1
|
Zafar A, Khalid M, Alsaidan OA, Mujtaba MA. Exploring the molecular pathways of advanced rectal cancer: A focus on genetic, RNA, and biological technique. Pathol Res Pract 2025; 270:155956. [PMID: 40215670 DOI: 10.1016/j.prp.2025.155956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 05/20/2025]
Abstract
Colorectal cancer (CRC) is the third most frequently diagnosed cancer, with rectal cancer (RC) accounting for approximately 35 % of cases, posing a significant health burden. The early phase of R progression is characterized by the accumulation of genetic and epigenetic changes that promote cell growth. These rapidly dividing cells form a benign adenoma, which can eventually transform into malignant tumors and metastasize to other organs. Among the key molecular alterations, a mutation in the Wnt/β-catenin signaling pathway plays a crucial role. Additionally, BRAF mutation contributes to 8-10 % of CRC cases, while mutation in PIK3C pathways is responsible for 20-25 % of cases. The RC involves complex biological mechanisms. This review article highlights the pivotal role of mRNA in diagnosing and predicting the prognosis of RC, explores the various functions of non-coding RNAs (ncRNA,s), and examines the impact of RNA editing and modification on the progression of tumor genesis. Furthermore, we discuss the cellular signaling pathways and microenvironment interaction and pathways like PI3K/Akt/mTOR and Wnt/β-catenin. Advancements in molecular, RNA, and genetic research have evolved the treatment of cancer. Techniques like next-generation sequencing have tremendously opened the biological field of research. Along with this, techniques like CRISPR/Cas9 aid in the developing therapeutic strategies. Proteomics and metabolomics approach further contribute to novel research direction in oncology.
Collapse
Affiliation(s)
- Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia.
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Md Ali Mujtaba
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Dash P, Yadav V, Das B, Satapathy SR. Experimental toolkit to study the oncogenic role of WNT signaling in colorectal cancer. Biochim Biophys Acta Rev Cancer 2025:189354. [PMID: 40414319 DOI: 10.1016/j.bbcan.2025.189354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Colorectal cancer (CRC) is linked to the WNT/β-catenin signaling as its primary driver. Aberrant activation of WNT/β-catenin signaling is closely correlated with increased incidence, malignancy, poorer prognosis, and even higher cancer-related death. Research over the years has postulated various experimental models that have facilitated an understanding of the complex mechanisms underlying WNT signaling in CRC. In the present review, we have comprehensively summarized the in vitro, in vivo, patient-derived, and computational models used to study the role of WNT signaling in CRC. We discuss the use of CRC cell lines and organoids in capturing the molecular intricacies of WNT signaling and implementing xenograft and genetically engineered mouse models to mimic the tumor microenvironment. Patient-derived models, including xenografts and organoids, provide valuable insights into personalized medicine approaches. Additionally, we elaborated on the role of computational models in simulating WNT signaling dynamics and predicting therapeutic outcomes. By evaluating the advantages and limitations of each model, this review highlights the critical contributions of these systems to our understanding of WNT signaling in CRC. We emphasize the need to integrate diverse model systems to enhance translational research and clinical applications, which is the primary goal of this review.
Collapse
Affiliation(s)
- Pujarini Dash
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Biswajit Das
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, USA
| | - Shakti Ranjan Satapathy
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
3
|
Peng W, Zeng Z. Epigenetic Activation of PTCD3 Promotes CRC Glutamine Metabolism and Metastasis via IGF2BP2-Mediated SLC38A2 m6A Modification. FASEB J 2025; 39:e70558. [PMID: 40304977 DOI: 10.1096/fj.202401788rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
Cancer cells undergo metabolic reprogramming, shifting their programs toward aerobic glycolysis and enhanced glutaminolysis to fulfill the requirements of rapid proliferation. Investigating the mechanisms underlying glutaminolysis and its connection with colorectal cancer (CRC) could aid in identifying novel therapeutic targets. PTCD3, a mitochondrial RNA-binding protein, is implicated in cancer progression, and IGF2BP2 regulates mRNA stability and translation. SLC38A2, a key transporter in glutamine metabolism, plays a crucial role in supporting cancer cell growth. This study aims to develop inhibitors of PTCD3 or SLC38A2 to prevent metabolic changes in cancer cells that facilitate rapid growth and metastasis in CRC. RT-qPCR, western blot, IHC, and IF staining assays confirmed the targeted gene and protein expression. Proliferation, migration, and invasion were evaluated using CCK-8 assay, scratch assay, and Transwell assay, respectively. Co-IP, RIP, and dual-luciferase assays were conducted to investigate the interactions among PTCD3, IGF2BP2, and SLC38A2. A CRC xenograft nude mice model was established for additional in vivo validation. PTCD3 was upregulated in CRC and positively correlated with GLS1. PTCD3 knockdown suppressed CRC cell glutaminolysis, thereby inhibiting CRC migration and invasion. PTCD3 promoted SLC38A2 mRNA stability in an IGF2BP2-dependent manner. KAT2A promoted the expression of PTCD3 by increasing H3K27 acetylation. The inhibitory effect of PTCD3 depletion on the glutaminolysis of CRC cells, as well as CRC cell proliferation and migration, was reversed by SLC38A2 overexpression. The in vivo mouse experiments further confirmed that silencing of PTCD3 inhibited CRC tumor growth. In summary, KAT2A upregulates PTCD3 expression by promoting H3K27 acetylation, which promotes glutaminolysis and metastasis in CRC via enhancing SLC38A2 mRNA stability in an IGF2BP2-dependent manner.
Collapse
Affiliation(s)
- Weihui Peng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zhijun Zeng
- Department of Geriatric Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
4
|
Song W, Rahimian N, Hasanzade Bashkandi A. GRP78: A new promising candidate in colorectal cancer pathogenesis and therapy. Eur J Pharmacol 2025; 995:177308. [PMID: 39870235 DOI: 10.1016/j.ejphar.2025.177308] [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: 10/02/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) is a significant global health challenge, marked by varying incidence and mortality rates across different regions. The pathogenesis of CRC involves multiple stages, including initiation, promotion, progression, and metastasis, influenced by genetic and epigenetic factors. The chaperone protein glucose-regulated protein 78 (GRP78), crucial in regulating the unfolded protein response (UPR) during endoplasmic reticulum (ER) stress, plays a pivotal role in CRC pathogenesis. This review discusses the expression profile of GRP78 in CRC, highlighting its potential as a prognostic biomarker and its role in modulating the cellular mechanisms of CRC, including ER response regulation, cell proliferation, migration and invasion. The complex molecular interactions of GRP78 with key signaling pathways such as protein kinase B (Akt), Wnt, protein kinase R-like ER kinase (PERK), vascular endothelial growth factor (VEGF), and Kirsten rat sarcoma virus (Kras) are explored, elucidating its contributions to tumor survival, proliferation, invasion, and chemoresistance. GRP78's involvement in autophagy, glycolysis, and immune regulation further underscores its importance in CRC progression. The review also covers the therapeutic potential of targeting GRP78 in CRC, examining various natural products like curcumin, epigallocatechin gallate (EGCG), and aloe-emodin, which modulate GRP78 expression and activity. Additionally, GRP78's role in mediating resistance to chemotherapeutic agents like 5-fluorouracil (5-FU) and oxaliplatin is discussed, emphasizing its significance in the development of resistance mechanisms in CRC. In conclusion, GRP78 emerges as a central player in CRC pathogenesis and a promising target for therapeutic interventions aimed at improving treatment outcomes and overcoming chemoresistance in colorectal cancer.
Collapse
Affiliation(s)
- Wang Song
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China.
| | - Neda Rahimian
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
5
|
Yii CY, Toh DE, Chen TA, Hsu WL, Lai HJ, Wang YC, Liu CR, Kuo YC, Young SH, Chang FM, Lin C. Use of artificial intelligence to measure colorectal polyp size without a reference object. Endosc Int Open 2025; 13:a25561836. [PMID: 40376021 PMCID: PMC12080523 DOI: 10.1055/a-2556-1836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/27/2025] [Indexed: 05/18/2025] Open
Abstract
Background and study aims Polyp size is crucial for determining colonoscopy surveillance intervals. We present an artificial intelligence (AI) model for colorectal polyp size measurement without a reference object. Methods The regression model for polyp size estimation was developed using outputs from two SegFormer models, segmentation and depth estimation. Initially built on colonoscopic images of polyp phantoms, the model underwent transfer learning with 1,304 real-world images. Testing was conducted on 178 images from 52 polyps, independent of the training set, using a snare as the ground truth for size comparison with the AI-based model. Polyps were classified into three size groups: ≤ 5 mm, 5-10 mm, and ≥ 10 mm. Error rates were calculated to evaluate discrepancies in actual size values between the AI model and the snare method. Precision indicated the positive predictive value per size group and recall and Bland-Altman were also conducted. Results The Bland-Altman analysis showed a mean bias of -0.03 mm between methods, with limits of agreement from -1.654 mm to 1.596 mm. AI model error rates for actual size discrepancies were 10.74%, 12.36%, and 9.89% for the ≤ 5 mm, 5-10 mm, and ≥ 10 mm groups, respectively, averaging 11.47%. Precision values were 0.870, 0.911, and 0.857, with overall recall of 0.846. Conclusions Our study shows that colorectal polyp size measurement by AI model is practical and clinically useful, exhibiting low error rates and high precision. AI shows promise as an accurate tool for measurement without the need for a reference object during screening colonoscopy.
Collapse
Affiliation(s)
- Chin-Yuan Yii
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
- Department of Biomedical Sciences and Engineering, National Central University, Zhongli, Taiwan
| | - Ding-Ek Toh
- Department of Gastroenterology, Flinders Medical Centre, Bedford Park, Australia
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Tzu-An Chen
- Division of Colorectal Surgery, Department of Surgery, Landseed International Medical Group, Taoyuan, Taiwan
| | - Wei-Lun Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
- Institute of Computer Science and Information Engineering, National Central University, Zhongli, Taiwan
| | - Huang-Jen Lai
- Division of Colorectal Surgery, Department of Surgery, Landseed International Medical Group, Taoyuan, Taiwan
| | - Yin-Chen Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
| | - Chang-Ru Liu
- Nursing Department, Landseed International Medical Group, Taoyuan, Taiwan
| | - Yow-Chii Kuo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
| | - Shih-Hao Young
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
| | - Fu-Ming Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Medical Group, Taoyuan, Taiwan
| | - Chen Lin
- Department of Biomedical Sciences and Engineering, National Central University, Zhongli, Taiwan
| |
Collapse
|
6
|
Yoon J, Moon H, Jeon Y, Choe S, Yoon H. Signature Gene Mutations in Colorectal Cancer: Potential Neoantigens for Cancer Vaccines. Int J Mol Sci 2025; 26:4559. [PMID: 40429703 DOI: 10.3390/ijms26104559] [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: 04/08/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Colorectal cancer (CRC), the third most common cancer worldwide, is one of the deadliest cancers. CRC is known as a cold tumor, characterized by a low immune response that makes it difficult for immune cells to infiltrate and exhibits strong resistance to immunotherapy with checkpoint inhibition. This restricted response is largely attributed to signature gene mutations including mismatch repair (MMR) genes, KRAS, BRAF, APC, and TP53, which are also the main oncogenes in CRC. Mutated signature genes continuously upregulate abnormal signaling pathways, leading to excessive proliferation, cancer progression, and metastasis. Furthermore, it reorganizes the tumor microenvironment (TME) by recruiting immunosuppressive cells. However, the mutation can produce neoantigens that can provoke an immune response, making it a potential target for immunotherapy. In particular, cancer vaccines that leverage the strong neoantigenic properties of these mutations are considered promising for overcoming immune resistance and eliciting anti-tumor responses. In this review, we will describe signature gene mutations in CRC and focus on cancer vaccines targeting these mutations as potential therapies for CRC.
Collapse
Affiliation(s)
- Jaegoo Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Haeun Moon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Yuna Jeon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Soohyun Choe
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| |
Collapse
|
7
|
Teixeira N, Baião A, Dias S, Sarmento B. The progress and challenges in modeling colorectal cancer and the impact on novel drug discovery. Expert Opin Drug Discov 2025; 20:565-574. [PMID: 39282980 DOI: 10.1080/17460441.2024.2404238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/10/2024] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality worldwide. This disease is complex and heterogeneous, influenced by a variety of genetic, epigenetic, and environmental factors that drive CRC initiation and progression. Despite advances in therapeutic strategies, the five-year survival rate for metastatic CRC is alarmingly low. Traditional two-dimensional (2D) cell culture systems have been the foundation of cancer research, but their inability to replicate the complex tumor microenvironment (TME) limits their effectiveness. AREAS COVERED This paper explores the evolution of CRC models, starting with the limitations of traditional 2D cell culture systems and the significant advancements offered by 3D models. Additionally, it highlights 3D bioprinting and on-chip CRC models, which have enhanced the ability to mimic in vivo conditions. EXPERT OPINION The transition to advanced 3D models represents a pivotal shift in CRC research, offering considerable improvements over the established 2D models. These models hold promise for the development of patient-specific models that better mimic in vivo conditions. However, the inherent complexity of CRC continues to pose challenges in developing models that can fully capture the disease's multifaceted nature. This complexity and high costs associated with these technologies, along with the need for standardized protocols, pose significant challenges to their widespread adoption.
