1
|
Chung DJ, Wang CH, Liu PJ, Ng SK, Luo CK, Jwo SH, Li CT, Hsu DY, Fan CC, Wei TT. Targeting CREB-binding protein (CBP) abrogates colorectal cancer stemness through epigenetic regulation of C-MYC. Cancer Gene Ther 2024:10.1038/s41417-024-00838-9. [PMID: 39358564 DOI: 10.1038/s41417-024-00838-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Colorectal cancer (CRC) is a common cancer worldwide with an increasing annual incidence. Cancer stem cells (CSCs) play important roles in the occurrence, development, recurrence, and metastasis of CRC. The molecular mechanism regulating the development of colorectal CSCs remains unclear. The discovery of human induced pluripotent stem cells (hiPSCs) through somatic cell reprogramming has revolutionized the fields of stem cell biology and translational medicine. In the present study, we converted hiPSCs into cancer stem-like cells by culture with conditioned medium (CM) from CRC cells. These transformed cells, termed hiPSC-CSCs, displayed cancer stem-like properties, including a spheroid morphology and the expression of both pluripotency and CSC markers. HiPSC-CSCs showed tumorigenic and metastatic abilities in mouse models. The epithelial-mesenchymal transition phenotype was observed in hiPSC-CSCs, which promoted their migration and angiogenesis. Interestingly, upregulation of C-MYC was observed during the differentiation of hiPSC-CSCs. Mechanistically, CREB binding protein (CBP) bound to the C-MYC promoter, while histone deacetylase 1 and 3 (HDAC1/3) dissociated from the promoter, ultimately leading to an increase in histone acetylation and C-MYC transcriptional activation during the differentiation of hiPSC-CSCs. Pharmacological treatment with a CBP inhibitor or abrogation of CBP expression with a CRISPR/Cas9-based strategy reduced the stemness of hiPSC-CSCs. This study demonstrates for the first time that colorectal CSCs can be generated from hiPSCs. The upregulation of C-MYC via histone acetylation plays a crucial role during the conversion process. Inhibition of CBP is a potential strategy for attenuating the stemness of colorectal CSCs.
Collapse
Affiliation(s)
- Dai-Jung Chung
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chun-Hao Wang
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | - Pin-Jung Liu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shang-Kok Ng
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Cong-Kai Luo
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Si-Han Jwo
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chin-Tzu Li
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Dai-Yi Hsu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chia-Chu Fan
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Tzu-Tang Wei
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program in Chemical Biology and Molecular Biophysics (TIGP-CBMB), Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
2
|
Miyazaki K, Hoshino D, Kasajima R, Koizume S, Koshikawa N, Miyagi Y. Oncofetal morphogenesis similar to embryonic gut formation by a subpopulation of DLD-1 human colon cancer cells. Exp Cell Res 2024; 442:114188. [PMID: 39128553 DOI: 10.1016/j.yexcr.2024.114188] [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: 01/18/2024] [Revised: 06/25/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
Cancer stem cells (CSC) are thought to be responsible for cancer phenotypes and cellular heterogeneity. Here we demonstrate that the human colon cancer cell line DLD1 contains two types of CSC-like cells that undergo distinct morphogenesis in the reconstituted basement membrane gel Matrigel. In our method with cancer cell spheroids, the parent cell line (DLD1-P) developed grape-like budding structures, whereas the other (DLD1-Wm) and its single-cell clones dynamically developed worm-like ones. Gene expression analysis suggested that the former mimicked intestinal crypt-villus morphogenesis, while the latter mimicked embryonic hindgut development. The organoids of DLD1-Wm cells rapidly extended in two opposite directions by expressing dipolar proteolytic activity. The invasive morphogenesis required the expression of MMP-2 and CD133 genes and ROCK activity. These cells also exhibited gastrula-like morphogenesis even in two-dimensional cultures without Matrigel. Moreover, the two DLD1 cell lines showed clear differences in cellular growth, tumor growth and susceptibility to paclitaxel. This study also provides a simple organoid culture method for human cancer cell lines. HT-29 and other cancer cell lines underwent characteristic morphogenesis in direct contact with normal fibroblasts. Such organoid cultures would be useful for investigating the nature of CSCs and for screening anti-cancer drugs. Our results lead to the hypothesis that CSC-like cells with both invasive activity and a fetal phenotype, i. e. oncofetal CSCs, are generated in some types of colon cancers.
Collapse
Affiliation(s)
- Kaoru Miyazaki
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Daisuke Hoshino
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Naohiko Koshikawa
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuda-cho, Midori-ku, Yokohama, 226-8501, Japan.
