1
|
Hu Y, Zhou P, Deng K, Zhou Y, Hu K. Targeting the gut microbiota: a new strategy for colorectal cancer treatment. J Transl Med 2024; 22:915. [PMID: 39379983 PMCID: PMC11460241 DOI: 10.1186/s12967-024-05671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND How to reduce the high incidence rate and mortality of colorectal cancer (CRC) effectively is the focus of current research. Endoscopic treatment of early-stage CRC and colorectal adenomas (CAC) has a high success rate, but although several treatments are available for advanced CRC, such as surgery, radiotherapy, chemotherapy, and immunotherapy, the 5-year survival rate remains low. In view of the high incidence rate and mortality of CRC, early rational drug prevention for high-risk groups and exploration of alternative treatment modalities are particularly warranted. Gut microbiota is the target of and interacts with probiotics, prebiotics, aspirin, metformin, and various Chinese herbal medicines (CHMs) for the prevention of CRC. In addition, the anti-cancer mechanisms of probiotics differ widely among bacterial strains, and both bacterial strains and their derivatives and metabolites have been found to have anti-cancer effects. Gut microbiota plays a significant role in early drug prevention of CRC and treatment of CRC in its middle and late stages, targeting gut microbiota may be a new strategy for colorectal cancer treatment.
Collapse
Affiliation(s)
- Yue Hu
- Health Science Center, Ningbo University, Ningbo, China
| | - Peng Zhou
- Health Science Center, Ningbo University, Ningbo, China
| | - Kaili Deng
- Health Science Center, Ningbo University, Ningbo, China
| | - Yuping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
- Institute of Digestive Disease of Ningbo University, Ningbo, China.
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo, China.
| | - Kefeng Hu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
2
|
Abdalla Abdelaziz MA, Nelson VK, Kumarasamy M, Peraman R. Anticancer effect of polyphenolic acid enriched fractions from Grewia bracteata Roth on tumor cells and their p53 gene independent ROS mediated apoptosis in colon cancer cells. Toxicon 2023; 233:107243. [PMID: 37567418 DOI: 10.1016/j.toxicon.2023.107243] [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] [Revised: 07/12/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
It is the first report on leaves of Grewia bracteata Roth for its anticancer effect. In this study, three polarity-guided solvent extracts of Grewia bracteata leaves from n-hexane (GLH), ethyl acetate (GLE), and methanol (GLM) were screened for anticancer effects on HeLa, HCT-116, MCF-7, HCT-116 p53-/- and PC-3 cells via methyl thiazoldiphenyltetrazolium bromide (MTT) assay. Based on the results, GLM was fractionated, and the obtained fractions were tested on HCT-116 cells. Further, FT-IR, HPLC analysis, clonogenic assay, wound healing assay, DCFDA, and cell cycle experiments were conducted on HCT-116 cells. The extracts from methanol (GLM) and ethyl-acetate (GLE) demonstrated a more selective and promising inhibition on HCT-116 cells than others. Notably, GLM recorded superior inhibition on HCT-116 p53-/- than GLE. Amongst, the methanol column fraction (GMCF) showed prominent inhibition on HCT-116 (IC50:63.55 ± 0.61 μg/ml) and HCT-116 p53-/- (IC50: 84.51 ± 0.58 μg/ml) cells. Further, the test on normal cells (NKE) revealed minimal toxicity of GMCF. The phytochemical test, FT-IR, HPLC, and LC-HRMS analyses confirmed the high abundance of polyphenolic acid/polyphenols in GMCF. Further, the clonogenic and wound healing assays on HCT-116 cells were also performed. Later, the probable cell death mechanism was identified using DCFDA and cell cycle experiments. These experiments disclosed that GMCF induced HCT-116 cell death was probably due to reactive oxygen species (ROS) upregulation and cells cycle arrest at SubG0 phase. It inferred that the activity is most probably p53 independent, a tumor suppressor gene responsible for drug resistance in colon cancer.
Collapse
Affiliation(s)
| | - Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, 515742, India
| | - Murali Kumarasamy
- National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, 844102, India
| | - Ramalingam Peraman
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, 515742, India; National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, 844102, India.
| |
Collapse
|
3
|
Sompel K, Dwyer-Nield LD, Smith AJ, Elango A, Backos DS, Zhang B, Gross J, Ternyak K, Matsuda JL, Kopf K, Keith RL, Tennis MA. Iloprost requires the Frizzled-9 receptor to prevent lung cancer. iScience 2022; 25:104442. [PMID: 35707728 PMCID: PMC9189122 DOI: 10.1016/j.isci.2022.104442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/31/2022] Open
Abstract
Prevention of premalignant lesion progression is a promising approach to reducing lung cancer burden in high-risk populations. Substantial preclinical and clinical evidence has demonstrated efficacy of the prostacyclin analogue iloprost for lung cancer chemoprevention. Iloprost activates peroxisome proliferator-activated receptor gamma (PPARG) to initiate chemopreventive signaling and in vitro, which requires the transmembrane receptor Frizzled9 (FZD9). We hypothesized a Fzd 9 -/- mouse would not be protected by iloprost in a lung cancer model. Fzd 9 -/- mice were treated with inhaled iloprost in a urethane model of lung adenoma. We found that Fzd 9 -/- mice treated with iloprost were not protected from adenoma development compared to wild-type mice nor did they demonstrate increased activation of iloprost signaling pathways. Our results established that iloprost requires FZD9 in vivo for lung cancer chemoprevention. This work represents a critical advancement in defining iloprost's chemopreventive mechanisms and identifies a potential response marker for future clinical trials.