Collapse
Affiliation(s)
- Natália Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sofia Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- CESPU - IUCS - Instituto Universitário de Ciências da Saúde, Gandra, Portugal
| |
Collapse
|
8
|
Ramesh A, Srinivasan D, Subbarayan R, Chauhan A, Krishnamoorthy L, Kumar J, Krishnan M, Shrestha R. Enhancing Colorectal Cancer Treatment: The Role of Bifidobacterium in Modulating Gut Immunity and Mitigating Capecitabine-Induced Toxicity. Mol Nutr Food Res 2025; 69:e70023. [PMID: 40109200 DOI: 10.1002/mnfr.70023] [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: 12/11/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related mortality globally and presents significant challenges in treatment and patient care. Capecitabine, a widely used prodrug of 5-fluorouracil (5-FU), offers targeted delivery with reduced systemic toxicity compared to traditional chemotherapies. However, capacitabine is associated with adverse effects, such as hand-foot syndrome, gastrointestinal issues, and mucositis. Emerging evidence suggests that probiotics, particularly Bifidobacterium, play a pivotal role in gut microbiota modulation, promoting anti-inflammatory cytokines and short-chain fatty acids, such as butyrate, which possess both intestinal protective and anti-cancer properties. In this review, we explored the potential of Bifidobacterium to improve chemotherapy outcomes by mitigating inflammation and enhancing mucosal immunity in CRC patients. Furthermore, we demonstrated in silico approaches, including molecular docking and protein-protein interaction analysis, for Bifidobacterium and Toll-like receptor 2 (TLR-2), a key mediator of intestinal immunity. Docking results revealed strong binding affinity, suggesting the activation of anti-inflammatory pathways. Notably, this interaction enhanced IL-10 production while reducing pro-inflammatory cytokines, such as IL-6 and TNF-α, fostering gut homeostasis and mitigating chronic inflammation, a key driver of CRC progression. Therefore, future research should focus on personalized probiotics and validating their synergy with chemotherapy and immunotherapy to improve CRC treatment outcomes.
Collapse
Affiliation(s)
- Aswathi Ramesh
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Jeevan Kumar
- Department of Biomedical Sciences, The Apollo University, Chittoor, Andhra Pradesh, India
| | - Madhan Krishnan
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rupendra Shrestha
- Department of Natural and Applied Sciences, Nexus Institute of Research and Innovation (NIRI), Lalitpur, Nepal
| |
Collapse
|
9
|
Alhosani F, Alhamidi RS, Ilce BY, Altaie AM, Ali N, Hamad AM, Künstner A, Khandanpour C, Busch H, Al-Ramadi B, Harati R, Sayed K, AlFazari A, Bendardaf R, Hamoudi R. Transcriptome-Wide Analysis and Experimental Validation from FFPE Tissue Identifies Stage-Specific Gene Expression Profiles Differentiating Adenoma, Carcinoma In-Situ and Adenocarcinoma in Colorectal Cancer Progression. Int J Mol Sci 2025; 26:4194. [PMID: 40362431 PMCID: PMC12071244 DOI: 10.3390/ijms26094194] [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: 03/21/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Colorectal cancer (CRC) progression occurs through three stages: adenoma (pre-cancerous lesion), carcinoma in situ (CIS) and adenocarcinoma, with tumor stage playing a pivotal role in the prognosis and treatment outcomes. Despite therapeutic advancements, the lack of stage-specific biomarkers hinders the development of accurate diagnostic tools and effective therapeutic strategies. This study aims to identify stage-specific gene expression profiles and key molecular mechanisms in CRC providing insights into molecular alterations across disease progression. Our methodological approach integrates the use of absolute gene set enrichment analysis (absGSEA) on formalin-fixed paraffin-embedded (FFPE)-derived transcriptomic data, combined with large-scale clinical validation and experimental confirmation. A comparative whole transcriptomic analysis (RNA-seq) was performed on FFPE samples including adenoma (n = 10), carcinoma in situ (CIS) (n = 8) and adenocarcinoma (n = 11) samples. Using absGSEA, we identified significant cellular pathways and putative molecular biomarkers associated with each stage of CRC progression. Key findings were then validated in a large independent CRC patient cohort (n = 1926), with survival analysis conducted from 1336 patients to assess the prognostic relevance of the candidate biomarkers. The key differentially expressed genes were experimentally validated using real-time PCR (RT-qPCR). Pathway analysis revealed that in CIS, apoptotic processes and Wnt signaling pathways were more prominent than in adenoma samples, while in adenocarcinoma, transcriptional co-regulatory mechanisms and protein kinase activity, which are critical for tumor growth and metastasis, were significantly enriched compared to adenoma. Additionally, extracellular matrix organization pathways were significantly enriched in adenocarcinoma compared to CIS. Distinct gene signatures were identified across CRC stages that differentiate between adenoma, CIS and adenocarcinoma. In adenoma, ARRB1, CTBP1 and CTBP2 were overexpressed, suggesting their involvement in early tumorigenesis, whereas in CIS, RPS3A and COL4A5 were overexpressed, suggesting their involvement in the transition from benign to malignant stage. In adenocarcinoma, COL1A2, CEBPZ, MED10 and PAWR were overexpressed, suggesting their involvement in advanced disease progression. Functional analysis confirmed that ARRB1 and CTBP1/2 were associated with early tumor development, while COL1A2 and CEBPZ were involved in extracellular matrix remodeling and transcriptional regulation, respectively. Experimental validation with RT-qPCR confirmed the differential expression of the candidate biomarkers (ARRB1, RPS3A, COL4A5, COL1A2 and MED10) across the three CRC stages reinforcing their potential as stage-specific biomarkers in CRC progression. These findings provide a foundation to distinguish between the CRC stages and for the development of accurate stage-specific diagnostic and prognostic biomarkers, which helps in the development of more effective therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Faisal Alhosani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
- Forensic Laboratory Department, Sharjah Police Headquarters, Sharjah P.O. Box 1965, United Arab Emirates
| | - Reem Sami Alhamidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Burcu Yener Ilce
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Muayad Altaie
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Nival Ali
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Mohamed Hamad
- College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates;
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Cyrus Khandanpour
- Department of Hematology and Oncology, University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, University of Lübeck, 23562 Lübeck, Germany;
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Rania Harati
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Kadria Sayed
- Department of Pathology and Laboratory Medicine, American Hospital Dubai, Dubai P.O. Box 3050, United Arab Emirates;
| | - Ali AlFazari
- Mediclinic Welcare Hospital, Dubai P.O. Box 31500, United Arab Emirates;
| | - Riyad Bendardaf
- Oncology Unit, University Hospital Sharjah, Sharjah P.O. Box 72772, United Arab Emirates;
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
| |
Collapse
|
10
|
Xie DL, Fan JH, Fan CJ, Gao YH, Cheng JP. A randomized, controlled trial of oral sulfate solution versus polyethylene glycol for bowel preparation for colonoscopy. BMC Gastroenterol 2025; 25:292. [PMID: 40269724 PMCID: PMC12020202 DOI: 10.1186/s12876-025-03885-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The quality of colonoscopy is significantly influenced by the effectiveness of bowel preparation. In this study, we aimed to evaluate the efficacy, safety, and tolerability of bowel cleansing between a new oral sulfate solution (OSS) and standard polyethylene glycol electrolyte powder (PEG). METHODS This single center, randomized, superiority study recruited 679 outpatients who were assigned to either the new OSS group (Group A) or standard PEG group (Group B). The quality of bowel cleansing was evaluated using the Boston Bowel Preparation Scale (BBPS) and compared between the two groups. Furthermore, data pertaining to the duration of bowel preparation, patient tolerability, and the occurrence of adverse events were also analyzed. RESULTS According to BBPS scores, group A demonstrated significantly higher bowel preparation cleanliness than group B. Additionally, group A achieved superior bowel cleansing, as evidenced by a greater proportion of patients with BBPS scores ≥ 8 compared to group B (75.3% vs. 55.2%, P < 0.05). No severe adverse events were reported during examinations in either group. CONCLUSIONS The magnesium sulfate, sodium sulfate, and potassium sulfate concentrated oral solution is a novel, safe, and effective bowel preparation for colonoscopy. TRIAL REGISTRATION This study was registered in the Chinese Clinical Trial Registry on 20/02/2024 (clinical trial registration number: ChiCTR2400081004).
Collapse
Affiliation(s)
- Dong-Ling Xie
- Department of Gastroenterology and Oncology, Civil Aviation General Hospital, No. 76 Chaoyang Road, Chaoyang District, Beijing, 100123, China
| | - Jin-Hui Fan
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing, China
| | - Chan-Juan Fan
- Department of Gastroenterology and Oncology, Civil Aviation General Hospital, No. 76 Chaoyang Road, Chaoyang District, Beijing, 100123, China
| | - Ying-Hui Gao
- Department of Gastroenterology and Oncology, Civil Aviation General Hospital, No. 76 Chaoyang Road, Chaoyang District, Beijing, 100123, China
| | - Jian-Ping Cheng
- Department of Gastroenterology and Oncology, Civil Aviation General Hospital, No. 76 Chaoyang Road, Chaoyang District, Beijing, 100123, China.
| |
Collapse
|
11
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [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: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Li KJ, Zhang ZY, Wang K, Sulayman S, Zeng XY, Liu J, Chen Y, Zhao ZL. Prognostic scoring system using inflammation- and nutrition-related biomarkers to predict prognosis in stage I-III colorectal cancer patients. World J Gastroenterol 2025; 31:104588. [PMID: 40248373 PMCID: PMC12001188 DOI: 10.3748/wjg.v31.i14.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/22/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy that has become a global burden. The prognostic prediction of CRC patients on the basis of inflammatory biomarkers and nutritional biomarkers has shown some potential but has not been fully explored. AIM To develop and validate a prognostic model for CRC based on inflammation and nutrition-related biomarkers and to evaluate its predictive value for patient outcomes. METHODS Patients were randomized at a 3:2 ratio into a training cohort (n = 282) or a validation cohort (n = 188). To identify the optimal prognostic factors for constructing the risk score (RS), LASSO Cox regression analysis was conducted. The association between the RS and overall survival (OS) was evaluated using receiver operating characteristic (ROC) curves and Kaplan-Meier (K-M) survival analysis. Independent risk factors were screened by multivariate Cox regression analysis. Nomograms were constructed and validated on the basis of these factors. RESULTS In the training cohort, univariate analysis of all the inflammatory and nutritional biomarkers demonstrated some predictive value. A LASSO-Cox analysis included four biomarkers and constructed an RS. Through ROC analysis, the area under the prognostic curve was 0.795. K-M survival curve analyses revealed that the five-year OS was significantly greater in the Low-RS group than in the High-RS group (P < 0.001). Multivariate analysis demonstrated that the degree of differentiation (P = 0.001), degree of nerve invasion (P = 0.022), and RS (P < 0.001) were independent risk factors. We constructed a nomogram to predict the OS of CRC patients and validated it in a separate cohort. The calibration curve showed high accuracy. Additionally, decision curve analysis for 1-year, 3-year, and 5-year survival probabilities indicated significant clinical utility in predicting survival outcomes. CONCLUSION This study developed a nomogram based on the RS to predict the OS of CRC patients. This nomogram can guide treatment decisions and enable the formulation of personalized follow-up strategies on the basis of predicted recurrence risk, aiming to improve long-term prognosis.
Collapse
Affiliation(s)
- Ke-Jin Li
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Zi-Yi Zhang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Kuan Wang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Subinur Sulayman
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiang-Yue Zeng
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Juan Liu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Yi Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Ze-Liang Zhao
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
13
|
Wang Z, Li T, Sun M, Liu N, Zhang H, Feng Z, Lei N. Metabolomics- and Proteomics-Based Disease Diagnostic Classifier Model for the Prediction and Diagnosis of Colorectal Carcinoma. J Proteome Res 2025; 24:2096-2111. [PMID: 40108892 PMCID: PMC11977543 DOI: 10.1021/acs.jproteome.5c00010] [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: 01/12/2025] [Revised: 02/28/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is a leading cause of cancer-related deaths globally. Diagnostic biomarkers are essential for risk stratification and early detection, potentially enhancing patient survival. Our study aimed to explore the potential biomarkers of CRC at the protein and metabolic levels. METHODS Blood serum from CRC patients and healthy controls was analyzed using metabolomic and proteomic techniques. A conjoint analysis was conducted, and samples were split into training and validation sets (7:3 ratio) to develop and evaluate a disease diagnosis classifier model. Immunohistochemistry (IHC) analyses were conducted to validate the results. RESULTS We identified 631 differential metabolites and 61 differentially expressed proteins (DEPs) in CRC, involved in pathways such as arginine and proline metabolism, central carbon metabolism in cancer, and signaling pathways including TGF-β, mTOR, PI3K-Akt, and others. Key proteins (CILP2, SLC3A2, EXTL2, hydroxypyruvate isomerase (HYI), ENPEP, LRG1, CTSS, thyrotropin-releasing hormone-degrading ectoenzyme (TRHDE), SELE, and HSPA1A) showed significant expression differences between CRC patients and controls. IHC results showed that compared with the paracancerous tissues, the expression of CILP2, EXTL2, and HYI was significantly downregulated in the CRC tissues (P < 0.05). The classifier model, comprising l-arginine, Harden-Young ester, l-aspartic acid, oxoglutaric acid, l-proline, octopine, l-valine, and progesterone, achieved AUC values of 0.998 and 0.914 in training and validation data sets, respectively. CONCLUSIONS The identified metabolites and DEPs are promising CRC biomarkers. The developed classifier model based on eight metabolites demonstrates high accuracy for CRC assessment and diagnosis.