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| |
Collapse
|
3
|
Zhu M, Yu R, Liu Y, Geng X, Liu Q, Liu S, Zhu Y, Li G, Guo Y, Xi X, Du B. LncRNA H19 Participates in Leukemia Inhibitory Factor Mediated Stemness Promotion in Colorectal Cancer Cells. Biochem Genet 2024; 62:3695-3708. [PMID: 38198021 DOI: 10.1007/s10528-023-10627-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: 05/24/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024]
Abstract
Colorectal cancer (CRC) is a common human malignancy and the third leading cause of cancer-related death worldwide. Cancer stem cells (CSCs) were considered to play important roles in the genesis and development of many tumors. In recent years, it has been observed that leukemia inhibitory factor (LIF) might be involved in the regulation of stemness in cancer cells. In this study, we observed that LIF could increase the spheroid formation and stemness marker expression (inculding Nanog and SOX2) in CRC cell lines, such as HCT116 and Caco2 cells. Meanwhile, we also observed that LIF could upregulate LncRNA H19 expression via PI3K/AKT pathway. Knockdown of the expression of LncRNA H19 could decrease the spheroid formation and SOX2 expression in LIF-treated HCT116 and Caco2 cells, and thereby LncRNA H19 knockdown could compensate for the stemness enhancement effects induced by LIF. Our results indicated that LncRNA H19 might participate in the stemness promotion of LIF in CRC cells.
Collapse
Affiliation(s)
- Min Zhu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ruihong Yu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Yirui Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Xiaoqing Geng
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Qiong Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Shuaitong Liu
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Yunhe Zhu
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Gang Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yang Guo
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China
| | - Xueyan Xi
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China.
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Boyu Du
- Institute of Basic Medical Sciences, Hubei University of Medicine, No.30 Renmin Nanlu, Shiyan, 442000, Hubei, China.
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
4
|
Yang R, Lai C, Huang L, Li F, Peng W, Wu M, Xin J, Lu Y, Ouyang M, Bai Y, Lei H, He S, Lin Y. Role of disulfidptosis in colorectal adenocarcinoma: implications for prognosis and immunity. Front Immunol 2024; 15:1409149. [PMID: 39399504 PMCID: PMC11466812 DOI: 10.3389/fimmu.2024.1409149] [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: 03/29/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024] Open
Abstract
Background Recent research has found a new way of cell death: disulfidptosis. Under glucose starvation, abnormal accumulation of disulfide molecules such as Cystine in Solute Carrier Family 7 Member 11 (SLC7A11) overexpression cells induced disulfide stress to trigger cell death. The research on disulfidptosis is still in its early stages, and its role in the occurrence and development of colorectal malignancies is still unclear. Method In this study, we employed bioinformatics methods to analyze the expression and mutation characteristics of disulfidptosis-related genes (DRGs) in colorectal cancer. Consensus clustering analysis was used to identify molecular subtypes of Colorectal Adenocarcinoma (COAD) associated with disulfidptosis. The biological behaviors between subtypes were analyzed to explore the impact of disulfidptosis on the tumor microenvironment. Constructing and validating a prognostic risk model for COAD using diverse data. The influence of key genes on prognosis was evaluated through SHapley Additive exPlanations (SHAP) analysis, and the predictive capability of the model was assessed using Overall Survival analysis, Area Under Curve and risk curves. The immunological status of different patients and the prediction of drug treatment response were determined through immune cell infiltration, TMB, MSI status, and drug sensitivity analysis. Single-cell analysis was employed to explore the expression of genes at the cellular level, and finally validated the expression of key genes in clinical samples. Result By integrating the public data from two platforms, we identified 2 colorectal cancer subtypes related to DRGs. Ultimately, we established a prognosis risk model for COAD using 7 genes (FABA4+GIPC2+EGR3+HOXC6+CCL11+CXCL10+ITLN1). SHAP analysis can further explained the positive or negative impact of gene expression on prognosis. By dividing patients into high-risk and low-risk groups, we found that patients in the high-risk group had poorer prognosis, higher TMB, and a higher proportion of MSI-H and MSI-L statuses. We also predicted that drugs such as 5-Fluorouracil, Oxaliplatin, Gefitinib, and Sorafenib would be more effective in low-risk patients, while drugs like Luminesib and Staurosporine would be more effective in high-risk patients. Single-cell analysis revealed that these 7 genes not only differ at the level of immune cells but also in epithelial cells, fibroblasts, and myofibroblasts, among other cell types. Finally, the expression of these key genes was verified in clinical samples, with consistent results. Conclusions Our research findings provide evidence for the role of disulfidptosis in COAD and offer new insights for personalized and precise treatment of COAD.
