1
|
Chen Y, Fang H, Chen H, Liu X, Zhao J, Stanton C, Ross RP, Chen W, Yang B. Bifidobacterium inhibits the progression of colorectal tumorigenesis in mice through fatty acid isomerization and gut microbiota modulation. Gut Microbes 2025; 17:2464945. [PMID: 39924893 PMCID: PMC11812354 DOI: 10.1080/19490976.2025.2464945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Colorectal cancer (CRC) represents the third most common cancer worldwide. Consequently, there is an urgent need to identify novel preventive and therapeutic strategies for CRC. This study aimed to screen for beneficial bacteria that have a preventive effect on CRC and to elucidate the potential mechanisms. Initially, we compared gut bacteria and bacterial metabolites of healthy volunteers and CRC patients, which demonstrated that intestinal conjugated linoleic acid (CLA), butyric acid, and Bifidobacterium in CRC patients were significantly lower than those in healthy volunteers, and these indicators were significantly negatively correlated with CRC. Next, spontaneous CRC mouse model were conducted to explore the effect of supplemental CLA-producing Bifidobacterium on CRC. Supplementation of mice with CLA-producing Bifidobacterium breve CCFM683 and B. pseudocatenulatum MY40C significantly prevented CRC. Moreover, molecular approaches demonstrated that CLA and the CLA-producing gene, bbi, were the key metabolites and genes for CCFM683 to prevent CRC. Inhibitor intervention results showed that PPAR-γ was the key receptor for preventing CRC. CCFM683 inhibited the NF-κB signaling pathway, up-regulated MUC2, Claudin-1, and ZO-1, and promoted tumor cell apoptosis via the CLA-PPAR-γ axis. Additionally, fecal microbiota transplantation (FMT) and metagenomic analysis showed that CCFM683 up-regulated Odoribacter splanchnicus through CLA production, which then prevented CRC by producing butyric acid, up-regulating TJ proteins, regulating cytokines, and regulating gut microbiota. These results will contribute to the clinical trials of Bifidobacterium and the theoretical research and development of CRC dietary products.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Huiting Fang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Catherine Stanton
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R. Paul Ross
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
2
|
Chen Y, Ma W, Zhao J, Stanton C, Ross RP, Zhang H, Chen W, Yang B. Lactobacillus plantarum Ameliorates Colorectal Cancer by Ameliorating the Intestinal Barrier through the CLA-PPAR-γ Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19766-19785. [PMID: 39186442 DOI: 10.1021/acs.jafc.4c02824] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Colorectal cancer (CRC) is the third-largest cancer worldwide. Lactobacillus can regulate the intestinal barrier and gut microbiota. However, the mechanisms of Lactobacillus that alleviate CRC remained unknown. This study aimed to explore the regulatory effect of Lactobacillus plantarum on CRC and its potential mechanism. CCFM8661 treatment significantly ameliorated CRC compared with phosphate-buffered solution (PBS) treatment in ApcMin/+ mice. In addition, conjugated linoleic acid (CLA) was proved to be the key metabolite for CCFM8661 in ameliorating CRC by molecular biology techniques. Peroxisome proliferator-activated receptor γ (PPAR-γ) was proved to be the key receptor in ameliorating CRC by inhibitor intervention experiments. Moreover, supplementation with CCFM8661 ameliorated CRC by producing CLA to inhibit NF-κB pathway and pro-inflammatory cytokines, up-regulate ZO-1, Claudin-1, and MUC2, and promote tumor cell apoptosis in a PPAR-γ-dependent manner. Metagenomic analysis showed that CCFM8661 treatment significantly increased Odoribacter splanchnicus, which could ameliorate CRC by repairing the intestinal barrier. Clinical results showed that intestinal CLA, butyric acid, PPAR-γ, and Lactobacillus were significantly decreased in CRC patients, and these indicators were significantly negatively correlated with CRC. CCFM8661 alleviated CRC by ameliorating the intestinal barrier through the CLA-PPAR-γ axis. These results will promote the development of dietary probiotic supplements for CRC.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan 430068, Hubei, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Catherine Stanton
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - R Paul Ross
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
3
|