Collapse
Affiliation(s)
- Zhaorui Wang
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Tianyuan Li
- Department
of Gastroenterology, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Mengyao Sun
- Department
of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Na Liu
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Haozhe Zhang
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Zhikun Feng
- Department
of Pathology, The Fifth Clinical Medical
College of Henan University of Chinese Medicine (Zhengzhou People’s
Hospital), Zhengzhou, Henan 450000, China
| | - Ningjing Lei
- Department
of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
14
|
Ma G, Wu X, Agudamu, Shen J, Bao Z, Yang Z. M2 Macrophage-Extracellular Vesicle-Derived lncRNA-NEAT1 Regulates miR-204-5p/RRS1-mediated Cell Cycle to Promote the Occurrence and Development of Colorectal Cancer. J Biochem Mol Toxicol 2025; 39:e70168. [PMID: 40165491 DOI: 10.1002/jbt.70168] [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: 06/25/2024] [Revised: 12/06/2024] [Accepted: 01/26/2025] [Indexed: 04/02/2025]
Abstract
Colorectal cancer (CRC) is a prevalent malignancy of the digestive system. Here, we explored the role of M2 macrophage-derived extracellular vesicles (EVs) carrying long non-coding RNA-nuclear paraspeckle assembly transcript 1 (lncRNA-NEAT1) in promoting CRC progression via regulation of the miR-204-5p/regulator of ribosome synthesis 1 (RRS1) axis and cell cycle dynamics. Firstly, we differentiated WTHP-1 cells into M0 and M2 macrophages and transfected M2 macrophages with sh-NEAT1 lentivirus plasmids. EVs were isolated from M2 macrophages and administered to SW480 cells along with miR-204-5p inhibitors or si-RRS1 for 24 h. M2-EVs-derived lncRNA-NEAT1 enhanced CRC cell proliferation, migration, invasion, and viability while reducing apoptosis. This was accompanied by increased expression of RRS1, WEE1 G2 checkpoint kinase (WEE1), cyclin-dependent kinase 1 (CDK1), and CyclinB1, reduced miR-204-5p levels, and a lower proportion of cells in the G2/M phase. Knockdown of lncRNA-NEAT1 in M2 macrophages reversed these effects. Mechanistically, M2-EVs-derived lncRNA-NEAT1 functioned as a competing endogenous RNA (ceRNA), sponging miR-204-5p to upregulate RRS1 expression. In summary, M2 macrophage-derived EVs carrying lncRNA-NEAT1 promote CRC development by modulating the miR-204-5p/RRS1 axis, influencing the cell cycle and apoptosis. These findings provide insights into the tumor-promoting mechanisms of macrophage-derived EVs in CRC.
Collapse
Affiliation(s)
- Guorui Ma
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xinlin Wu
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Agudamu
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Junjie Shen
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhe Bao
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhiwen Yang
- Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
15
|
Nourbakhsh ST, Mirzaei SA, Mohamadhashem F, Naghizadeh MM, Razavi AN, Mansoori Y, Daraei A, Mohamadhashem F. Pathological expression of mitochondrial genome-derived circRNA SCAR/mc-COX2 and its ceRNA network in colorectal cancer: implications for clinical significance. BMC Cancer 2025; 25:466. [PMID: 40082804 PMCID: PMC11907809 DOI: 10.1186/s12885-025-13886-7] [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: 01/09/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Mitochondrial-encoded circular RNAs (mecciRNAs) are a newly discovered class of mitochondrial-encoded non-coding RNAs (mt-ncRNAs) that play important biological roles in the cell. This study aimed to examine the expression profile of SCAR/mc-COX2 (has_circ_0089762) in colorectal cancer (CRC) and its relationship with clinicopathological variables. Furthermore, to better understand SCAR/mc-COX2's functional role in CRC, we constructed a competing endogenous RNA (ceRNA) network. METHODS Quantitative real-time PCR (qRT-PCR) was employed to analyze the expression levels of SCAR/mc-COX2 in 40 pairs of CRC samples, consisting of 40 tumor samples and 40 adjacent non-tumoral samples from patients. The ceRNA regulatory network was constructed using online bioinformatics tools. Furthermore, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) enrichment analysis were conducted using the Enrichr database. RESULTS The results demonstrated a significant decrease in SCAR/mc-COX2 expression in tumor tissues compared to adjacent non-tumoral tissues (p-value<0.05). In another finding, a significant relationship was observed between pathological T staging and the expression status of SCAR/mc-COX2 (p-value=0.02). Additionally, the Receiver Operating Characteristic (ROC) curve analysis revealed that SCAR/mc-COX2 had an area under the curve (AUC) of 0.77, with 80% sensitivity and 75% specificity. Finally, a ceRNA regulatory network including SCAR/mc-COX2, 5 miRNA, and 9 mRNAs was found. CONCLUSION These findings suggest that SCAR/mc-COX2 may act as a tumor suppressor in CRC, and its dysregulation could play a crucial role in the pathophysiology of this cancer. The significant association with pathological T staging and its robust diagnostic performance (AUC = 0.77, sensitivity = 80%, specificity = 75%) highlight its potential as a novel biomarker for CRC detection and prognosis. Further functional studies are required to elucidate its precise role in CRC tumorigenesis and clinical applicability.
Collapse
Affiliation(s)
- Seyed Taha Nourbakhsh
- Department of Medical Genetics, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Mohamadhashem
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amir Nader Razavi
- Iran National Tumor Bank, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Faezeh Mohamadhashem
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
16
|
Mitea G, Iancu IM, Schröder V, Roșca AC, Iancu V, Crețu RM, Mireșan H. Therapeutic Potential of Prunus Species in Gastrointestinal Oncology. Cancers (Basel) 2025; 17:938. [PMID: 40149274 PMCID: PMC11940452 DOI: 10.3390/cancers17060938] [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: 02/05/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Gastrointestinal tract cancers represent a significant worldwide health concern, accounting for almost one-third of cancer-related deaths. The existing chemotherapy drugs used in gastrointestinal cancers are ineffective, so prognosis is poor, recurrence and metastasis rates are high, and survival time remains short, necessitating the development of novel antitumor drugs that exhibit low toxicity and less potential for the development of drug resistance. This challenge is considerable, but evidence from the past decades supports the medicinal properties and functionalities of bioactive compounds such as flavonoids and acid phenolics with anticancer activities. Our purpose was to find data on the relationship between gastrointestinal cancer and bioactive compounds from Prunus species, focusing on their molecular mechanisms of action. RESULTS Studies highlight the potential of bioactive compounds from Prunus species to modulate the cancer cell signaling pathways involved in gastrointestinal tumorigenesis. CONCLUSIONS The studies reviewed suggest that polyphenols from Prunus species exhibit promising gastrointestinal anticancer activities and could represent an adjunctive therapeutic strategy in cancer treatment. Further studies are necessary to validate these compounds' therapeutic potential and their feasibility as cost-effective treatments for cancer.
Collapse
Affiliation(s)
- Gabriela Mitea
- Department of Pharmacology, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania;
| | - Irina Mihaela Iancu
- Department of Toxicology, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania;
| | - Verginica Schröder
- Department of Cellular and Molecular Biology, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania
| | - Adrian Cosmin Roșca
- Department of Analysis and Quality Control of Drugs, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania;
| | - Valeriu Iancu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania;
| | - Ruxandra-Mihaela Crețu
- National Institute of Research and Development for Biological Sciences, “Stejarul” Biological Research Centre, 060031 Bucharest, Romania;
| | - Horațiu Mireșan
- Department of Toxicology, Faculty of Pharmacy, Ovidius University of Constanta, 900470 Constanta, Romania;
| |
Collapse
|
17
|
Imtiaz I, Schloss J, Bugarcic A. Interplay Between Traditional and Scientific Knowledge: Phytoconstituents and Their Roles in Lung and Colorectal Cancer Signaling Pathways. Biomolecules 2025; 15:380. [PMID: 40149916 PMCID: PMC11940637 DOI: 10.3390/biom15030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Natural plant products have been used for cancer treatment since ancient times and continue to play a vital role in modern anticancer drug development. However, only a small fraction of identified medicinal plants has been thoroughly investigated, particularly for their effects on cellular pathways in lung and colorectal cancers, two under-researched cancers with poor prognostic outcomes (lung cancers). This review focuses on the lung and colorectal cancer signaling pathways modulated by bioactive compounds from eleven traditional medicinal plants: Curcuma longa, Astragalus membranaceus, Glycyrrhiza glabra, Althaea officinalis, Echinacea purpurea, Sanguinaria canadensis, Codonopsis pilosula, Hydrastis canadensis, Lobelia inflata, Scutellaria baicalensis, and Zingiber officinale. These plants were selected based on their documented use in traditional medicine and modern clinical practice. Selection criteria involved cross-referencing herbs identified in a scoping review of traditional cancer treatments and findings from an international survey on herbal medicine currently used for lung and colorectal cancer management by our research group and the availability of existing literature on their anticancer properties. The review identifies several isolated phytoconstituents from these plants that exhibit anticancer properties by modulating key signaling pathways such as PI3K/Akt/mTOR, RAS/RAF/MAPK, Wnt/β-catenin, and TGF-β in vitro. Notable constituents include sanguinarine, berberine, hydrastine, lobeline, curcumin, gingerol, shogaol, caffeic acid, echinacoside, cichoric acid, glycyrrhizin, 18-β-glycyrrhetinic acid, astragaloside IV, lobetyolin, licochalcone A, baicalein, baicalin, wogonin, and glycyrol. Curcumin and baicalin show preclinical effectiveness but face bioavailability challenges, which may be overcome by combining them with piperine or using oral extracts to enhance gut microbiome conversion, integrating traditional knowledge with modern strategies for improved outcomes. Furthermore, herbal extracts from Echinacea, Glycyrrhiza, and Codonopsis, identified in traditional knowledge, are currently in clinical trials. Notably, curcumin and baicalin also modulate miRNA pathways, highlighting a promising intersection of modern science and traditional medicine. Thus, the development of anticancer therapeutics continues to benefit from the synergy of traditional knowledge, scientific innovation, and technological advancements.
Collapse
Affiliation(s)
| | | | - Andrea Bugarcic
- National Centre for Naturopathic Medicine, Faculty of Health, Southern Cross University, Military Road, Lismore, NSW 2480, Australia; (I.I.); (J.S.)
| |
Collapse
|
18
|
Fan Y, Wang Q, Zhang Y, Wang Y, Li W, Jiang S, Duan JN. Mechanism of Guishao Yigong decoction in treating colorectal cancer based on network pharmacology and experimental validation. J Pharm Pharmacol 2025; 77:430-445. [PMID: 39352002 DOI: 10.1093/jpp/rgae045] [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: 11/17/2023] [Accepted: 04/05/2024] [Indexed: 03/06/2025]
Abstract
OBJECTIVES To explore the effective components of Guishao Yigong decoction (GYD) in the treatment of colorectal cancer and reveal its potential mechanism of action. METHODS Through network pharmacology, the main target and signaling pathway of GYD therapy for colorectal cancer (CRC) were found. Subsequently, the effect of GYD was verified by in vitro cell viability measurements, colony formation, and scratch healing tests. The effects of GYD on metabolic pathways in vivo were found through plasma metabolomics. Finally, flow cytometry and qPCR experiments were used to verify the cycle-blocking effect of GYD on CRC cells. KEY FINDINGS Based on the network pharmacological analysis and molecular docking technology, it was found that GYD could restrain the growth of CRC cells by affecting lipid metabolic pathways and mitogen-activated protein kinase (MAPK) signaling pathways. A series of cell experiments showed that GYD could inhibit the proliferation, migration and clonogenic ability of CRC cells. Furthermore, the plasma metabolomics results showed that GYD could affect the production of unsaturated fatty acids in mice. Flow cytometry and qPCR experiments further proved that GYD blocked the CRC cells in the G1 phase and modulated the expression of cell cycle-related targets, such as AKT, TP53, CDKN1A, and CDK2. CONCLUSIONS All the results indicated that GYD could regulate the related metabolism of unsaturated fatty acids. Thus, the cell cycle was blocked and the expressions of the key proteins such as AKT and TP53 were regulated, which achieved the purpose of intervention in colorectal cancer.