Collapse
Affiliation(s)
- Ruanruan Yang
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunxiao Lai
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luji Huang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Feng Li
- Department of Gastroenterology, Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen, China
| | - Weiqi Peng
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meiyan Wu
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinge Xin
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Lu
- Department of Good Clinical Practice (GCP), Shunde Hospital, Southern Medical University, Foshan, China
| | - Manzhao Ouyang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Yang Bai
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoqiang Lei
- Huangpu People’s Hospital of Zhongshan, Zhongshan, China
| | - Shunhui He
- Department of Gastroenterology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Yu Lin
- Department of Gastroenterology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Verhagen MP, Xu T, Stabile R, Joosten R, Tucci FA, van Royen M, Trerotola M, Alberti S, Sacchetti A, Fodde R. The SW480 cell line as a model of resident and migrating colon cancer stem cells. iScience 2024; 27:110658. [PMID: 39246444 PMCID: PMC11379671 DOI: 10.1016/j.isci.2024.110658] [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: 03/13/2024] [Revised: 06/27/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Intra-tumor heterogeneity, i.e., the presence of diverse cell types and subpopulations within tumors, presents a significant obstacle in cancer treatment due to its negative consequences for resistance to therapy and disease recurrence. However, the mechanisms that underlie intra-tumor heterogeneity and result in the plethora of different cancer cells within a single lesion remain poorly understood. Here, we leverage the SW480 cell line as a model system to investigate the molecular and functional diversity of colon cancer cells. Through a combination of fluorescence-activated cell sorting (FACS) analysis and transcriptomic profiling, we identified three distinct subpopulations, namely resident cancer stem cells (rCSCs), migratory CSCs (mCSCs), and high-relapse cells (HRCs). These subpopulations show varying Wnt signaling levels and gene expression profiles mirroring their stem-like and functional properties. Examination of publicly available spatial transcriptomic data confirms the presence of these subpopulations in patient-derived cancers and reveals their distinct spatial distribution relative to the tumor microenvironment.
Collapse
Affiliation(s)
- Mathijs P Verhagen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tong Xu
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roberto Stabile
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rosalie Joosten
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Francesco A Tucci
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Martin van Royen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marco Trerotola
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Saverio Alberti
- Department of Biomedical Sciences, University of Messina, Messina, Italy
| | - Andrea Sacchetti
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Riccardo Fodde
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
6
|
Gou S, Wu A, Luo Z. Integrins in cancer stem cells. Front Cell Dev Biol 2024; 12:1434378. [PMID: 39239559 PMCID: PMC11375753 DOI: 10.3389/fcell.2024.1434378] [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: 05/20/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Integrins are a class of adhesion receptors on cell membranes, consisting of α and β subunits. By binding to the extracellular matrix, integrins activate intracellular signaling pathways, participating in every step of cancer initiation and progression. Tumor stem cells possess self-renewal and self-differentiation abilities, along with strong tumorigenic potential. In this review, we discussed the role of integrins in cancer, with a focus on their impact on tumor stem cells and tumor stemness. This will aid in targeting tumor stem cells as a therapeutic approach, leading to the exploration of novel cancer treatment strategies.
Collapse
Affiliation(s)
- Siqi Gou
- The Second Affiliated Hospital, Department of urology, Hengyang Medical School, University of South China, Hengyang, China
| | - Anqi Wu
- The Second Affiliated Hospital, Department of Clinical Research Center, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhigang Luo
- The Second Affiliated Hospital, Department of urology, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
7
|
Georvasili V, Markopoulos G, Lampri E, Lianos G, Vartholomatos G, Mitsis M, Bali C. Could Flow Cytometry Provide New Prognostic Markers in Colorectal Cancer? J Clin Med 2024; 13:4753. [PMID: 39200895 PMCID: PMC11355755 DOI: 10.3390/jcm13164753] [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: 06/29/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Background/Objectives: Colorectal cancer (CRC) is still accompanied by significant mortality, which poses the necessity of novel markers to predict treatment success and patient survival. This study aims to evaluate the prognostic and survival impact of flowytometry (FC) in CRC patients. Methods: In this prospective study, 106 surgically resectable CRC patients were included. Tissue specimens from tumor and normal mucosa were collected and analyzed by FC. DNA and tumor index were calculated. In a subgroup of 46 patients, the CD26 expression on tumor cells was estimated. These parameters were compared with patients' tumor characteristics as stage, histology data, responsiveness to treatment, metastasis/recurrence, and, finally, patients' survival to identify possible new biomarkers. Results: The overall survival and the disease-specific survival in our study group was 76% and 72%, respectively, during the 7-year follow up period. Diploid tumors had better median survival than the aneuploid ones. The DNA index had significant correlation to the tumor index and response to neoadjuvant treatment. Similarly, the tumor index was also significantly related to the response to neoadjuvant treatment. Patients with a higher tumor index had worst survival rates. Surprisingly, CD26 levels were not associated with any of the parameters examined and were negatively related to tumor stage and differentiation. Conclusions: FC is a rapid and reliable method of cell analysis. In CRC, it has been used for prognostic and diagnostic purposes. In this study, we have shown that DNA and tumor index could become predictive biomarkers of tumor response to neoadjuvant treatment and survival of resectable CRC patients.