Collapse
Affiliation(s)
- Kayla Sompel
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lori D. Dwyer-Nield
- Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alex J. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alamelu Elango
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Don S. Backos
- Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | | - Robert L. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Meredith A. Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
4
|
Feng J, Wang X, Ye X, Ares I, Lopez-Torres B, Martínez M, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Mitochondria as an important target of metformin: The mechanism of action, toxic and side effects, and new therapeutic applications. Pharmacol Res 2022; 177:106114. [DOI: 10.1016/j.phrs.2022.106114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/25/2022]
|
5
|
Dutta N, Pemmaraju DB, Ghosh S, Ali A, Mondal A, Majumder C, Nelson VK, Mandal SC, Misra AK, Rengan AK, Ravichandiran V, Che CT, Gurova KV, Gudkov AV, Pal M. Alkaloid-rich fraction of Ervatamia coronaria sensitizes colorectal cancer through modulating AMPK and mTOR signalling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114666. [PMID: 34592338 DOI: 10.1016/j.jep.2021.114666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/12/2021] [Accepted: 09/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ervatamia coronaria, a popular garden plant in India and some other parts of the world is known traditionally for its anti-inflammatory and anti-cancer properties. The molecular bases of these functions remain poorly understood. AIM OF THE STUDY Efficacies of the existing therapies for colorectal cancer (CRC) are limited by their life-threatening side effects and unaffordability. Therefore, identifying a safer, efficient, and affordable therapeutic is urgent. We studied the anti-CRC activity of an alkaloid-rich fraction of E. coronaria leaf extracts (AFE) and associated underlying mechanism. MATERIALS AND METHODS Activity guided solvant fractionation was adopted to identify the activity in AFE. Different cell lines, and tumor grown in syngeneic mice were used to understand the anti-CRC effect. Methodologies such as LCMS, MTT, RT-qPCR, immunoblot, immunohistochemistry were employed to understand the molecular basis of its activity. RESULTS We showed that AFE, which carries about six major compounds, is highly toxic to colorectal cancer (CRC) cells. AFE induced cell cycle arrest at G1 phase and p21 and p27 genes, while those of CDK2, CDK-4, cyclin-D, and cyclin-E genes were downregulated in HCT116 cells. It predominantly induced apoptosis in HCT116p53+/+ cells while the HCT116p53-/- cells under the same treatment condition died by autophagy. Notably, AFE induced upregulation of AMPK phosphorylation, and inhibition of both of the mTOR complexes as indicated by inhibition of phosphorylation of S6K1, 4EBP1, and AKT. Furthermore, AFE inhibited mTOR-driven conversion of cells from reversible cell cycle arrest to senescence (geroconversion) as well as ERK activity. AFE activity was independent of ROS produced, and did not primarily target the cellular DNA or cytoskeleton. AFE also efficiently regressed CT26-derived solid tumor in Balb/c mice acting alone or in synergy with 5FU through inducing autophagy as a major mechanism of action as indicated by upregulation of Beclin 1 and phospho-AMPK, and inhibition of phospho-S6K1 levels in the tumor tissue lysates. CONCLUSION AFE induced CRC death through activation of both apoptotic and autophagy pathways without affecting the normal cells. This study provided a logical basis for consideration of AFE in future therapy regimen to overcome the limitations associated with existing anti-CRC chemotherapy.
Collapse
Affiliation(s)
- Naibedya Dutta
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Deepak Bharadwaj Pemmaraju
- Division of Molecular Medicine, Bose Institute, Kolkata, India; Department of Biomedical Engineering, IIT, Hyderabad, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Asif Ali
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Ayan Mondal
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Vinod K Nelson
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Subhash C Mandal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Anup K Misra
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | | | - Chun-Tao Che
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, USA
| | - Katerina V Gurova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata, India.