Collapse
Affiliation(s)
- Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Ji-Nao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
19
|
Shahzad M, Hameed H, Amjad A, Khan MA, Qureshi IS, Hameed A, Saeed A, Munir R. An updated landscape on nanopharmaceutical delivery for mitigation of colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2107-2125. [PMID: 39361171 DOI: 10.1007/s00210-024-03482-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 03/19/2025]
Abstract
Globally, colorectal cancer (CRC) continues to rank among the leading causes of cancer-related death. Systemic toxicity, multidrug resistance, and nonspecific targeting often pose challenges to conventional therapy for CRC. Because it is a complex disease with a complex genetic and environmental pathophysiology, advanced therapeutic strategies are needed. Nanotechnology presents a potential solution that may maximize therapeutic efficacy while minimizing negative effects by enabling personalized delivery of anticancer drugs. This review focuses on recent developments in colorectal drug delivery systems based on nanotechnology. Numerous nanomaterials, including liposomes, dendrimers, micelles, exosomes, and gold nanoparticles, are developed and used. Distinctive characteristics of mentioned nanocarriers are discussed along with strategies that can be employed for enhancing the delivery of drugs to colorectal cancer cells. The review also quotes the most relevant preclinical and clinical studies that show how these nanomaterials improve drug solubility, stability, and targeted delivery while overcoming the shortcomings of conventional therapies. Nanotechnology has made CRC treatment very efficient and advanced, which has opened up new possibilities for targeted drug delivery. Preclinical and clinical studies have also proved that the use of nano-formulations in colon-specific delivery systems have significant results, indicating potential for better patient outcomes. Future research can be done in order to overcome the hurdles regarding biocompatibility, expansion, and regulatory challenges. Large-scale clinical trials and nanomaterial formulation optimization should be the main goals of future research to confirm the efficacy and safety of these novel treatments.
Collapse
Affiliation(s)
- Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ayesha Amjad
- Faculty of Food Technology and Nutrition Sciences, Lahore University of Biological and Applied Sciences, Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Inaba Shujaat Qureshi
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Asad Saeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
20
|
Sun YK, Wang JF, Sun XW, Zhang M. hnRNPA2B1 drives colorectal cancer progression via the circCDYL/EIF4A3/PHF8 axis. Kaohsiung J Med Sci 2025; 41:e12943. [PMID: 39810713 DOI: 10.1002/kjm2.12943] [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: 09/10/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
The RNA-binding protein hnRNPA2B1 acts as an m6A reader and plays a role in tumor development. This study investigates the potential mechanism of hnRNPA2B1 in colorectal cancer (CRC) progression. The expression profiles of hnRNPA2B1, circCDYL, and PHF8 in CRC cell lines were analyzed. Following si-hnRNPA2B1 transfection, CRC cell proliferation, invasion, and migration were evaluated by CCK-8 and Transwell. CDYL expression was detected after actinomycin D and RNase R treatment. RIP was conducted to assess the enrichment of hnRNPA2B1 and m6A on circCDYL. RIP and RNA pull-down assays established the interaction between circCDYL and EIF4A3/PHF8. EIF4A3 expression was evaluated using RT-qPCR and Western blot techniques. hnRNPA2B1 and PHF8 displayed high expression levels, whereas circCDYL showed low expression levels in colorectal cancer cells. Inhibition of hnRNPA2B1 reduced CRC cell proliferation, migration, and invasion. hnRNPA2B1 mechanistically elevated the m6A level of circCDYL while decreasing its expression, which in turn reduced the binding of circCDYL to EIF4A3 and enhanced PHF8 expression. In summary, hnRNPA2B1-mediated m6A modification decreases circCDYL expression, which inhibits the interaction of circCDYL with EIF4A3, enhances PHF8 expression, and ultimately facilitates CRC progression.
Collapse
Affiliation(s)
- Yu-Kai Sun
- Experimental and Clinical Research Center, Charité University Medicine Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, AG, Translational Oncology of Solid Tumors, Berlin, Germany
| | - Jin-Fu Wang
- Department of General Surgery, People's Hospital of Rehabilitation, Weifang, China
| | - Xi-Wen Sun
- Department of Gastrointestinal surgery, Linyi People's Hospital, Linyi, China
| | - Ming Zhang
- Department of Gastrointestinal Surgery Center, Weifang People's Hospital, Weifang, China
| |
Collapse
|
21
|
Muradi Muhar A, Velaro AJ, Prananda AT, Nugraha SE, Halim P, Syahputra RA. Precision medicine in colorectal cancer: genomics profiling and targeted treatment. Front Pharmacol 2025; 16:1532971. [PMID: 40083375 PMCID: PMC11903709 DOI: 10.3389/fphar.2025.1532971] [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: 11/27/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Precision medicine has revolutionized the treatment of colorectal cancer by enabling a personalized approach tailored to each patient's unique genetic characteristics. Genomic profiling allows for the identification of specific mutations in genes such as KRAS, BRAF, and PIK3CA, which play a crucial role in cell signaling pathways that regulate cell proliferation, apoptosis, and differentiation. This information enables doctors to select targeted therapies that inhibit specific molecular pathways, maximizing treatment effectiveness and minimizing side effects. Precision medicine also facilitates adaptive monitoring of tumor progression, allowing for adjustments in therapy to maintain treatment effectiveness. While challenges such as high costs, limited access to genomic technology, and the need for more representative genomic data for diverse populations remain, collaboration between researchers, medical practitioners, policymakers, and the pharmaceutical industry is crucial to ensure that precision medicine becomes a standard of care accessible to all. With continued advances and support, precision medicine has the potential to improve treatment outcomes, reduce morbidity and mortality rates, and enhance the quality of life for colorectal cancer patients worldwide.
Collapse
Affiliation(s)
- Adi Muradi Muhar
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Adrian Joshua Velaro
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Arya Tjipta Prananda
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Sony Eka Nugraha
- Department of Pharmaceutical Biology, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Universitas Sumatera Utara, Medan, Indonesia
| | | |
Collapse
|
22
|
Fernandez-Muñoz KV, Sánchez-Barrera CÁ, Meraz-Ríos M, Reyes JL, Pérez-Yépez EA, Ortiz-Melo MT, Terrazas LI, Mendoza-Rodriguez MG. Natural Alternatives in the Treatment of Colorectal Cancer: A Mechanisms Perspective. Biomolecules 2025; 15:326. [PMID: 40149862 PMCID: PMC11940303 DOI: 10.3390/biom15030326] [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: 01/24/2025] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Colorectal cancer (CRC) is one of the deadliest neoplasia. Intrinsic or acquired resistance is the main cause of failure of therapy regimens that leads to relapse and death in CRC patients. The widely used chemotherapeutic agent 5-fluorouracil (5-FU) remains the mainstay for therapeutic combinations. Unfortunately, chemotherapeutic resistance and side effects are frequent events that compromise the success of these therapies; the dysregulation of enzymes that regulate 5-FU metabolism increases the expression and activity of efflux pumps. Additional tumor cell adaptations such as epithelial-mesenchymal transition (EMT), autophagy shaping of the tumor microenvironment, and inflammation contribute to chemoresistance. Finding new strategies and alternatives to enhance conventional chemotherapies has become necessary. Recently, the study of natural compounds has been gaining strength as an alternative to chemotherapeutics in different cancers. Curcumin, trimethylglycine, resveratrol, artemisinin, and some helminth-derived molecules, among others, are some natural compounds studied in the context of CRC. This review discusses the main benefits, mechanisms, advances, and dark side of conventional chemotherapeutics currently evaluated in CRC treatment. We also analyzed the landscape of alternative non-conventional compounds and their underlying mechanisms of action, which could, in the short term, provide fundamental knowledge to harness their anti-tumor effects and allow them to be used as alternative adjuvant therapies.
Collapse
Affiliation(s)
- Karen Vanessa Fernandez-Muñoz
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Avenida Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - Cuauhtémoc Ángel Sánchez-Barrera
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
| | - Marco Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Avenida Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - Jose Luis Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
| | | | - Maria Teresa Ortiz-Melo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
| | - Luis I. Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Monica Graciela Mendoza-Rodriguez
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; (K.V.F.-M.); (C.Á.S.-B.); (M.T.O.-M.)
| |
Collapse
|
23
|
Catalano T, Selvaggi F, Cotellese R, Aceto GM. The Role of Reactive Oxygen Species in Colorectal Cancer Initiation and Progression: Perspectives on Theranostic Approaches. Cancers (Basel) 2025; 17:752. [PMID: 40075600 PMCID: PMC11899472 DOI: 10.3390/cancers17050752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Altered levels of reactive oxygen species (ROS) are recognized as one of the key factors in mediating tumor cell survival in the tissue microenvironment, where they play a role in the initiation, progression and recurrence/relapse of colorectal cancer (CRC). Tumor cells can adapt to oxidative stress (OS) using genetic or metabolic reprogramming in the long or short term. In addition, tumor cells defend themselves through positive regulation of antioxidant molecules, enhancing ROS-driven proliferation. Balanced oxidative eustress levels can influence chemotherapy resistance, allowing tumor cells to survive treatment. Secondary effects of chemotherapy include increased ROS production and redox stress, which can kill cancer cells and eliminate drug resistance. Anticancer treatments based on manipulating ROS levels could represent the gold standard in CRC therapy. Therefore, exploring the modulation of the response to OS in deregulated signaling pathways may lead to the development of new personalized CRC treatments to overcome therapy resistance. In this review, we explore the role of ROS in the initiation and progression of CRC and their diagnostic implications as biomarkers of disease. Furthermore, we focused on the involvement of ROS in different CRC therapeutic options, such as surgery, radiotherapy, theranostic imaging, chemotherapy and immunotherapy and other precision medicine approaches.
Collapse
Affiliation(s)
- Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Federico Selvaggi
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Roberto Cotellese
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Gitana Maria Aceto
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
- Department of Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
24
|
Cheng J, Yu W, Zhou W. Acute exposure to nitrogen mustard induces rapid nuclear component regulation and delayed stress to exogenous stimuli. Int Immunopharmacol 2025; 147:113976. [PMID: 39787759 DOI: 10.1016/j.intimp.2024.113976] [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: 09/29/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Nitrogen mustard (NM) is a vesicant agent with potent toxic effects on various tissues. Numerous theories have been proposed to explain its toxic mechanisms, yet research on the interconnections among these theories is lacking. This study focuses on analyzing the characteristics of genes involved in NM-induced bronchial injury within the Comparative Toxicogenomics Database (CTD). Subsequently, based on the CTD, we compared and analyzed the acute exposure and delayed changes following exposure in 16HBE cells. The injury processes caused by NM to bronchial and skin tissues are similar, primarily involving metabolism and regulation of nuclear constituents and inflammatory responses within the cellular matrix. During the acute exposure phase, NM rapidly induces nuclear stress, with the JUN family at the core of regulating metabolic and nucleic acid activities, and various nuclear binding proteins exhibit abnormalities. Delayed reactions following acute exposure are primarily centered in the cytoplasmic region, with diverse reaction types, including oxidative stress and responses to exogenous stimuli. Abnormalities in the activity of multiple cellular matrix enzymes are observed, with a relatively even involvement of various stress responses. Communication between the nucleus and cytoplasm is extreme active during the injury, and the content of the communication changes over time. These results suggest a temporal sequence in which NM causes chromatin damage and mediates cytoplasmic stress responses. In prevention and first aid, rapid DNA repair should be the primary focus, while subsequent treatment after acute exposure should focus more on delayed inflammatory and oxidative stress responses.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Public Health and Emergency Management, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Wenpei Yu
- Department of Chemical Defense Medicine, School of Preventive Medicine, The Third Military Medical University (Army Medical University),Chongqing, China
| | - Wenzheng Zhou
- Clinical and Public Health Research Center, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Health Center for Women and Children, Chongqing, China; Chongqing Research Center for Prevention & Control of Matermal and Child Disease and Public Health, Chongqing, China.
| |
Collapse
|
25
|
Saadh MJ, Menon SV, Verma R, Siva Prasad GV, Allela OQB, Mahdi MS, Ahmad N, Husseen B. LncRNA CRNDE and HOTAIR: Molecules behind the scenes in the progression of gastrointestinal cancers through regulating microRNAs. Pathol Res Pract 2025; 266:155778. [PMID: 39721094 DOI: 10.1016/j.prp.2024.155778] [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: 08/21/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Gastrointestinal (GI) cancers, such as gastric cancer, hepatocellular carcinoma, colorectal cancer, and esophageal cancer, pose a significant medical and economic burden globally, accounting for the majority of new cancer cases and deaths each year. A lack of knowledge about the molecular mechanisms of GI cancers is reflected in the low efficacy of treatment for individuals with late stage and recurring illness. Understanding the molecular pathways that promote the growth of GI cancers may open doors for their therapy. Numerous long non-coding RNAs (lncRNAs) that are produced differently in normal and malignant tissues have been discovered by genome-wide techniques. The role of lncRNAs in the diagnosis, proliferation, metastasis, and drug resistance of different GI cancers has been investigated in recent research. LncRNAs may affect transcription, epigenetic modifications, protein/RNA stability, translation, and post-translational modifications via their interactions with DNA, RNAs, and proteins. Also, by functioning as competing endogenous RNAs (ceRNAs), they control the synthesis of certain microRNAs (miRNAs), which in turn modify the downstream target molecules of these miRNAs. Based on recent studies, lncRNAs in particular, CRNDE and HOTAIR, sponge different miRNAs and their downstream genes, which in turn regulate GI cancers development, including cell proliferation, invasion, migration, and chemoresistance. In this comprehensive review, we present an overview of the biological roles of CRNDE and HOTAIR and their associated mechanisms, miRNAs/mRNA pathways, in various GI cancers, encompassing colorectal cancer, hepatocellular carcinoma, esophageal cancer, and gastric cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Sahibzada Ajit Singh Nagar, Jhanjeri, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | | | | | - Nabeel Ahmad
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India; Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand- 831001, India.