Collapse
Affiliation(s)
- Vaia Georvasili
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece; (V.G.); (G.L.); (M.M.)
| | - Georgios Markopoulos
- Unit of Molecular Biology, University Hospital of Ioannina, 45500 Ioannina, Greece; (G.M.); (G.V.)
| | - Evangeli Lampri
- Department of Pathology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Georgios Lianos
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece; (V.G.); (G.L.); (M.M.)
| | - George Vartholomatos
- Unit of Molecular Biology, University Hospital of Ioannina, 45500 Ioannina, Greece; (G.M.); (G.V.)
| | - Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece; (V.G.); (G.L.); (M.M.)
| | - Christina Bali
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece; (V.G.); (G.L.); (M.M.)
| |
Collapse
|
8
|
Dang Q, Zuo L, Hu X, Zhou Z, Chen S, Liu S, Ba Y, Zuo A, Xu H, Weng S, Zhang Y, Luo P, Cheng Q, Liu Z, Han X. Molecular subtypes of colorectal cancer in the era of precision oncotherapy: Current inspirations and future challenges. Cancer Med 2024; 13:e70041. [PMID: 39054866 PMCID: PMC11272957 DOI: 10.1002/cam4.70041] [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: 03/29/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most hackneyed malignancies. Even patients with identical clinical symptoms and the same TNM stage still exhibit radically different clinical outcomes after receiving equivalent treatment regimens, indicating extensive heterogeneity of CRC. Myriad molecular subtypes of CRC have been exploited for decades, including the most compelling consensus molecular subtype (CMS) classification that has been broadly applied for patient stratification and biomarker-drug combination formulation. Encountering barriers to clinical translation, however, CMS classification fails to fully reflect inter- or intra-tumor heterogeneity of CRC. As a consequence, addressing heterogeneity and precisely managing CRC patients with unique characteristics remain arduous tasks for clinicians. REVIEW In this review, we systematically summarize molecular subtypes of CRC and further elaborate on their clinical applications, limitations, and future orientations. CONCLUSION In recent years, exploration of subtypes through cell lines, animal models, patient-derived xenografts (PDXs), organoids, and clinical trials contributes to refining biological insights and unraveling subtype-specific therapies in CRC. Therapeutic interventions including nanotechnology, clustered regulatory interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9), gut microbiome, and liquid biopsy are powerful tools with the possibility to shift the immunologic landscape and outlook for CRC precise medicine.
Collapse
Affiliation(s)
- Qin Dang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Lulu Zuo
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinru Hu
- Department of Cardiology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Chen
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
9
|
Tang J, Zhang Y, Zhou L, Song X, Wei Y, Qi J, Wu J, Song Z, Zhan L. Design, synthesis and biological evaluation of indoline-maleimide conjugates as potential antitumor agents for the treatment of colorectal cancer. Bioorg Med Chem 2024; 108:117786. [PMID: 38843656 DOI: 10.1016/j.bmc.2024.117786] [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: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/17/2024]
Abstract
An efficient protocol for direct coupling of maleimides and indolines at the C7-position was achieved under Rh(III) catalysis. Thirty four novel indoline-maleimide conjugates were prepared in good to excellent yields using this method. All compounds were evaluated for their anti-proliferative effect against colorectal cell lines. Among them, compound 3ab showed the most potent anti-proliferative activity against the CRC cells, and displayed low toxicity in the normal cell. Further investigation indicated that 3ab could effectively suppress the proliferation and migration of CRC cells, along with inducing cell cycle arrest and apoptosis. Mechanistic studies revealed that compound 3ab inhibited the proliferation of CRC cells via suppressing the AKT/GSK-3β pathway. In vivo evaluation demonstrated remarkable antitumor effect of 3ab (10 mg/kg) in the HCT116 xenograft model with no obvious toxicity, which is superior to that of 5-Fluorouracil (20 mg/kg). Therefore, conjugate 3ab could be considered as a potential CRC therapy agent for further development.