Xiong F, Zhou YW, Hao YT, Wei GX, Chen XR, Qiu M. Combining Anti-epidermal Growth Factor Receptor (EGFR) Therapy with Immunotherapy in Metastatic Colorectal Cancer (mCRC). Expert Rev Gastroenterol Hepatol 2024; 18:185-192. [PMID: 37705376 DOI: 10.1080/17474124.2023.2232718] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/30/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION Monoclonal antibodies binding the EGFR, such as cetuximab and panitumumab, have been extensively used as targeted therapy for the treatment of mCRC. However, in clinical practice, it has been found that these treatment options have some limitations and fail to fully exploit their immunoregulatory activities. Meanwhile, because of the limited effects of current treatments, immunotherapy is being widely studied for patients with mCRC. However, previous immunotherapy trials in mCRC patients have had unsatisfactory outcomes as monotherapy. Thus, combinatorial treatment strategies are being researched. AREAS COVERED The authors retrieved relevant documents of combination therapy for mCRC from PubMed and Medline. This review elaborates on the knowledge of immunomodulatory effects of anti-EGFR therapy alone and in combination with immunotherapy for mCRC. EXPERT OPINION Although current treatment options have improved median overall survival (OS) for advanced disease to 30 months, the prognosis remains challenging for those with metastatic disease. More recently, the combination of anti-EGFR therapy with immunotherapy has been shown activity with complementary mechanisms. Hence, anti-EGFR therapy in combination with immunotherapy may hold the key to improving the therapeutic effect of refractory mCRC.
Collapse
Affiliation(s)
- Feng Xiong
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Ya-Ting Hao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Gui-Xia Wei
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao-Rong Chen
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Susanto AC, Hartajanie L, Wu CC. α‑Phellandrene enhances the apoptosis of HT‑29 cells induced by 5‑fluorouracil by modulating the mitochondria‑dependent pathway. Oncol Rep 2024; 51:61. [PMID: 38456489 PMCID: PMC10940876 DOI: 10.3892/or.2024.8720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
α‑Phellandrene (α‑PA), a natural constituent of herbs, inhibits cancer cell viability and proliferation. 5‑Fluorouracil (5‑FU) is a frequently utilized chemotherapeutic medicine for the treatment of colon cancer, which works by triggering cancer cell apoptosis. The present study examined how the combination of α‑PA and 5‑FU affects the suppression of human colon cancer cells by promoting apoptosis. The impact of this treatment on cell viability, apoptosis, and the expression levels of Bcl‑2 family members, caspase family members and mitochondria‑related molecules in HT‑29 cells was assessed by the MTT assay, immunocytochemistry, western blotting and quantitative PCR. The combination of 5‑FU and α‑PA had a synergistic inhibitory effect on cell viability, as determined by assessing the combination index value. Bax protein expression levels were higher in the 50, 100 or 250 µM α‑PA combined with 5‑FU groups compared with those in the 5‑FU alone group (P<0.05). By contrast, Bcl‑2 protein expression levels and mitochondrial membrane potential (MMP, ΔΨm) were lower in the 100 or 250 µM α‑PA combined with 5‑FU groups than those in the 5‑FU alone group (P<0.05). In addition, hexokinase‑2 (HK‑2) protein expression levels were lower in the 50, 100 or 250 µM α‑PA combined with 5‑FU groups than those in the 5‑FU alone group (P<0.05). Compared with 5‑FU alone, after HT‑29 cells were treated with 50, 100 or 250 µM α‑PA combined with 5‑FU, the mRNA expression levels of extrinsic‑induced apoptotic molecules, including caspase‑8 and Bid, were higher (P<0.05). Treatment with 50, 100 or 250 µM α‑PA combined with 5‑FU also increased the mRNA expression levels of cytochrome c, caspase‑9 and caspase‑3, regulating intrinsic apoptosis (P<0.05). These results showed that α‑PA and 5‑FU had a synergistic effect on reducing the viability of human colon cancer HT‑29 cells by inducing extrinsic and intrinsic apoptosis pathways. The mechanism by which apoptosis is induced may involve the intrinsic apoptosis pathway that activates the mitochondria‑dependent pathway, including regulating the expression levels of Bcl‑2 family members, including Bax, Bcl‑2 and Bid, regulating MMP and HK‑2 expression levels, and increasing the expression of caspase cascade molecules, including caspase‑9 and caspase‑3. In addition, it may involve the extrinsic apoptosis pathway that activates caspase‑8 and caspase‑3 leading to apoptosis.