| |
Collapse
|
6
|
Yang J, Fang W, Wu W, Tian Z, Gao R, Yu L, Chen D, Weng X, Zhu S, Yang C. A Novel Diagnostic Biomarker, PZP, for Detecting Colorectal Cancer in Type 2 Diabetes Mellitus Patients Identified by Serum-Based Mass Spectrometry. Front Mol Biosci 2021; 8:736272. [PMID: 34917649 PMCID: PMC8670180 DOI: 10.3389/fmolb.2021.736272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Growing evidence has confirmed that populations with type 2 diabetes mellitus (T2DM) have an increasing risk of developing colorectal cancer (CRC). Thus, convenient and effective screening strategies for CRC should be developed for the T2DM population to increase the detection rate of CRC. Methods: Twenty serum samples extracted from five healthy participants, five T2DM patients, five CRC patients and five T2DM patients with CRC (T2DM + CRC) were submitted to data-independent acquisition mass spectrometry (DIA-MS) analysis to discover unique differentially altered proteins (DAPs) for CRC in patients with T2DM. Then, the diagnostic value of pregnancy zone protein (PZP) was validated by ELISA analysis in the validated cohort. Results: Based on DIA-MS analysis, we found eight unique proteins specific to T2DM patients with CRC. Among these proteins, four proteins showed different expression between the T2DM + CRC and T2DM groups, and PZP exhibited the largest difference. Next, the diagnostic value of serum PZP was validated by ELISA analysis with an AUC of 0.713. Moreover, the combination of PZP, CA199 and CEA exhibited encouraging diagnostic value, and the AUC reached 0.916. Conclusion: Overall, our current research implied that PZP could be regarded as a newfound serum biomarker for CRC medical diagnosis in T2DM patients.
Collapse
Affiliation(s)
- Jiayue Yang
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Weigang Fang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Wenjun Wu
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhen Tian
- Department of Clinical Laboratory, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Rong Gao
- Department of Clinical Laboratory, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Lu Yu
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Dayang Chen
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xiaohua Weng
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Shengwei Zhu
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Cheng Yang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
7
|
Siri M, Behrouj H, Dastghaib S, Zamani M, Likus W, Rezaie S, Hudecki J, Khazayel S, Łos MJ, Mokarram P, Ghavami S. Casein Kinase-1-Alpha Inhibitor (D4476) Sensitizes Microsatellite Instable Colorectal Cancer Cells to 5-Fluorouracil via Authophagy Flux Inhibition. Arch Immunol Ther Exp (Warsz) 2021; 69:26. [PMID: 34536148 PMCID: PMC8449776 DOI: 10.1007/s00005-021-00629-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Adjuvant chemotherapy with 5-fluorouracil (5-FU) does not improve survival of patients suffering from a form of colorectal cancer (CRC) characterized by high level of microsatellite instability (MSI-H). Given the importance of autophagy and multi-drug-resistant (MDR) proteins in chemotherapy resistance, as well as the role of casein kinase 1-alpha (CK1α) in the regulation of autophagy, we tested the combined effect of 5-FU and CK1α inhibitor (D4476) on HCT116 cells as a model of MSI-H colorectal cancer. To achieve this goal, the gene expression of Beclin1 and MDR genes, ABCG2 and ABCC3 were analyzed using quantitative real-time polymerase chain reaction. We used immunoblotting to measure autophagy flux (LC3, p62) and flow cytometry to detect apoptosis. Our findings showed that combination treatment with 5-FU and D4476 inhibited autophagy flux. Moreover, 5-FU and D4476 combination therapy induced G2, S and G1 phase arrests and it depleted mRNA of both cell proliferation-related genes and MDR-related genes (ABCG2, cyclin D1 and c-myc). Hence, our data indicates that targeting of CK1α may increase the sensitivity of HCT116 cells to 5-FU. To our knowledge, this is the first description of sensitization of CRC cells to 5-FU chemotherapy by CK1α inhibitor.
Collapse
Affiliation(s)
- Morvarid Siri
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Hamid Behrouj
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Sanaz Dastghaib
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Wirginia Likus
- Department of Anatomy, School of Health Science in Katowice, Medical University of Silesia, ul. Medyków 18, 40-762, Katowice, Poland
| | - Sedigheh Rezaie
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Jacek Hudecki
- Laryngology Department, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Saeed Khazayel
- Department of Research and Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marek J Łos
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pathology, Unii Lubelskiej 1, Pomeranian Medical University, 71-344, Szczecin, Poland.
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran.
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada.
- Faculty of Medicine, Katowice School of Technology, Katowice, Poland.
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
8
|
Sompel K, Elango A, Smith AJ, Tennis MA. Cancer chemoprevention through Frizzled receptors and EMT. Discov Oncol 2021; 12:32. [PMID: 34604862 PMCID: PMC8429367 DOI: 10.1007/s12672-021-00429-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Frizzled (FZD) transmembrane receptors are well known for their role in β-catenin signaling and development and now understanding of their role in the context of cancer is growing. FZDs are often associated with the process of epithelial to mesenchymal transition (EMT) through β-catenin, but some also influence EMT through non-canonical pathways. With ten different FZDs, there is a wide range of activity from oncogenic to tumor suppressive depending on the tissue context. Alterations in FZD signaling can occur during development of premalignant lesions, supporting their potential as targets of chemoprevention agents. Agonizing or antagonizing FZD activity may affect EMT, which is a key process in lesion progression often targeted by chemoprevention agents. Recent studies identified a specific FZD as important for activity of an EMT inhibiting chemopreventive agent and other studies have highlighted the previously unrecognized potential for targeting small molecules to FZD receptors. This work demonstrates the value of investigating FZDs in chemoprevention and here we provide a review of FZDs in cancer EMT and their potential as chemoprevention targets.
Collapse
Affiliation(s)
- K. Sompel
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. Elango
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. J. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - M. A. Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| |
Collapse
|