Collapse
Affiliation(s)
- Jielin Tang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yuxin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lingling Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiangrui Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yusi Wei
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ji Qi
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianmin Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Lingling Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
10
|
Pereira F, Fernández-Barral A, Larriba MJ, Barbáchano A, González-Sancho JM. From molecular basis to clinical insights: a challenging future for the vitamin D endocrine system in colorectal cancer. FEBS J 2024; 291:2485-2518. [PMID: 37699548 DOI: 10.1111/febs.16955] [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: 05/15/2023] [Revised: 08/03/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023]
Abstract
Colorectal cancer (CRC) is one of the most life-threatening neoplasias in terms of incidence and mortality worldwide. Vitamin D deficiency has been associated with an increased risk of CRC. 1α,25-Dihydroxyvitamin D3 [1,25(OH)2D3], the most active vitamin D metabolite, is a pleiotropic hormone that, through its binding to a transcription factor of the nuclear receptor superfamily, is a major regulator of the human genome. 1,25(OH)2D3 acts on colon carcinoma and stromal cells and displays tumor protective actions. Here, we review the variety of molecular mechanisms underlying the effects of 1,25(OH)2D3 in CRC, which affect multiple processes that are dysregulated during tumor initiation and progression. Additionally, we discuss the epidemiological data that associate vitamin D deficiency and CRC, and the most relevant randomized controlled trials of vitamin D3 supplementation conducted in both healthy individuals and CRC patients.
Collapse
Affiliation(s)
- Fábio Pereira
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Ourense, Spain
| | - Asunción Fernández-Barral
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (Hospital Universitario La Paz-Universidad Autónoma de Madrid), Spain
| | - María Jesús Larriba
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (Hospital Universitario La Paz-Universidad Autónoma de Madrid), Spain
| | - Antonio Barbáchano
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (Hospital Universitario La Paz-Universidad Autónoma de Madrid), Spain
| | - José Manuel González-Sancho
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ (Hospital Universitario La Paz-Universidad Autónoma de Madrid), Spain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| |
Collapse
|
11
|
Huang Z, Hu X, Wei Y, Lai Y, Qi J, Pang J, Huang K, Li H, Cai P. ADAMTSL2 is a potential prognostic biomarker and immunotherapeutic target for colorectal cancer: Bioinformatic analysis and experimental verification. PLoS One 2024; 19:e0303909. [PMID: 38814950 PMCID: PMC11139340 DOI: 10.1371/journal.pone.0303909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
The ADAMTS Like 2 (ADAMTSL2) mutation has been identified to be associated with different human genetic diseases. The role of ADAMTSL2 is unclear in colorectal cancer (CRC). The study investigated the expression of ADAMTSL2 in both pan cancer and CRC, using data from The Cancer Genome Atlas (TCGA) database to assess its diagnostic value. The study examined the correlation between ADAMTSL2 expression levels and clinical characteristics, as well as prognosis in CRC. The study explored potential regulatory networks involving ADAMTSL2, including its association with immune infiltration, immune checkpoint genes, tumor mutational burden (TMB) / microsatellite instability (MSI), tumor stemness index (mRNAsi), and drug sensitivity in CRC. ADAMTSL2 expression was validated using GSE71187 and quantitative real-time PCR (qRT-PCR). ADAMTSL2 was aberrantly expressed in pan cancer and CRC. An increased level of ADAMTSL2 expression in patients with CRC was significantly associated with the pathologic N stage (p < 0.001), pathologic stage (p < 0.001), age (p < 0.001), histological type (p < 0.001), and neoplasm type (p = 0.001). The high expression of ADAMTSL2 in patients with CRC was found to be significantly associated with a poorer overall survival (OS) (HR: 1.67; 95% CI: 1.18-2.38; p = 0.004), progression-free survival (PFS) (HR: 1.55; 95% CI: 1.14-2.11; p = 0.005) and disease-specific survival (DSS) (HR: 1.83; 95% CI: 1.16-2.89; p = 0.010). The expression of ADAMTSL2 in patients with CRC (p = 0.009) was identified as an independent prognostic determinant. ADAMTSL2 was associated with extracellular matrix receptor (ECM-receptor) interaction, transforming growth factor β (TGF-β) signaling pathway, and more. ADAMTSL2 expression was correlated with immune infiltration, immune checkpoint genes, TMB / MSI and mRNAsi in CRC. ADAMTSL2 expression was significantly and negatively correlated with 1-BET-762, Trametinib, and WZ3105 in CRC. ADAMTSL2 was significantly upregulated in CRC cell lines. The high expression of ADAMTSL2 is significantly correlated with lower OS and immune infiltration of CRC. ADAMTSL2 may be a potential prognostic biomarker and immunotherapeutic target for CRC patients.