Collapse
Affiliation(s)
- Anita Caroline Susanto
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan, R.O.C
- Faculty of Food Technology, Soegijapranata Catholic University, Semarang 50234, Indonesia
| | - Laksmi Hartajanie
- Faculty of Food Technology, Soegijapranata Catholic University, Semarang 50234, Indonesia
| | - Chih-Chung Wu
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan, R.O.C
| |
Collapse
|
5
|
Imanbayev N, Iztleuov Y, Koishybaev AK, Kereyeva N, Tulyayeva A, Zholmukhamedova D, Zharylgapov A. The Role of Tumor Parenchymal and Stromal Ratios in Colorectal Cancer. J Family Reprod Health 2024; 18:1-8. [PMID: 38863846 PMCID: PMC11162885 DOI: 10.18502/jfrh.v18i1.15433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024] Open
Abstract
Objective To evaluate the implications and significant role of parenchymal and stromal ratio in colorectal cancer (CRC). Materials and methods In our review, we involved English studies from common databases such as Web of Science, Scopus, Google Scholar, PubMed, and the Cochrane Library using the following keywords "colorectal cancer", "tumor stromal ratio", "tumor parenchymal ratio", and "prognostic marker" till December 2023. Results The majority of CRC patients are usually diagnosed at late stages, which may lead to missing out on the chance to get radical therapy. Patients with unfavorable prognosis have epithelial malignant tumors with a high amount of stroma, more than 50% stroma, while tumors with a low amount of stroma, less than 50%, and abundant carcinoma tissue have a better prognosis. Conclusion Tumor-stromal ratio is a valuable, cheap, and fast modality that provides valuable prognostic data of colorectal carcinoma and other cancers.
Collapse
Affiliation(s)
- Nauryzbay Imanbayev
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerbolat Iztleuov
- Department of Radiologists of the NJSC ZKMU named after Marat Ospanov, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Arip K. Koishybaev
- Department of Oncology of the NJSC ZKMU named after M. Ospanov MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Nurgul Kereyeva
- Department of Oncology ZKMU named after Marat Ospanova, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Anar Tulyayeva
- Department of Oncology Medical Center of West Kazakhstan Medical University named after Marat Ospanov, Aktobe, Kazakhstan
| | - Dinara Zholmukhamedova
- Department Oncology MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Aktobe, Kazakhstan
| | - Azamat Zharylgapov
- Department Oncology MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Aktobe, Kazakhstan
| |
Collapse
|
6
|
El-Hazek RMM, Zaher NH, Emam HES, El-Gazzar MG, Khalil A. Pyrazole-sulfonamide scaffold featuring dual-tail strategy as apoptosis inducers in colon cancer. Sci Rep 2023; 13:5782. [PMID: 37031294 PMCID: PMC10082777 DOI: 10.1038/s41598-023-32820-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/03/2023] [Indexed: 04/10/2023] Open
Abstract
Dual-tail strategy has been successfully utilized in the development of novel carbonic anhydrase IX (CA IX) inhibitors. Herein we adopted this approach in the design and synthesis of a series of novel pyridine sulfonamide-pyrazole hybrid scaffold mimicking dual-tail inhibitors of CA IX. A library of 15 compounds was synthesized and assessed for their potential cytotoxic effects against colorectal cancer cells. Compounds 3, and 11 induced potential cytotoxic effects against the three cancer cell lines (HCT-116, HT-29, and SW-620) with IC50s' of 45.88, 28.27, and 16.57 uM, 25.01, 8.99, and 3.27 µM, respectively. Both compounds induced cellular apoptosis on HCT-116 and SW-620 cells, while compound 3 induced necrosis as well. In addition, both compounds induced cell cycle arrest on G0/G1, and S phases. Also, compound 11 showed potential autophagy induction on both colon cancer cell lines (HCT-116, and HT-29), and a little bit on metastatic type. Both compounds were less cytotoxic than the reference drug on normal epithelial cell. The migration rates of HCT-116 and the metastatic one SW-620 were reduced by both compounds. Finally, molecular docking of compounds 3 and 11 into the active site of CA IX confirmed in vitro inhibitory activity for both compounds.