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
26
|
Nayak A, Streiff H, Gonzalez I, Adekoya OO, Silva I, Shenoy AK. Wnt Pathway-Targeted Therapy in Gastrointestinal Cancers: Integrating Benchside Insights with Bedside Applications. Cells 2025; 14:178. [PMID: 39936971 PMCID: PMC11816596 DOI: 10.3390/cells14030178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
The Wnt signaling pathway is critical in the onset and progression of gastrointestinal (GI) cancers. Anomalies in this pathway, often stemming from mutations in critical components such as adenomatous polyposis coli (APC) or β-catenin, lead to uncontrolled cell proliferation and survival. In the case of colorectal cancer, dysregulation of the Wnt pathway drives tumor initiation and growth. Similarly, aberrant Wnt signaling contributes to tumor development, metastasis, and resistance to therapy in other GI cancers, such as gastric, pancreatic, and hepatocellular carcinomas. Targeting the Wnt pathway or its downstream effectors has emerged as a promising therapeutic strategy for combating these highly aggressive GI malignancies. Here, we review the dysregulation of the Wnt signaling pathway in the pathogenesis of GI cancers and further explore the therapeutic potential of targeting the various components of the Wnt pathway. Furthermore, we summarize and integrate the preclinical evidence supporting the therapeutic efficacy of potent Wnt pathway inhibitors with completed and ongoing clinical trials in GI cancers. Additionally, we discuss the challenges of Wnt pathway-targeted therapies in GI cancers to overcome these concerns for effective clinical translation.
Collapse
|
27
|
Zhang H, Zhuo C, Lin R, Ke F, Wang M, Yang C. ASO Author Reflections: Why are the Identification and Verification of Key Genes in Colorectal Cancer Liver Metastases so Important? Ann Surg Oncol 2025; 32:182-183. [PMID: 39489828 DOI: 10.1245/s10434-024-16334-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Hui Zhang
- Department of Hepatopancreatobiliary Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Changhua Zhuo
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Ruirong Lin
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Fayong Ke
- Department of Hepatopancreatobiliary Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Ming Wang
- Department of Hepatopancreatobiliary Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
28
|
Zhang J, Li Z, Han J, Tian Z, Meng Q, Niu W. KLF7 enhances the invasion and migration of colorectal cancer cells via the miR-139-5p/TPD52 axis. Cancer Biol Ther 2024; 25:2385172. [PMID: 39097779 PMCID: PMC11299624 DOI: 10.1080/15384047.2024.2385172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
In this study, we aimed to investigate the molecular mechanism of Krüppel-like factor 7 (KLF7) in colorectal cancer (CRC) cell invasion and migration. The expression pattern of KLF7 in CRC tissues and the correlation between KLF7 expression and clinical symptoms of CRC were analyzed. CRC cell lines were transfected with si-KLF7, followed by qRT-PCR or western blot detection of KLF7, miR-139-5p, and tumor protein D52 (TPD52) expression, cell counting kit-8 (CCK-8) assay to detect cell viability, and transwell detection of invasion and migration. Chromatin immunoprecipitation (ChIP) analyzed the enrichment KLF7 in the miR-139-5p promoter. The dual-luciferase reporter assay verified the binding relationship between KLF7 and miR-139-5p, and between miR-139-5p and TPD52. In the subcutaneous tumorigenesis experiment, tumor growth was observed and ki67-positive expression was detected. KLF7 is abundantly expressed in CRC cells KLF7 silencing inhibits CRC cell viability, invasion, and migration. KLF7 represses miR-139-5p expression by binding to the miR-139-5p promoter. miR-139-5p targets TPD52 expression. miR-13-5p inhibition or TPD52 overexpression partially counteracted the effect of KLF7 silencing in CRC cells. KLF7 silencing suppresses tumor growth in vivo. In conclusion, KLF7 suppresses miR-139-5p expression by binding to the miR-139-5p promoter, thereby upregulating TPD52 expression and enhancing CRC cell invasion and migration.
Collapse
Affiliation(s)
- Juan Zhang
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhihan Li
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaxu Han
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhongtao Tian
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qingyu Meng
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenbo Niu
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
29
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
30
|
Luo P, Huang C, Guo J, Yao X, Pan C, Bao A, Li F, Li YY. Network pharmacology and anticancer mechanism study of Dendrobium nobile dendrobine in the treatment of colorectal cancer. Discov Oncol 2024; 15:768. [PMID: 39692955 DOI: 10.1007/s12672-024-01641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE This study aims to explore the potential targets and anticancer mechanisms of dendrobine from Dendrobium nobile in the treatment of colorectal cancer through network pharmacology, and to experimentally validate its specific effects. METHODS Initially, potential targets of dendrobine were identified using the ITCM Traditional Chinese Medicine database, while colorectal cancer-related genes were obtained from the NCBI Gene database, with the intersection of these datasets taken for further analysis. Functional enrichment analysis was conducted using the Metascape database, and a protein-protein interaction (PPI) network was constructed. Additionally, cell culture, cell proliferation assays, and wound healing assays were performed. The Wnt/β-catenin and NF-κB/COX-2/PGE2 signaling pathways were analyzed using PCR and Western blot experiments. RESULTS The PPI network constructed from 152 intersecting genes revealed that these genes play crucial roles in processes such as cell proliferation, apoptosis, and signal transduction. Cell-based assays demonstrated that dendrobine significantly inhibits the proliferation and migration of colorectal cancer cells. Furthermore, PCR and Western blot results indicated that dendrobine suppresses colorectal cancer cell proliferation and migration by modulating the Wnt/β-catenin and NF-κB/COX-2/PGE2 signaling pathways. CONCLUSION Dendrobine exhibits significant anticancer potential against colorectal cancer by regulating the Wnt/β-catenin and NF-κB/COX-2/PGE2 signaling pathways, providing a theoretical foundation and experimental evidence for its therapeutic application in colorectal cancer.
Collapse
Affiliation(s)
- Pei Luo
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Can Huang
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Jun Guo
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Xin Yao
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Chao Pan
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Aijin Bao
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Fei Li
- Department of Gastroenterology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China
| | - Ying-Ying Li
- Department of Gerontology, People's Hospital of Qianxinan Prefecture, Xingyi, 562400, Guizhou, China.
| |
Collapse
|
31
|
Lin Q, Wang Z, Wang J, Xu M, Zhang X, Sun P, Yuan Y. Innovative strategies to optimise colorectal cancer immunotherapy through molecular mechanism insights. Front Immunol 2024; 15:1509658. [PMID: 39717768 PMCID: PMC11663906 DOI: 10.3389/fimmu.2024.1509658] [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: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 12/25/2024] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related deaths globally. The heterogeneity of the tumor microenvironment significantly influences patient prognosis, while the diversity of tumor cells shapes its unique characteristics. A comprehensive analysis of the molecular profile of tumor cells is crucial for identifying novel molecular targets for drug sensitivity analysis and for uncovering the pathophysiological mechanisms underlying CRC. Methods We utilized single-cell RNA sequencing technology to analyze 13 tissue samples from 4 CRC patients, identifying key cell types within the tumor microenvironment. Intercellular communication was assessed using CellChat, and a risk score model was developed based on eight prognostic genes to enhance patient stratification for immunotherapeutic approaches. Additionally, in vitro experiments were performed on DLX2, a gene strongly associated with poor prognosis, to validate its potential role as a therapeutic target in CRC progression. Results Eight major cell types were identified across the tissue samples. Within the tumor cell population, seven distinct subtypes were recognized, with the C0 FXYD5+ tumor cells subtype being significantly linked to cancer progression and poor prognosis. CellChat analysis indicated extensive communication among tumor cells, fibroblasts, and immune cells, underscoring the complexity of the tumor microenvironment. The risk score model demonstrated high accuracy in predicting 1-, 3-, and 5-year survival rates in CRC patients. Enrichment analysis revealed that the C0 FXYD5+ tumor cell subtype exhibited increased energy metabolism, protein synthesis, and oxidative phosphorylation, contributing to its aggressive behavior. In vitro experiments confirmed DLX2 as a critical gene associated with poor prognosis, suggesting its viability as a target for improving drug sensitivity. Conclusion In summary, this study advances our understanding of CRC progression by identifying critical tumor subtypes, molecular pathways, and prognostic markers that can inform innovative strategies for predicting and enhancing drug sensitivity. These findings hold promise for optimizing immunotherapeutic approaches and developing new targeted therapies, ultimately aiming to improve patient outcomes in CRC.
Collapse
Affiliation(s)
- Quanjun Lin
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqiang Wang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jue Wang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Xu
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihang Yuan
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Zhang H, Zhuo C, Lin R, Ke F, Wang M, Yang C. Identification and Verification of Key Genes in Colorectal Cancer Liver Metastases Through Analysis of Single-Cell Sequencing Data and TCGA Data. Ann Surg Oncol 2024; 31:8664-8679. [PMID: 39382748 PMCID: PMC11549235 DOI: 10.1245/s10434-024-16194-9] [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: 05/01/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is highly prevalent worldwide, with more patients experiencing colorectal cancer liver metastases (CRLM). This study aimed to identify key genes in CRLM through single-cell sequencing data reanalysis and experimental validation. METHODS The study analyzed single-cell RNA-sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) database. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for gene functional enrichment analysis. The Cancer Genome Atlas (TCGA) data enabled bulk-RNA expression and survival prognosis analysis. Real-time polymerase chain reaction (qPCR) detected mRNA expression, whereas Western blot determined protein levels. Cell function experiments assessed SPARC's impact on CRC cell behavior. RESULTS Cluster analysis showed 23 classes among 17 CRLM samples, representing six cell types. A GO and KEGG analysis identified interleukin-1 beta (IL1B), CD2 molecule (CD2), and C-X-C motif chemokine ligand 8 (CXCL8) as significant prognostic factors in CRC. Secreted protein acidic and cysteine rich (SPARC) was one of the top differentially expressed genes (DEGs) in tissue stem cells, confirmed in primary and metastatic lesions. Metastatic lesions showed higher expression of SPARC and CRC stem cell marker leucine-rich repeat containing G protein-coupled receptor 5 (LGR5), which was significantly correlated positively with LGR5 expression. Knockdown of SPARC reduced CRC cell sphere- and colony-formation, invasion, and migration abilities. Overexpression of SPARC significantly increased the malignancy of CRC cells. CONCLUSIONS Several key genes were identified in the process of CRLM. In CRLM samples and those corresponding to CRC stem cells, SPARC was significantly upregulated. In the therapy of CRLM, SPARC might be a potential target.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Hepatopancreatobiliary Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Changhua Zhuo
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ruirong Lin
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Fayong Ke
- Department of Hepatopancreatobiliary Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Ming Wang
- Department of Hepatopancreatobiliary Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
33
|
Calafato G, Alquati C, Bernardi A, Di Paola FJ, Ricciardiello L. Comparative Analysis of Commercial and Home-Made Media on RSPO1/S6R Axis in Organoids with Different Wnt Backgrounds: A Methodological Guide for the Selection of Intestinal Patient-Derived Organoids Culture Media. Int J Mol Sci 2024; 25:11526. [PMID: 39519079 PMCID: PMC11546270 DOI: 10.3390/ijms252111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
WNT3A is an intestinal ligand triggering the Wnt/β-catenin (Wnt) pathway, which can be enhanced by R-spondin 1 (RSPO1) through the RSPO1-LGR axis or antagonized by the adenomatous polyposis coli (APC) protein supporting β-catenin-degradation. Wnt interplays with several pathways including PI3K/mTOR (mTOR). In this study, we evaluated the influence of WNT3A-commercial and home-made culture media and RSPO1 protein on the Wnt and mTOR interplay in non-APC and APC-mutated intestinal patient-derived organoids (PDOs). Normal mucosa (NM) of sporadic CRC and FAP PDOs were cultured with: WNT3A-lacking/containing commercial (A/A+B) or home-made (BASAL/WNT3A-conditioned medium (CM)±RSPO1) media. In non-APC-mutated-PDOs (CRC-NM), WNT3A-CM, over commercial A+B, strongly activated Wnt-target-genes CCND1 and c-MYC. Most importantly, the addition of RSPO1 to home-made WNT3A-CM or A+B led to the downregulation of the mTOR-downstream-effector phospho-S6 ribosomal protein (p-S6R), highlighting the activation of the RSPO1-pS6R in both non-APC (CRC-NM) and APC-mutated (FAP-NM) PDOs, independently from LGR5 gene expression modulation. Our work demonstrates that home-made WNT3A-CM strongly impacts the crosstalk between Wnt and mTOR over commercial media, and proposes RSPO1 as a key regulator of the RSPO1-p-S6R axis in both non-APC and APC-mutated PDOs. Together, these findings represent an important methodological guide for scientists working in these fields to select the most appropriate intestinal PDO media.