Collapse
Affiliation(s)
- Zhe Huang
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xu Hu
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yiqiu Wei
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yousheng Lai
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jiaming Qi
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jinglin Pang
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kang Huang
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huagui Li
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Pengzhu Cai
- Department of Anorectal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
12
|
Zhang X, Zhou W, Wu C, Jiang J, Guo Q, Feng L, Cheng X, Zhang X. Cetuximab inhibits colorectal cancer development through inactivating the Wnt/β-catenin pathway and modulating PLCB3 expression. Sci Rep 2024; 14:10642. [PMID: 38724565 PMCID: PMC11081956 DOI: 10.1038/s41598-024-59676-2] [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: 08/12/2023] [Accepted: 04/13/2024] [Indexed: 05/12/2024] Open
Abstract
Colorectal cancer (CRC) often necessitates cetuximab (an EGFR-targeting monoclonal antibody) for treatment. Despite its clinical utility, the specific operative mechanism of cetuximab remains elusive. This research investigated the influence of PLCB3, a potential CRC oncogene, on cetuximab treatment. We extracted differentially expressed genes from the GSE140973, the overlapping genes combined with 151 Wnt/β-Catenin signaling pathway-related genes were identified. Then, we conducted bioinformatics analysis to pinpoint the hub gene. Subsequently, we investigated the clinical expression characteristics of this hub gene, through cell experimental, scrutinized the impact of cetuximab and PLCB3 on CRC cellular progression. The study identified 26 overlapping genes. High expression of PLCB3, correlated with poorer prognosis. PLCB3 emerged as a significant oncogene associated with patient prognosis. In vitro tests revealed that cetuximab exerted a cytotoxic effect on CRC cells, with PLCB3 knockdown inhibiting CRC cell progression. Furthermore, cetuximab treatment led to a reduction in both β-catenin and PLCB3 expression, while simultaneously augmenting E-cadherin expression. These findings revealed PLCB3 promoted cetuximab inhibition on Wnt/β-catenin signaling. Finally, simultaneous application of cetuximab with a Wnt activator (IM12) and PLCB3 demonstrated inhibited CRC proliferation, migration, and invasion. The study emphasized the pivotal role of PLCB3 in CRC and its potential to enhance the efficacy of cetuximab treatment. Furthermore, cetuximab suppressed Wnt/β-catenin pathway to modulate PLCB3 expression, thus inhibiting colorectal cancer progression. This study offered fresh perspectives on cetuximab mechanism in CRC.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Fengxian District Center Hospital Graduate Student Training Base, Jinzhou Medical University, No. 6600 Nanfeng Road, Shanghai, 201499, China
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Wenming Zhou
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Chenqu Wu
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Jun Jiang
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Qianqian Guo
- Fengxian District Center Hospital Graduate Student Training Base, Jinzhou Medical University, No. 6600 Nanfeng Road, Shanghai, 201499, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Xun Cheng
- Endoscopy Center, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Xingxing Zhang
- Fengxian District Center Hospital Graduate Student Training Base, Jinzhou Medical University, No. 6600 Nanfeng Road, Shanghai, 201499, China.
- Department of Gastroenterology, Shanghai Jiaotong University Affiliated Sixth People Hospital South Campus, No. 6600 Nanfeng Road, Shanghai, 201499, China.
| |
Collapse
|
13
|
Naser AN, Xing T, Tatum R, Lu Q, Boyer PJ, Chen YH. Colonic crypt stem cell functions are controlled by tight junction protein claudin-7 through Notch/Hippo signaling. Ann N Y Acad Sci 2024; 1535:92-108. [PMID: 38598500 PMCID: PMC11111361 DOI: 10.1111/nyas.15137] [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: 07/13/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The tight junction protein claudin-7 is essential for tight junction function and intestinal homeostasis. Cldn7 deletion in mice leads to an inflammatory bowel disease-like phenotype exhibiting severe intestinal epithelial damage, weight loss, inflammation, mucosal ulcerations, and epithelial hyperplasia. Claudin-7 has also been shown to be involved in cancer metastasis and invasion. Here, we test our hypothesis that claudin-7 plays an important role in regulating colonic intestinal stem cell function. Conditional knockout of Cldn7 in the colon led to impaired epithelial cell differentiation, hyperproliferative epithelium, a decrease in active stem cells, and dramatically altered gene expression profiles. In 3D colonoid culture, claudin-7-deficient crypts were unable to survive and form spheroids, emphasizing the importance of claudin-7 in stem cell survival. Inhibition of the Hippo pathway or activation of Notch signaling partially rescued the defective stem cell behavior. Concurrent Notch activation and Hippo inhibition resulted in restored colonoid survival, growth, and differentiation to the level comparable to those of wild-type derived crypts. In this study, we highlight the essential role of claudin-7 in regulating Notch and Hippo signaling-dependent colonic stem cell functions, including survival, self-renewal, and differentiation. These new findings may shed light on potential avenues to explore for drug development in colorectal cancer.