Collapse
Affiliation(s)
- Reham M M El-Hazek
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Nashwa H Zaher
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Hagar E S Emam
- Biomedical Research Division, Nawah Scientific, Cairo, Egypt
| | - Marwa G El-Gazzar
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Amira Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt.
| |
Collapse
|
7
|
Ahmed AA, Monir M, Sabry D, Mostafa A. In vitro study to evaluate the effect of granulocyte colony stimulating factor on colorectal adenocarcinoma and on mesenchymal stem cells trans differentiation into cancer stem cells by cancer cells derived exosomes. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2023. [DOI: 10.1186/s43088-023-00351-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
Abstract
Abstract
Background
Colorectal cancer (CRC) is a common and lethal malignancies with poor prognosis. CRC cells release extracellular vesicles called exosomes to facilitate tumor progression by passing bioactive molecules such as proteins and nucleic acids between cells of the tumor and their microenvironment. Granulocyte colony stimulating factor (G-CSF) is a hematopoietic growth factor which mainly affects the lineage of neutrophil and exerts direct anti-tumor effects on various tumor types. The purpose of our study is to investigate the effect of G-CSF on CRC cells and to evaluate its capability to attenuate the potentiality of CRC cells derived exosomes to induce bone marrow-derived mesenchymal stem cells (BM-MSCs) malignant transformation into cancer stem cells (CSCs).
Results
The level of both lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT-1) (p = 0.014) & β-catenin (p = 0.01) was significantly decreased, whereas programmed cell death 4 (PDCD4) (p = 0.018) was increased in CRC exosomes pre-treated with G-CSF compared to untreated CRC exosomes. Additionally, there was a significant decrease in the cell proliferation in CRC cells pre-treated with G-CSF compared to untreated CRC cells (p = 0.008). Flow cytometric analysis of BM-MSCs showed that G-CSF could attenuate their transformation into CSCs.
Conclusion
G-CSF can be a promising therapeutic agent for CRC treatment.
Collapse
|
8
|
Al-Hujaily EM, Al-Sowayan BS, Alyousef Z, Uddin S, Alammari F. Recruiting Immunity for the Fight against Colorectal Cancer: Current Status and Challenges. Int J Mol Sci 2022; 23:13696. [PMID: 36430176 PMCID: PMC9697544 DOI: 10.3390/ijms232213696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Cancer immunotherapies have changed the landscape of cancer management and improved the standard treatment protocols used in multiple tumors. This has led to significant improvements in progression-free survival and overall survival rates. In this review article, we provide an insight into the major immunotherapeutic methods that are currently under investigation for colorectal cancer (CRC) and their clinical implementations. We emphasize therapies that are based on monoclonal antibodies (mAbs) and adoptive cell therapy, their mechanisms of action, their advantages, and their potential in combination therapy. We also highlight the clinical trials that have demonstrated both the therapeutic efficacy and the toxicities associated with each method. In addition, we summarize emerging targets that are now being evaluated as potential interventions for CRC. Finally, we discuss current challenges and future direction for the cancer immunotherapy field.