Collapse
Affiliation(s)
- Giulia Calafato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.C.)
| | - Chiara Alquati
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (C.A.)
| | - Alice Bernardi
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (C.A.)
| | | | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (C.A.)
- Centre for Applied Biomedical Research (CRBA), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
34
|
Sahebnasagh R, Deli H, Shadboorestan A, Vakili-Ghartavol Z, Salehi N, Komeili-Movahhed T, Azizi Z, Ghahremani MH. Identification of key lncRNAs associated with oxaliplatin resistance in colorectal cancer cells and isolated exosomes: From In-Silico prediction to In-Vitro validation. PLoS One 2024; 19:e0311680. [PMID: 39401197 PMCID: PMC11472961 DOI: 10.1371/journal.pone.0311680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/23/2024] [Indexed: 10/17/2024] Open
Abstract
One of the critical challenges in managing colorectal cancer (CRC) is the development of oxaliplatin (OXP) resistance. Long non-coding RNAs (lncRNAs) have a crucial role in CRC progression and chemotherapy resistance, with exosomal lncRNAs emerging as potential biomarkers. This study aimed to predict key lncRNAs involved in OXP-resistance using in-silico methods and validate them using RT-qPCR methods in CRC cells and their isolated exosomes. Two public datasets, GSE42387 and GSE119481, were downloaded from the GEO database to identify differentially expressed genes (DEGs) and miRNAs (DEmiRNAs) associated with OXP-resistance in the HCT116 cell line. The analysis of GSE42387 revealed 210 DEGs, and GSE119481 identified 73 DEmiRNAs. A protein-protein interaction (PPI) network analysis of the DEGs identified 133 interconnected genes, from which the top ten genes with the highest degree scores were selected. By intersecting predicted miRNAs targeting these genes with the DEmiRNAs, 38 common miRNAs were found. Subsequently, 224 lncRNAs targeting these common miRNAs were predicted. LncRNA-miRNA-mRNA network were constructed and the top five lncRNAs with the highest degree scores were identified. Analysis using the Kaplan-Meier plotter database revealed that the key lncRNAs NEAT1, OIP5-AS1, and MALAT1 are significantly associated with the overall survival of CRC patients. To validate these lncRNAs, OXP-resistant HCT116 sub-cell line (HCT116/OXR) was developed by exposing parental HCT116 cells to gradually increasing concentrations of OXP. Exosomes derived from both HCT116 and HCT116/OXR cells were isolated and characterized utilizing dynamic light scattering (DLS), transmission electron microscopy (TEM), and Western blotting. RT-qPCR confirmed elevated levels of NEAT1, OIP5-AS1, and MALAT1 in HCT116/OXR cells and their exosomes compared to parental HCT116 cells and their exosomes. This study concludes that NEAT1, OIP5-AS1, and MALAT1 are associated with the OXP-resistance in CRC. The high levels of these lncRNAs in exosomes of resistant cells suggest their involvement in intercellular communication and resistance propagation. This positioning makes them promising biomarkers for OXP-resistance in CRC.
Collapse
Affiliation(s)
- Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Deli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zeynab Vakili-Ghartavol
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Salehi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | | | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
El Din N, El‑Shenawy R, Moustafa R, Khairy A, Farouk S. Association between the expression level of miRNA‑374a and TGF‑β1 in patients with colorectal cancer. WORLD ACADEMY OF SCIENCES JOURNAL 2024; 6:68. [DOI: 10.3892/wasj.2024.283] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Affiliation(s)
- Noha El Din
- Microbial Biotechnology Department, National Research Centre, Dokki 12622, Egypt
| | - Reem El‑Shenawy
- Microbial Biotechnology Department, National Research Centre, Dokki 12622, Egypt
| | - Rehab Moustafa
- Microbial Biotechnology Department, National Research Centre, Dokki 12622, Egypt
| | - Ahmed Khairy
- Endemic Medicine Department, Faculty of Medicine, Cairo University, Giza 11559, Egypt
| | - Sally Farouk
- Microbial Biotechnology Department, National Research Centre, Dokki 12622, Egypt
| |
Collapse
|
36
|
Mokhfi FZ, Al Amin M, Zehravi M, Sweilam SH, Arjun UVNV, Gupta JK, Vallamkonda B, Balakrishnan A, Challa M, Singh J, Prasad PD, Ali SS, Ahmad I, Doukani K, Emran TB. Alkaloid-based modulators of the PI3K/Akt/mTOR pathway for cancer therapy: Understandings from pharmacological point of view. Chem Biol Interact 2024; 402:111218. [PMID: 39209016 DOI: 10.1016/j.cbi.2024.111218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
This review aims to summarize the role of alkaloids as potential modulators of the PI3K/Akt/mTOR (PAMT) pathway in cancer therapy. The PAMT pathway plays a critical role in cell growth, survival, and metabolism, and its dysregulation contributes to cancer hallmarks. In healthy cells, this pathway is tightly controlled. However, this pathway is frequently dysregulated in cancers and becomes abnormally active. This can happen due to mutations in genes within the pathway itself or due to other factors. This chronic overactivity promotes cancer hallmarks such as uncontrolled cell division, resistance to cell death, and increased blood vessel formation to nourish the tumor. As a result, the PAMT pathway is a crucial therapeutic target for cancer. Researchers are developing drugs that specifically target different components of this pathway, aiming to turn it off and slow cancer progression. Alkaloids, a class of naturally occurring nitrogen-containing molecules found in plants, have emerged as potential therapeutic agents. These alkaloids can target different points within the PAMT pathway, inhibiting its activity and potentially resulting in cancer cell death or suppression of tumor growth. Research is ongoing to explore the role of various alkaloids in cancer treatment. Berberine reduces mTOR activity and increases apoptosis by targeting the PAMT pathway, inhibiting cancer cell proliferation. Lycorine inhibits Akt phosphorylation and mTOR activation, increasing pro-apoptotic protein production and decreasing cell viability. In glioblastoma models, harmine suppresses mTORC1. This review focuses on alkaloids such as evodiamine, hirsuteine, chaetocochin J, indole-3-carbinol, noscapine, berberine, piperlongumine, and so on, which have shown promise in targeting the PAMT pathway. Clinical studies evaluating alkaloids as part of cancer treatment are underway, and their potential impact on patient outcomes is being investigated. In summary, alkaloids represent a promising avenue for targeting the dysregulated PAMT pathway in cancer, and further research is warranted.
Collapse
Affiliation(s)
- Fatima Zohra Mokhfi
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Md Al Amin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Uppuluri Varuna Naga Venkata Arjun
- Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai, 600117, Tamil Nadu, India
| | | | - Bhaskar Vallamkonda
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN'S Foundation for Science, Technology & Research, Vadlamudi, Andhra Pradesh, India
| | - Anitha Balakrishnan
- Department of Pharmaceutics, GRT Institute of Pharmaceutical Education and Research, Tiruttani, Tamil Nadu, India
| | - Manjula Challa
- Department of Pharmaceutics, Vasavi Institute of Pharmaceutical Sciences, Vasavi Nagar, Peddapalli Village, Sidhout Mandal Kadapa District, Andhra Pradesh, India
| | - Jyoti Singh
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - P Dharani Prasad
- Depertment of Pharmacology, Mohan Babu University, MB School of Pharmaceutical Sciences, (Erstwhile, Sree Vidyaniketan College of Pharmacy), Tirupati, India
| | - Syed Salman Ali
- Lloyd Institute of Management and Technology, Plot No.-11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201306, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Department of Biology, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh; Department of Pathology and Laboratory Medicine and Legorreta Cancer Center Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA; Legorreta Cancer Center, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
37
|
Zhang L, Li Y, Yao L, He R, Wu J. Establishment and Clinical Significance of the Patient-Derived Xenograft Model of Colorectal Cancer. Cureus 2024; 16:e71116. [PMID: 39525113 PMCID: PMC11544153 DOI: 10.7759/cureus.71116] [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] [Accepted: 08/10/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES Patient-derived xenograft (PDX) models are widely acknowledged for their ability to reflect the heterogeneity of human cancers and can be used to improve preclinical models. In this study, we evaluated the factors affecting the tumor formation rate of the PDX colorectal cancer (CRC) model and conducted preliminary drug sensitivity tests. METHODS CRC patients who underwent elective surgery at Shaoxing People's Hospital from November 2019 to October 2020 were included. The tumor tissue obtained from surgery was transplanted to the back of NSG mice, and the PDX model was established and subcultured to the F3 generation. Factors that affected tumorigenicity were analyzed and compared histologically. Drug interventions included 5-fluorouracil, oxaliplatin, and propofol. RESULTS Sixty CRC patients were included in this study, and tumorigenesis was observed in CRC tissue derived from 37 cases (62%). The primary tumor malignancy degree (tumor stage and degree of cell differentiation), preoperative carcinoembryonic antigen level, and tumor location in CRC patients could affect the tumorigenicity of the PDX model. Histopathological analysis of CRC-PDX transplanted tumor tissue was highly consistent with the patient's tumor tissue. All four chemotherapy regimens could inhibit tumor growth and cause tumor tissue damage. Propofol could inhibit diarrhea in mice and protect intestinal mucosa. CONCLUSIONS The CRC-PDX model established in this study can maintain the biological characteristics of primary tumors and can be used as a reference model for the individualized treatment of CRC patients. The degree of malignancy of the primary tumor is the primary factor affecting the tumorigenesis rate of the PDX model.
Collapse
Affiliation(s)
- Li Zhang
- Anesthesiology, Hangzhou Linping Qiaosi Community Health Service Center, Hangzhou, CHN
| | - Yuhong Li
- Anesthesiology, Zhejiang Shuren University, Hangzhou, CHN
| | - Liuxu Yao
- Anesthesiology, Zhejiang People's Hospital, Hangzhou, CHN
| | - Rui He
- Anesthesiology, Shaoxing People's Hospital, Shaoxing, CHN
| | - Jianqiang Wu
- Anesthesiology, Hangzhou Linping Qiaosi Community Health Service Center, Hangzhou, CHN
| |
Collapse
|
38
|
Wu X, Liu P, Wang Q, Sun L, Wang Y. A prognostic model established using bile acid genes to predict the immunity and survival of patients with gastrointestinal cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:4594-4609. [PMID: 38606991 DOI: 10.1002/tox.24287] [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: 01/24/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND The metabolism of abnormal bile acids (BAs) is implicated in the initiation and development of gastrointestinal (GI) cancer. However, there was a lack of research on the molecular mechanisms of BAs metabolism in GI. METHODS Genes involved in BAs metabolism were excavated from public databases of The Cancer Genome Atlas (TCGA) database, Gene Expression Omnibus (GEO) database, and Molecular Signatures Database (MSigDB). ConsensusClusterPlus was used to classify molecular subtypes for GI. To develop a RiskScore model for predicting GI prognosis, univariate Cox analysis was performed on the genes in protein-protein interaction (PPI) network, followed by using Lasso regression and stepwise regression to refine the model and to determine the key prognostic genes. Tumor immune microenvironment in GI patients from different risk groups was assessed using the ESTIMATE algorithm and enrichment analysis. Reverse transcription-quantitative real-time PCR (RT-qPCR), Transwell assay, and wound healing assay were carried out to validate the expression and functions of the model genes. RESULTS This study defined three molecular subtypes (C1, C2, and C3). Specifically, C1 had the best prognosis, while C3 had the worst prognosis with high immune checkpoint gene expression levels and TIDE scores. We selected nine key genes (AXIN2, ATOH1, CHST13, PNMA2, GYG2, MAGEA3, SNCG, HEYL, and RASSF10) that significantly affected the prognosis of GI and used them to develop a RiskScore model accordingly. Combining the verification results from a nomogram, the prediction of the model was proven to be accurate. The high RiskScore group was significantly enriched in tumor and immune-related pathways. Compared with normal gastric mucosal epithelial cells, the mRNA levels of the nine genes were differential in the gastric cancer cells. Inhibition of PNMA2 suppressed migration and invasion of the cancer cells. CONCLUSION We distinguished three GI molecular subtypes with different prognosis based on the genes related to BAs metabolism and developed a RiskScore model, contributing to the diagnosis and treatment of patients with GI.