Collapse
Affiliation(s)
- Amna N. Naser
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
| | - Tiaosi Xing
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Neural and Behavioral Science Department, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Rodney Tatum
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
| | - Qun Lu
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Philip J. Boyer
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Yan-Hua Chen
- Department of Anatomy and Cell Biology, University of South Carolina, Columbia, South Carolina, USA
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
14
|
Radu P, Zurzu M, Tigora A, Paic V, Bratucu M, Garofil D, Surlin V, Munteanu AC, Coman IS, Popa F, Strambu V, Ramboiu S. The Impact of Cancer Stem Cells in Colorectal Cancer. Int J Mol Sci 2024; 25:4140. [PMID: 38673727 PMCID: PMC11050141 DOI: 10.3390/ijms25084140] [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: 03/10/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Despite incessant research, colorectal cancer (CRC) is still one of the most common causes of fatality in both men and women worldwide. Over time, advancements in medical treatments have notably enhanced the survival rates of patients with colorectal cancer. Managing metastatic CRC involves a complex tradeoff between the potential benefits and adverse effects of treatment, considering factors like disease progression, treatment toxicity, drug resistance, and the overall impact on the patient's quality of life. An increasing body of evidence highlights the significance of the cancer stem cell (CSC) concept, proposing that CSCs occupy a central role in triggering cancer. CSCs have been a focal point of extensive research in a variety of cancer types, including CRC. Colorectal cancer stem cells (CCSCs) play a crucial role in tumor initiation, metastasis, and therapy resistance, making them potential treatment targets. Various methods exist for isolating CCSCs, and understanding the mechanisms of drug resistance associated with them is crucial. This paper offers an overview of the current body of research pertaining to the comprehension of CSCs in colorectal cancer.
Collapse
Affiliation(s)
- Petru Radu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mihai Zurzu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Anca Tigora
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Vlad Paic
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mircea Bratucu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Dragos Garofil
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Valeriu Surlin
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Alexandru Claudiu Munteanu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Ionut Simion Coman
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
- General Surgery Department, “Bagdasar-Arseni” Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Florian Popa
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Victor Strambu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Sandu Ramboiu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| |
Collapse
|
15
|
Zhou H, Chen M, Zhao C, Shao R, Xu Y, Zhao W. The Natural Product Secoemestrin C Inhibits Colorectal Cancer Stem Cells via p38-S100A8 Feed-Forward Regulatory Loop. Cells 2024; 13:620. [PMID: 38607060 PMCID: PMC11011747 DOI: 10.3390/cells13070620] [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: 02/24/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Cancer stem cells (CSCs) are closely associated with tumor initiation, metastasis, chemoresistance, and recurrence, which represent some of the primary obstacles to cancer treatment. Targeting CSCs has become an important therapeutic approach to cancer care. Secoemestrin C (Sec C) is a natural compound with strong anti-tumor activity and low toxicity. Here, we report that Sec C effectively inhibited colorectal CSCs and non-CSCs concurrently, mainly by inhibiting proliferation, self-renewal, metastasis, and drug resistance. Mechanistically, RNA-seq analysis showed that the pro-inflammation pathway of the IL17 axis was enriched, and its effector S100A8 was dramatically decreased in Sec C-treated cells, whose roles in the stemness of CSCs have not been fully clarified. We found that the overexpression of S100A8 hindered the anti-CSCs effect of Sec C, and S100A8 deficiency attenuated the stemness traits of CSCs to enhance the Sec C killing activity on them. Meanwhile, the p38 signal pathway, belonging to the IL17 downstream axis, can also mediate CSCs and counter with Sec C. Notably, we found that S100A8 upregulation increased the p38 protein level, and p38, in turn, promoted S100A8 expression. This indicated that p38 may have a mutual feedback loop with S100A8. Our study discovered that Sec C was a powerful anti-colorectal CSC agent, and that the positive feedback loop of p38-S100A8 mediated Sec C activity. This showed that Sec C could act as a promising clinical candidate in colorectal cancer treatment, and S100A8 could be a prospective drug target.
Collapse
Affiliation(s)
- Huimin Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (H.Z.); (C.Z.); (R.S.)
| | - Minghua Chen
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Tiantan Xili, Beijing 100050, China;
| | - Cong Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (H.Z.); (C.Z.); (R.S.)
| | - Rongguang Shao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (H.Z.); (C.Z.); (R.S.)
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Tiantan Xili, Beijing 100050, China;
| | - Wuli Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (H.Z.); (C.Z.); (R.S.)