Collapse
Affiliation(s)
- Ensaf M. Al-Hujaily
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Batla S. Al-Sowayan
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Zeyad Alyousef
- Department of Surgery, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 14611, Saudi Arabia
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Farah Alammari
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| |
Collapse
|
9
|
Bioinformatic Analysis of the Effect of Silver Nanoparticles on Colorectal Cancer Cell Line. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6828837. [PMID: 35445138 PMCID: PMC9015850 DOI: 10.1155/2022/6828837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/09/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Colorectal cancer (CRC) is the most diagnosed cancer with the highest mortality rate each year globally. Although there are treatments for CRC, the development of resistance to therapies decreases the success of treatments. In vitro studies using the Caco-2 cell line have revealed the anticancer properties of silver nanoparticles (AgNPs) as a possible treatment for this disease. This study considered four researches that evaluated the proteomic profiles of cells of the Caco-2 line exposed to AgNPs. We performed a bioinformatics analysis to predict protein-protein interaction, hub genes, Gene Ontology (molecular function, biological process, and cellular components), KEGG pathways, analysis of expression, and immune cell infiltration. For these analyses, the STRING, DAVID, UALCAN, GEPIA2, and TISIDB databases were used. The results in Gene Ontology show that AgNPs cause a deregulation of genes related to cell-cell adhesion, the cytoplasm, the centriole, and carbon metabolism. Hub genes were identified, including GADPH, ENO1, EEF2, and ATP5A1, which showed differential expression in patients with adenocarcinoma of the colon and rectum. Additionally, the expression of the hub genes and immune cells was correlated. It was found that ATP5A1 and ENO1 were positively correlated with the infiltration of CD4+ T lymphocytes in colon adenocarcinoma and a negative correlation between GADPH and PDIA3 with the infiltration of NK cells and CD4+ T lymphocytes in rectal adenocarcinoma, respectively. In conclusion, the administration of AgNPs causes an alteration of biological processes, cellular components, metabolic pathways, deregulation of hub genes, and the activity of immune cells leading to a potential anticancer effect.
Collapse
|
10
|
Ebrahimpour M, Mohammadian M, Pourheydar B, Moradi Z, Behrouzkia Z. Effects of Radiotherapy in Combination With Irinotecan and 17-AAG on Bcl-2 and Caspase 3 Gene Expression in Colorectal Cancer Cells. J Lasers Med Sci 2022; 13:e9. [DOI: 10.34172/jlms.2022.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 11/14/2021] [Indexed: 12/24/2022]
Abstract
Introduction: In this study, the cytotoxic and anti-cancer effects of Irinotecan as a conventional chemotherapeutic agent compared to 17-(allyl amino)-17-demethoxygeldanamycin (17-AAG) as possible radiosensitizers in the HCT-116 cell line were investigated. Methods: HCT-116 cells were treated with various concentrations of irinotecan and 17-AAG and also irradiated with a 2-Gy of X-ray radiation. Then, the cell viability was examined by a water-soluble tetrazolium-1 assay after 24 hours. For single therapies and double and triple combination cases, IC50, 0.5×IC50 and 0.25×IC50 concentrations of each drug were selected respectively for a terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) assay and other tests. In treated and untreated cells, the caspase 3 and Bcl-2 gene expression ratios were evaluated by the real-time PCR method. Likewise, caspase 3 activity was detected with a colorimetric assay. Results: In all combined treatments, including 17-AAG- radiation, irinotecan - radiation, irinotecan -17-AAG, and irinotecan-17-AAG-radiation, decreased cellular viability and increased TUNEL positive cells were presented versus the control group (P<0.05). There were increased TUNEL positive cells in the triple combination, in concentrations of 0.25×IC50 of each drug, in comparison with single and double agent treatments. Moreover, in triple combination, the caspase 3 mRNA level and caspase 3 activity increased versus related single treatments. Likewise, in the irinotecan-17-AAG-radiation combined treatment and the 17-AAG-radiation double treatment, the Bcl-2 gene expression level decreased in comparison with single therapies. Conclusion: It can be indicated that the combination of chemo-radiotherapy versus single treatments has significant anti-cancer effects.
