal cancer cells without disturbing the normal cells. The concept of focusing on multiple pathways for marking the colorectal cancer cells could help in activating one among the pathways if the other one fails. The activity of the 5-Fluorouracil can be enhanced with the help of curcumin which acts as a chemosensitizer, chemotherapeutic agent, and even for altering the resistance. As we eat to survive, so do the cancer cells. The cancer cells utilize the energy source to stay alive and survive. Fatty acids can be used as the energy source and this concept can be employed for targeting the colorectal cancer cells and also for altering the resistant part.
Collapse
|