Collapse
Affiliation(s)
- Xin Wu
- Department of General Surgical Medicine, The First Medicine Center of PLA General Hospital, Beijing, China
| | - Peifa Liu
- Pathology Department, The First Medicine Center of PLA General Hospital, Beijing, China
| | - Qing Wang
- Department of General Surgical Medicine, The First Medicine Center of PLA General Hospital, Beijing, China
| | - Linde Sun
- Department of General Surgical Medicine, The First Medicine Center of PLA General Hospital, Beijing, China
| | - Yu Wang
- Department of General Surgical Medicine, The First Medicine Center of PLA General Hospital, Beijing, China
| |
Collapse
|
39
|
Gatasheh MK. Identifying key genes against rutin on human colorectal cancer cells via ROS pathway by integrated bioinformatic analysis and experimental validation. Comput Biol Chem 2024; 112:108178. [PMID: 39191167 DOI: 10.1016/j.compbiolchem.2024.108178] [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: 03/13/2024] [Revised: 08/04/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
Colorectal cancer (CRC) poses a significant global health challenge, characterized by substantial prevalence variations across regions. This study delves into the therapeutic potential of rutin, a polyphenol abundant in fruits, for treating CRC. The primary objectives encompass identifying molecular targets and pathways influenced by rutin through an integrated approach combining bioinformatic analysis and experimental validation. Employing Gene Set Enrichment Analysis (GSEA), the study focused on identifying potential differentially expressed genes (DEGs) associated with CRC, specifically those involved in regulating reactive oxygen species, metabolic reprogramming, cell cycle regulation, and apoptosis. Utilizing diverse databases such as GEO2R, CTD, and Gene Cards, the investigation revealed a set of 16 targets. A pharmacological network analysis was subsequently conducted using STITCH and Cytoscape, pinpointing six highly upregulated genes within the rutin network, including TP53, PCNA, CDK4, CCNEB1, CDKN1A, and LDHA. Gene Ontology (GO) analysis predicted functional categories, shedding light on rutin's potential impact on antioxidant properties. KEGG pathway analysis enriched crucial pathways like metabolic and ROS signaling pathways, HIF1a, and mTOR signaling. Diagnostic assessments were performed using UALCAN and GEPIA databases, evaluating mRNA expression levels and overall survival for the identified targets. Molecular docking studies confirmed robust binding associations between rutin and biomolecules such as TP53, PCNA, CDK4, CCNEB1, CDKN1A, and LDHA. Experimental validation included inhibiting colorectal cell HT-29 growth and promoting cell growth with NAC through MTT assay. Flow cytometric analysis also observed rutin-induced G1 phase arrest and cell death in HT-29 cells. RT-PCR demonstrated reduced expression levels of target biomolecules in HT-29 cells treated with rutin. This comprehensive study underscores rutin's potential as a promising therapeutic avenue for CRC, combining computational insights with robust experimental evidence to provide a holistic understanding of its efficacy.
Collapse
Affiliation(s)
- Mansour K Gatasheh
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
40
|
Wu YJ, Xiong JF, Zhan CN, Xu H. Gut microbiota alterations in colorectal adenoma-carcinoma sequence based on 16S rRNA gene sequencing: A systematic review and meta-analysis. Microb Pathog 2024; 195:106889. [PMID: 39197689 DOI: 10.1016/j.micpath.2024.106889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/12/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Most sporadic colorectal cancers (CRC) develop through the adenoma-carcinoma sequence. While dysbiosis of the intestinal flora contributes to CRC's pathogenesis, precise microbial taxa closely associated with the colorectal adenoma-carcinoma sequence remain elusive. This meta-analysis aimed to summarize the features of intestinal flora in patients with AD and CRC. METHODS PubMed, Embase, Cochrane Library, and Web of Science were searched for case-control studies comparing the relative abundance of gut microbiota in the feces of patients with AD, CRC, and healthy controls (HC) from inception to January 2024. The weighted mean difference (WMD) with a 95 % confidence interval (CI) was used to display the results. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the entailed literature. Publication bias was evaluated with the Egger's and Begg's tests. RESULTS Eleven studies were included, involving 477 CRC patients, 628 AD patients, and 864 healthy controls. Compared with HC, the patients with AD had a significantly lower Chao 1 index (WMD = -30.17, 95 % CI [-41.10, -19.23], P < 0.001) and Shannon index (WMD = -0.11 95 % CI [-0.18, -0.04], P = 0.002). Compared with AD, the CRC patients had a significantly higher Chao1 index (WMD = 22.09, 95 % CI [7.59, 36.00], P = 0.003) and Shannon index (WMD = 0.08, 95 % CI [0.00, 0.15], P = 0.037). Enterobacteriaceae (WMD = 0.03 95 % CI [0.00,0.05], P = 0.047; WMD = 0.02 95 % CI [0.00,0.04], P = 0.027) significantly increased in the order of Control-AD-CRC, while that of Blautia (WMD = -0.00 95 % CI [-0.01, -0.00], P = 0.001; WMD = -0.00 95 % CI [-0.00, -0.00], P = 0.002) was reduced. Compared with HC, the relative abundance of Proteobacteria (WMD = 0.05 95 % CI [0.03,0.07], P < 0.001), Fusobacteria (WMD = 0.02 95 % CI [0.00,0.03], P = 0.042), Streptococcaceae (WMD = 0.03 95 % CI [0.01,0.05], P = 0.017), Prevotellaceae (WMD = 0.02 95 % CI [0.00,0.04], P = 0.040), and Escherichia-Shigella (WMD = 0.06 95 % CI [0.01, 0.11], P = 0.021) was enriched in the CRC group. The relative abundance of Alistipes (WMD = 0.00 95 % CI [0.00,0.01], P = 0.032) and Streptococcus (WMD = 0.00 95 % CI [0.00,0.00], P = 0.001) was increased in the AD vs HC. The relative abundance of Firmicutes (WMD = -0.07 95 % CI [-0.12, -0.03], P = 0.003), Bifidobacteria (WMD = -0.03 95 % CI [-0.05, -0.01], P = 0.016), and Klebsiella (WMD = -0.01 95 % CI [-0.01, -0.00], P = 0.001) was decreased in the CRC vs HC. Compared with AD, the relative abundance of Firmicutes (WMD = -0.04 95 % CI [-0.07, -0.02], P = 0.002), Peptostreptococcaceae (WMD = -0.03 95 % CI [-0.05, -0.00], P = 0.021), Lachnospiraceae (WMD = -0.04 95 % CI [-0.08,-0.00], P = 0.037), Ruminococcaceae (WMD = -0.06 95 % CI [-0.09,-0.03], P < 0.001), Faecalibacterium (WMD = -0.01 95 % CI [-0.02, -0.01], P = 0.001), and Lachnoclostridium (WMD = -0.02 95 % CI [-0.03, -0.00], P = 0.040) was decreased in the CRC group, while Proteobacteria (WMD = 0.04 95 % CI [0.02,0.05], P < 0.001) was increased. CONCLUSIONS The dysbiosis characterized by reduced levels of short-chain fatty acid (SCFA)-producing bacteria, decreased anti-inflammatory bacteria, increased pro-inflammatory bacteria, and an elevation of bacteria with cytotoxic effects damaging to DNA may represent the specific microbial signature of colorectal adenoma/carcinoma. Further research is required to elucidate the mechanisms by which gut dysbiosis leads to the progression from AD to CRC and to explore the potential of specific microbiota markers in clinical treatment and non-invasive screening.
Collapse
Affiliation(s)
- Yi-Jun Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing-Fang Xiong
- Department of Geriatrics, Hangzhou Red Cross Hospital, Hangzhou, China
| | - Cheng-Nan Zhan
- Medical Service Community, Hangzhou Xiaoshan Hospital of TCM, Hangzhou, China
| | - Hong Xu
- Department of Gastroenterology and Hepatology, Hangzhou Red Cross Hospital, Hangzhou, China.
| |
Collapse
|
41
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Fan Y, Zhong J. LINC01094: A key long non-coding RNA in the regulation of cancer progression and therapeutic targets. Heliyon 2024; 10:e37527. [PMID: 39309878 PMCID: PMC11415682 DOI: 10.1016/j.heliyon.2024.e37527] [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: 04/27/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
LINC01094 is a long non-coding RNA that plays a crucial role in cancer progression by modulating key signaling pathways, such as PI3K/AKT, Wnt/β-catenin and TGF-β Signaling Pathway Feedback Loop. In this review we summarize the recent research on the functional mechanisms of LINC01094 in various cancers, including its impact on tumor growth, metastasis, and resistance to therapy. We also discuss the therapeutic potential of targeting LINC01094 and highlight the current strategies and challenges in this area. Perspectives on future development of LINC01094-based therapies are also provided.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Yu Fan
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, 341000, China
| |
Collapse
|
42
|
Piotrowska U, Orzechowska K. Advances in Chitosan-Based Smart Hydrogels for Colorectal Cancer Treatment. Pharmaceuticals (Basel) 2024; 17:1260. [PMID: 39458901 PMCID: PMC11510048 DOI: 10.3390/ph17101260] [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: 09/04/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Despite advancements in early detection and treatment in developed countries, colorectal cancer (CRC) remains the third most common malignancy and the second-leading cause of cancer-related deaths worldwide. Conventional chemotherapy, a key option for CRC treatment, has several drawbacks, including poor selectivity and the development of multiple drug resistance, which often lead to severe side effects. In recent years, the use of polysaccharides as drug delivery systems (DDSs) to enhance drug efficacy has gained significant attention. Among these polysaccharides, chitosan (CS), a linear, mucoadhesive polymer, has shown promise in cancer treatment. This review summarizes current research on the potential applications of CS-based hydrogels as DDSs for CRC treatment, with a particular focus on smart hydrogels. These smart CS-based hydrogel systems are categorized into two main types: stimuli-responsive injectable hydrogels that undergo sol-gel transitions in situ, and single-, dual-, and multi-stimuli-responsive CS-based hydrogels capable of releasing drugs in response to various triggers. The review also discusses the structural characteristics of CS, the methods for preparing CS-based hydrogels, and recent scientific advances in smart CS-based hydrogels for CRC treatment.
Collapse
Affiliation(s)
- Urszula Piotrowska
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | | |
Collapse
|
43
|
纪 凯, 于 冠, 周 乐, 张 天, 凌 潜, 满 文, 朱 冰, 张 卫. [ HNRNPA1 gene is highly expressed in colorectal cancer: its prognostic implications and potential as a therapeutic target]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1685-1695. [PMID: 39505336 PMCID: PMC11744076 DOI: 10.12122/j.issn.1673-4254.2024.09.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To investigate the expression level of HNRNP A1 in colorectal cancer (CRC) and its prognostic implications. METHODS We investigated HNRNP A1 expression level in CRC using HPA, TIMER, and GEPIA databases and analyzed its association with Ki-67 and VEGFA expressions. Kaplan-Meier Plotter database was used to analyze the correlation of HNRNP A1 mRNA levels with the survival rates of CRC patients. Pathway enrichment analysis was performed for predicting the biological roles of HNRNP A1 in CRC progression. Immunohistochemistry and Western blotting were used to examine the protein levels of HNRNP A1 in CRC versus adjacent tissues, and TIMER was used for assessing its expression in the infiltrating immune cells. In RKO/Caco2 cells, the effects of lentivirus-mediated knockdown of HNRNP A1 on cell proliferation and migration were observed, and the inhibitory effect of VPC-80051 (a HNRNP A1 inhibitor) on cell proliferation was evaluated to assess its potential as a therapeutic agent. RESULTS HNRNP A1 was significantly overexpressed in CRC tissues and correlated with a poor prognosis of the patients. HNRNP A1 expression level was correlated with the infiltrating immune cells in CRC microenvironment and positively correlated with MKI67 and VEGFA expressions in CRC. A high HNRNP A1 expression predicted a in survival and progression-free survival of CRC patients and was involved in multiple biological processes related with CRC progression. In RKO/Caco2 cells, HNRNP A1 knockdown significantly suppressed cell proliferation and migration, and treatment with VPC-80051 also effectively inhibited CRC cell proliferation. Immunohistochemical study demonstrated a close correlation of HNRNP A1 overexpression with tumor stage of CRC. CONCLUSION HNRNP A1 is overexpressed in CRC tissues to modulate cell proliferation and migration and is correlated with a poorer prognosis. VPC-80051 can effectively inhibit CRC cell proliferation, suggesting the potential of HNRNP A1 as a therapeutic target for CRC.
Collapse
Affiliation(s)
- 凯 纪
- 蚌埠医科大学第一附属医院胃肠外科, 安徽 蚌埠 233000Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| | - 冠宇 于
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| | - 乐其 周
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| | - 天帅 张
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| | - 潜龙 凌
- 蚌埠医科大学第一附属医院胃肠外科, 安徽 蚌埠 233000Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| | - 文江 满
- 蚌埠医科大学第一附属医院胃肠外科, 安徽 蚌埠 233000Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| | - 冰 朱
- 蚌埠医科大学第一附属医院胃肠外科, 安徽 蚌埠 233000Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| | - 卫 张
- 海军军医大学第一附属医院//上海长海医院肛肠外科, 上海 200082Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200082, China
| |
Collapse
|
44
|
Gakinya S, Njau A, Abdallah A, Nzioka A, Ogutu J. Frequency of KRAS and BRAF mutations in colorectal carcinoma and their association with clinical-pathological characteristics in a tertiary hospital in Kenya. Front Med (Lausanne) 2024; 11:1433120. [PMID: 39364024 PMCID: PMC11446768 DOI: 10.3389/fmed.2024.1433120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction Colorectal carcinoma is a leading cause of cancer morbidity and mortality globally. Its management includes the use of targeted therapy which require assessment for biomarkers to choose eligible patients. KRAS and BRAF mutations are biomarkers predictive of response to anti-EGFR therapy. This study aimed at determining the frequency of BRAF V600E and KRAS exon 2,3,4 mutations in colorectal carcinoma patients at the Aga Khan University Hospital Nairobi, Kenya. Methods Study participants were patients who had colectomy for colorectal carcinoma. They were identified from the laboratory information system. The patients age, gender and tumor location were determined from the medical records. The histological diagnosis, pathological tumor and nodal stage were confirmed by examining hematoxylin and eosin-stained slides prepared from the colectomy specimen. DNA was extracted from the specimens using Qiagen QIAamp DNA FFPE Tissue Kit and PCR performed using EntroGen KRAS/BRAF mutation analysis kit following manufacturer's protocol. Results One hundred fourteen patients were enrolled. Colorectal carcinoma was significantly more common in males than females. The mean age at diagnosis was 58 years. Majority of the tumors were in the right colon, were of pathological tumor stage T3 and had nodal involvement. Forty six percent (46%) of the cases had KRAS mutations while 5.3% had BRAF V600E mutation. KRAS mutation was associated with a high pathological tumor stage and nodal involvement. Conclusion Colorectal carcinoma in our patients is more common in males and tend to occur at a younger age. The patients tend to have a high tumor pathological stage and nodal involvement at diagnosis. The high frequency of KRAS exon 2,3,4 mutation and low frequency of BRAF V600E mutations is similar to what has been reported in literature.