Collapse
Affiliation(s)
- Mahnaz Ebrahimpour
- Medical Physics Department, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahshid Mohammadian
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Bagher Pourheydar
- Neurophysiology Research Center, Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zhino Moradi
- Medical Physics Department, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zhaleh Behrouzkia
- Medical Physics Department, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
11
|
Talaat IM, Elemam NM, Saber-Ayad M. Complement System: An Immunotherapy Target in Colorectal Cancer. Front Immunol 2022; 13:810993. [PMID: 35173724 PMCID: PMC8841337 DOI: 10.3389/fimmu.2022.810993] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/14/2022] [Indexed: 12/26/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor and the second most fatal cancer worldwide. Several parts of the immune system contribute to fighting cancer including the innate complement system. The complement system is composed of several players, namely component molecules, regulators and receptors. In this review, we discuss the complement system activation in cancer specifically CRC and highlight the possible interactions between the complement system and the various TME components. Additionally, the role of the complement system in tumor immunity of CRC is reviewed. Hence, such work could provide a framework for researchers to further understand the role of the complement system in CRC and explore the potential therapies targeting complement activation in solid tumors such as CRC.
Collapse
Affiliation(s)
- Iman M. Talaat
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha Mousaad Elemam
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
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: 52] [Impact Index Per Article: 13.0] [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
|
13
|
Petre-Mandache CB, Margaritescu DN, Mitrut R, Kamal AM, Padureanu V, Cucu MG, Mitrut P. Risk Factors and Genetic Predisposition in Colorectal Cancer: A Study on Young and Old Adults. CURRENT HEALTH SCIENCES JOURNAL 2021; 47:84-88. [PMID: 34211752 PMCID: PMC8200615 DOI: 10.12865/chsj.47.01.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022]
Abstract
According to GLOBOCAN 2018 data Colorectal cancer (CRC) represents the third most commonly diagnosed cancer in the world and has the second-highest mortality rate. The incidence of CRC has been rising worldwide, the majority of cases being in developing countries mostly due to the adoption of an unhealthy lifestyle. The main driving factors behind CRC are a sedentary lifestyle, obesity, red meat consumption, alcohol, and tobacco; however, early detection screenings and standardized treatment options have reduced CRC mortality. Better family history and genetic testing can help those with a hereditary predisposition in taking preventative measures.
Collapse
Affiliation(s)
| | | | - Radu Mitrut
- Cardiology Department, University of Medicine and Pharmacy of Craiova, Romania, Emergency University Hospital Bucharest, Romania
| | - Adina-Maria Kamal
- Internal Medicine Department, University of Medicine and Pharmacy of Craiova, Romania
| | - Vlad Padureanu
- Internal Medicine Department, University of Medicine and Pharmacy of Craiova, Romania
| | - Mihai-Gabriel Cucu
- Medical Genetics Department, University of Medicine and Pharmacy of Craiova, Romania
| | - Paul Mitrut
- Internal Medicine Department, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
14
|
Network Pharmacology-Based Study on the Mechanism of Gegen Qinlian Decoction against Colorectal Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8897879. [PMID: 33294000 PMCID: PMC7714584 DOI: 10.1155/2020/8897879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/17/2020] [Accepted: 10/24/2020] [Indexed: 12/11/2022]
Abstract