Collapse
Affiliation(s)
- Samuel Gakinya
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Allan Njau
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | | | - Ancent Nzioka
- Department of Pathology, Kenyatta University Teaching, Referral & Research Hospital, Nairobi, Kenya
| | - James Ogutu
- Department of Microbiology, School of Medicine, Kenyatta University, Nairobi, Kenya
| |
Collapse
|
45
|
Chen JM, He J, Qiu JM, Yang GG, Wang D, Shen Z. Netrin-1-CD146 and netrin-1-S100A9 are associated with early stage of lymph node metastasis in colorectal cancer. BMC Gastroenterol 2024; 24:308. [PMID: 39261771 PMCID: PMC11389491 DOI: 10.1186/s12876-024-03401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The netrin-1/CD146 pathway regulates colorectal cancer (CRC) liver metastasis, angiogenesis, and vascular development. However, few investigations have yet examined the biological function of netrin-1/CD146 complex in CRC. In this work, we investigated the relationship between the netrin-1/CD146 axis and S100 proteins in sentinel lymph node, and revealed a possible new clue for vascular metastasis of CRC. METHODS The expression levels of netrin-1 and CD146 proteins in CRC, as well as S100A8 and S100A9 proteins in the sentinel lymph nodes were determined by immunohistochemistry. Using GEPIA and UALCAN, we analyzed netrin-1 and CD146 gene expression in CRC, their association with CRC stage, and their expression levels and prognosis in CRC patients. RESULTS The expression level of netrin-1 in N1a+1b (CRC lymphatic metastasis groups, exculded N1c) was positively increased with N0 (p = 0.012). The level of netrin-1 protein was positively correlated with CD146 protein (p < 0.05). The level of S100A9 protein was positively correlated with CD146 protein (r = 0.492, p = 0.007). Moreover, netrin-1 expression was obviously correlated with S100A9 expression in the N1 stage (r = 0.867, p = 0.000). CD146 level was correlated with S100A9 level in the N2 stage (r = 0.731, p = 0.039). CD146 mRNA expression was higher in normal colorectal tissues than in CRC (p < 0.05). Netrin-1 and CD146 expression were not significantly associated with the tumor stages and prognosis of patients with CRC (p > 0.05). CONCLUSIONS The netrin-1/CD146 and netrin-1/S100A9 axis in CRC tissues might related with early stage of lymph node metastasis, thus providing potential novel channels for blocking lymphatic metastasis and guiding biomarker discovery in CRC patients.
Collapse
Affiliation(s)
- Jin-Ming Chen
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China.
| | - Jun He
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China
| | - Jian-Ming Qiu
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China
| | - Guan-Gen Yang
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China
| | - Dong Wang
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China
| | - Zhong Shen
- Department of Anorectal Surgery, the Third People's Hospital of Hangzhou, 38 West Lake Avenue, 310009, Hangzhou, People's Republic of China.
| |
Collapse
|
46
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
47
|
Feng Y, Wang K, Fan J, Wu X, Li T, Yang Z. Mindfulness intervention, homogeneous medical concept, and concentrated solution nursing for colorectal cancer patients: a retrospective study. BMC Cancer 2024; 24:1055. [PMID: 39192195 PMCID: PMC11348521 DOI: 10.1186/s12885-024-12508-y] [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: 02/09/2024] [Accepted: 06/12/2024] [Indexed: 08/29/2024] Open
Abstract
OBJECTIVE We aim to explore the differences of the psychological distress of postoperative chemotherapy patients with colorectal cancer between mindfulness intervention combined with homogeneous medical concepts and mindfulness intervention only. METHODS One hundred patients with colorectal cancer undergoing chemotherapy after surgery from Sep 2020 to Sep 2022 were enrolled and divided into active control group (Solution centered nursing interventions; homogenized medical and nursing professional teams; dedicated personnel responsible for "admission notices"; Regular follow-up after discharge) and mindfulness group (homogeneous medical concept + and concentrated solution + Mindfulness intervention) with 50 cases in each group according to different nursing methods. RESULTS After nursing, the physical function, emotional function, cognitive function, and social function of the patients in the mindfulness group were significantly higher than those in the active control group. However, the overall life and economic difficulties of the patients in the mindfulness group were significantly lower than those in the active control group (P < 0.05). After nursing, the observation score, description score, action score, intrinsic experience score, non-judgment score and non-reaction score of the mindfulness group were significantly higher than those of the active control group (P < 0.05). CONCLUSION The implementation of mindfulness intervention in colorectal cancer patients undergoing chemotherapy can alleviate the patients' negative emotions, improve the level of mindfulness, and improve the quality of life of patients.
Collapse
Affiliation(s)
- Yaning Feng
- Physical Examination Center, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Kuanlei Wang
- Hospital Office, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Jianchun Fan
- Graduate School, Hebei North University, Zhangjiakou, 075000, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 12 Changqing Rd, Zhangjiakou, 075000, China.
- Tumor Research Institute, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhili Yang
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 12 Changqing Rd, Zhangjiakou, 075000, China.
- Department of Anorectal Surgery, Xinchang Country People's Hospital, 117 Gushan Middle Road, Xinchang, Zhejiang Province, 312500, China.
| |
Collapse
|
48
|
Abd Rahman NI, Tham CL, Abd Hamid R. Inhibition of angiogenesis and metastasis in colorectal cancer cell lines through KRAS-associated signaling pathways by 2-methoxy-6-undecyl-1,4-benzoquinone. Chem Biol Interact 2024; 399:111151. [PMID: 39025287 DOI: 10.1016/j.cbi.2024.111151] [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: 05/05/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Colorectal cancer (CRC), the third most prevalent cancer globally, presents formidable hurdles in treatment owing to factors such as therapeutic resistance and genetic mutations affecting primary drug targets. 2-methoxy-6-undecyl-1,4-benzoquinone (BQ), derived from Ardisia crispa roots, has emerged as a potent anti-inflammatory and anti-angiogenic compound with substantial potential, as evidenced by previous studies. This study aimed to explore the potential of BQ in suppressing angiogenesis and metastasis in the human CRC cell lines LoVo and HCT116. Various in vitro and in silico studies have been conducted to elucidate the potential pathway(s) of BQ. BQ was highly cytotoxic, with an IC50 of 7.01 ± 0.6 μM in HCT116 and 9.58 ± 0.8 μM in LoVo cells. Moreover, BQ induced notable apoptotic activity and suppressed migration, invasion, and adhesion in both cell lines. The inhibition of MMP-2 suggests the potential of BQ to impede extracellular matrix degradation and CRC cell metastasis. BQ inhibits the expression of key proteins involved in angiogenesis and metastasis, including VEGF-A, VEGF-C, BRAF, ERK, KRAS, PI3K, and AKT. Molecular docking simulations illustrated the robust binding of BQ to CRC protein receptors. BQ holds promise in impeding CRC progression by targeting angiogenesis and metastasis, particularly through inhibition of the KRAS/BRAF/ERK and KRAS/PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Noor Izzah Abd Rahman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Center of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Roslida Abd Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
49
|
Al-Nasser S, Abdulla MH, Alhassan N, Vaali-Mohammed MA, Al-Omar S, Hamdi N, Elnakady Y, Matou-Nasri S, Mansour L. A Benzimidazole-Based N-Heterocyclic Carbene Derivative Exhibits Potent Antiproliferative and Apoptotic Effects against Colorectal Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1379. [PMID: 39336420 PMCID: PMC11433580 DOI: 10.3390/medicina60091379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/20/2024] [Accepted: 08/05/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: Colorectal cancer (CRC) remains a major global health issue. Although chemotherapy is the first-line treatment, its effectiveness is limited due to drug resistance developed in CRC. To overcome resistance and improve the prognosis of CRC patients, investigating new therapeutic approaches is necessary. Materials and Methods: Using human colorectal adenocarcinoma (HT29) and metastatic CRC (SW620) cell lines, the potential anticancer properties of a newly synthesized compound 1-(Isobutyl)-3-(4-methylbenzyl) benzimidazolium chloride (IMBZC) were evaluated by performing MTT cytotoxicity, cell migration, and colony formation assays, as well as by monitoring apoptosis-related protein and gene expression using Western blot and reverse transcription-quantitative polymerase chain reaction technologies. Results: Tested at various concentrations, the half-maximal inhibitory concentrations (IC50) of IMBZC on HT29 and SW620 cell growth were determined to be 22.13 µM (6.97 μg/mL) and 15.53 µM (4.89 μg/mL), respectively. IMBZC did not alter the cell growth of normal HEK293 cell lines. In addition, IMBZC inhibited cell migration and significantly decreased colony formation, suggesting its promising role in suppressing cancer metastasis. Mechanistic analyses revealed that IMBZC treatment increased the expression of pro-apoptotic proteins p53 and Bax, while decreasing the expression of anti-apoptotic proteins Bcl-2 and Bcl-xL, thus indicating the induction of apoptosis in IMBZC-treated CRC cells, compared to untreated cells. Additionally, the addition of IMBZC to conventional chemotherapeutic drugs (i.e., 5-fluorouracil, irinotecan, and oxaliplatin) resulted in an increase in the cytotoxic potential of the drugs. Conclusions: This study suggests that IMBZC has substantial anticancer effects against CRC cells through its ability to induce apoptosis, inhibit cancer cell migration and colony formation, and enhance the cytotoxic effects of conventional chemotherapeutic drugs. These findings indicate that IMBZC could be a promising chemotherapeutic drug for the treatment of CRC. Further research should be conducted using in vivo models to confirm the anti-CRC activities of IMBZC.
Collapse
Affiliation(s)
- Sarah Al-Nasser
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Noura Alhassan
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Mansoor-Ali Vaali-Mohammed
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Suliman Al-Omar
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naceur Hamdi
- Research Laboratory of Environmental Sciences and Technologies (LR16ES09), Higher Institute of Environmental Sciences and Technology, University of Carthage, Hammam-Lif 2050, Tunisia
| | - Yasser Elnakady
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School of Systems Biology, George Mason University, Manassas, VA 20110, USA
| | - Lamjed Mansour
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
50
|
Lou S, Liu YX, Xia C, Zhang Q, Deng L, Tang JJ. Novel meroterpene-like compounds inhibit ferroptosis through Fe 2+ chelation. Int J Biochem Cell Biol 2024; 173:106610. [PMID: 38879152 DOI: 10.1016/j.biocel.2024.106610] [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: 03/29/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 07/22/2024]
Abstract
Colorectal cancer (CRC) is the third most common type of cancer in the world. It is characterized by complex crosstalk between various signaling pathways, as a result of which it is highly challenging to identify optimal therapeutic targets and design treatment strategies. In this study, we tested the effect of 700 compounds on the CRC cell line HT-29 by using the sulforhodamine B assay and screened out 17 compounds that exhibited high toxicity (indicated by an inhibition rate of ≥75 % when applied at a concentration of 10 µM) against the HT-29 cell line. Next, we investigated the mechanisms underlying the effects of these 17 highly toxic compounds. The results of ferroptosis analysis and electron microscopy showed that compounds 575 and 578 were able to significantly reverse RSL3-induced increase in ferroptosis, while compound 580 had a less pronounced ferroptosis-regulating effect. In subsequent experiments, western blotting showed that compounds 575, 578, and 580, which belong to a class of meroterpene-like compounds that affect ferroptosis, do not induce autophagy or apoptosis in the CRC cell line. Instead, Fe2+ chelation experiments showed that these three compounds can serve as iron chelators by chelating Fe2+ at a 1:1 (chelator: Fe2+) ratio. Specifically, the aldehyde and hydroxyl groups of the benzene ring in these compounds may chelate Fe2+, thus reducing Fe2+ levels in cells and inhibiting ferroptosis. These results indicate that these novel meroterpene-like compounds are potential therapeutic small-molecule candidates for targeting ferroptosis in tumors.
Collapse
Affiliation(s)
- Shiyang Lou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Yan-Xiang Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Qiang Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China; Shenzhen Research Institute, Northwest A&F University, Shenzhen, Guangdong 518000, China.
| | - Jiang-Jiang Tang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China.
| |
Collapse
|