Purpose Gegen Qinlian decoction (GQD) has been used to treat gastrointestinal diseases, such as diarrhea and ulcerative colitis (UC). A recent study demonstrated that GQD enhanced the effect of PD-1 blockade in colorectal cancer (CRC). This study used network pharmacology analysis to investigate the mechanisms of GQD as a potential therapeutic approach against CRC. Materials and Methods Bioactive chemical ingredients (BCIs) of GQD were collected from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. CRC-specific genes were obtained using the gene expression profile GSE110224 from the Gene Expression Omnibus (GEO) database. Target genes related to BCIs of GQD were then screened out. The GQD-CRC ingredient-target pharmacology network was constructed and visualized using Cytoscape software. A protein-protein interaction (PPI) network was subsequently constructed and analyzed with BisoGenet and CytoNCA plug-in in Cytoscape. Gene Ontology (GO) functional and the Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis for target genes were then performed using the R package of clusterProfiler. Results One hundred and eighteen BCIs were determined to be effective on CRC, including quercetin, wogonin, and baicalein. Twenty corresponding target genes were screened out including PTGS2, CCNB1, and SPP1. Among these genes, CCNB1 and SPP1 were identified as crucial to the PPI network. A total of 212 GO terms and 6 KEGG pathways were enriched for target genes. Functional analysis indicated that these targets were closely related to pathophysiological processes and pathways such as biosynthetic and metabolic processes of prostaglandins and prostanoids, cytokine and chemokine activities, and the IL-17, TNF, Toll-like receptor, and nuclear factor-kappa B (NF-κB) signaling pathways. Conclusion The study elucidated the “multiingredient, multitarget, and multipathway” mechanisms of GQD against CRC from a systemic perspective, indicating GQD to be a candidate therapy for CRC treatment.
Collapse
|
15
|
Reszegi A, Horváth Z, Karászi K, Regős E, Postniková V, Tátrai P, Kiss A, Schaff Z, Kovalszky I, Baghy K. The Protective Role of Decorin in Hepatic Metastasis of Colorectal Carcinoma. Biomolecules 2020; 10:1199. [PMID: 32824864 PMCID: PMC7465536 DOI: 10.3390/biom10081199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/24/2022] Open
Abstract
Decorin, the prototype member of the small leucine-rich proteoglycan gene family of extracellular matrix (ECM) proteins, acts as a powerful tumor suppressor by inducing the p21Waf1/Cip1 cyclin-dependent kinase inhibitor, as well as through its ability to directly bind and block the action of several tyrosine kinase receptors. Our previous studies suggested that the lack of decorin promotes hepatic carcinogenesis in mice. Based on this, we set out to investigate whether excess decorin may protect against the liver metastases of colon carcinoma. We also analyzed the effect of decorin in tissue microarrays of human colon carcinoma liver metastasis and examined whether the tumor cells can directly influence the decorin production of myofibroblasts. In humans, low levels of decorin in the liver facilitated the development of colon carcinoma metastases in proportion with more aggressive phenotypes, indicating a possible antitumor action of the proteoglycan. In vitro, colon carcinoma cells inhibited decorin expression in LX2 hepatic stellate cells. Moreover, liver-targeted decorin delivery in mice effectively attenuated metastasis formation of colon cancer. Overexpressed decorin reduced the activity of multiple receptor tyrosine kinases (RTKs) including the epidermal growth factor receptor (EGFR), an important player in colorectal cancer (CRC) pathogenesis. Downstream of that, we observed weakened signaling of ERK1/2, PLCγ, Akt/mTOR, STAT and c-Jun pathways, while p38 MAPK/MSK/CREB and AMPK were upregulated culminating in enhanced p53 function. In conclusion, decorin may effectively inhibit metastatic tumor formation in the liver.
Collapse
Affiliation(s)
- Andrea Reszegi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | - Zsolt Horváth
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | - Katalin Karászi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | - Eszter Regős
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | - Victoria Postniková
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | | | - András Kiss
- 2nd Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary; (A.K.); (Z.S.)
| | - Zsuzsa Schaff
- 2nd Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary; (A.K.); (Z.S.)
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői Street 26, H-1085 Budapest, Hungary; (A.R.); (Z.H.); (K.K.); (E.R.); (V.P.); (I.K.)
| |
Collapse
|