| |
Collapse
|
16
|
Cao X, Liu Z, Qin X, Gu Y, Huang Y, Qian Y, Wang Z, Li H, Zhu Q, Wei W. LoC-SERS platform for rapid and sensitive detection of colorectal cancer protein biomarkers. Talanta 2024; 270:125563. [PMID: 38134815 DOI: 10.1016/j.talanta.2023.125563] [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: 04/25/2023] [Revised: 12/13/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023]
Abstract
Colorectal cancer (CRC) remains a significant contributor to the global mortality rate, and a single biomarker cannot meet the specificity required for CRC screening. To this end, we developed a multiplexed, pump-free surface-enhanced Raman scattering (SERS) microfluidic chip (LoC-SERS) using a one-step recognition release mechanism; the aptamer-functionalized novel Au nanocrown array (AuNCA) was used as the detection element embedded in the detection zone of the platform for rapid and specific detection of protein markers in multiple samples simultaneously. Here, the corresponding aptamer specifically captured the protein marker, causing the complementary strand of the aptamer carrying the Raman signal molecule to be shed, reducing the SERS signal. Based on this platform, sensitive and specific detection of the target can be accomplished within 15 min with detection limits of 0.031 pg/mL (hnRNP A1) and 0.057 pg/mL (S100P). Meanwhile, the platform was consistent with ELISA results when used to test clinical. By substituting different aptamers, this platform can provide a new solution for the rapid and sensitive detection of protein markers, which has promising applications in future disease detection.
Collapse
Affiliation(s)
- Xiaowei Cao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Zhengqing Liu
- Department of Endocrinology, Suzhou Ninth People's Hospital, Suzhou, China
| | - Xiaogang Qin
- Department of Gastroenterology, Traditional Chinese Medicine Hospital of Tongzhou District, Nantong, Jiangsu, China
| | - Yuexing Gu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yong Huang
- Department of General Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Yayun Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Zhenguang Wang
- Department of General Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hongbo Li
- Department of General Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Qunshan Zhu
- Department of General Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Wei Wei
- Department of General Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| |
Collapse
|
17
|
Zheng HD, Huang QY, Huang QM, Ke XT, Ye K, Lin S, Xu JH. T2-weighted imaging-based radiomic-clinical machine learning model for predicting the differentiation of colorectal adenocarcinoma. World J Gastrointest Oncol 2024; 16:819-832. [PMID: 38577440 PMCID: PMC10989374 DOI: 10.4251/wjgo.v16.i3.819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/30/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The study on predicting the differentiation grade of colorectal cancer (CRC) based on magnetic resonance imaging (MRI) has not been reported yet. Developing a non-invasive model to predict the differentiation grade of CRC is of great value. AIM To develop and validate machine learning-based models for predicting the differentiation grade of CRC based on T2-weighted images (T2WI). METHODS We retrospectively collected the preoperative imaging and clinical data of 315 patients with CRC who underwent surgery from March 2018 to July 2023. Patients were randomly assigned to a training cohort (n = 220) or a validation cohort (n = 95) at a 7:3 ratio. Lesions were delineated layer by layer on high-resolution T2WI. Least absolute shrinkage and selection operator regression was applied to screen for radiomic features. Radiomics and clinical models were constructed using the multilayer perceptron (MLP) algorithm. These radiomic features and clinically relevant variables (selected based on a significance level of P < 0.05 in the training set) were used to construct radiomics-clinical models. The performance of the three models (clinical, radiomic, and radiomic-clinical model) were evaluated using the area under the curve (AUC), calibration curve and decision curve analysis (DCA). RESULTS After feature selection, eight radiomic features were retained from the initial 1781 features to construct the radiomic model. Eight different classifiers, including logistic regression, support vector machine, k-nearest neighbours, random forest, extreme trees, extreme gradient boosting, light gradient boosting machine, and MLP, were used to construct the model, with MLP demonstrating the best diagnostic performance. The AUC of the radiomic-clinical model was 0.862 (95%CI: 0.796-0.927) in the training cohort and 0.761 (95%CI: 0.635-0.887) in the validation cohort. The AUC for the radiomic model was 0.796 (95%CI: 0.723-0.869) in the training cohort and 0.735 (95%CI: 0.604-0.866) in the validation cohort. The clinical model achieved an AUC of 0.751 (95%CI: 0.661-0.842) in the training cohort and 0.676 (95%CI: 0.525-0.827) in the validation cohort. All three models demonstrated good accuracy. In the training cohort, the AUC of the radiomic-clinical model was significantly greater than that of the clinical model (P = 0.005) and the radiomic model (P = 0.016). DCA confirmed the clinical practicality of incorporating radiomic features into the diagnostic process. CONCLUSION In this study, we successfully developed and validated a T2WI-based machine learning model as an auxiliary tool for the preoperative differentiation between well/moderately and poorly differentiated CRC. This novel approach may assist clinicians in personalizing treatment strategies for patients and improving treatment efficacy.
Collapse
Affiliation(s)
- Hui-Da Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Qiao-Yi Huang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Qi-Ming Huang
- Department of Computed Tomography/Magnetic Resonance Imaging, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Xiao-Ting Ke
- Department of Computed Tomography/Magnetic Resonance Imaging, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Kai Ye
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Jian-Hua Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
| |
Collapse
|
18
|
Cao Q, Zhao M, Su Y, Liu S, Lin Y, Da H, Yue C, Liu Y, Jing D, Zhao Q, Liu N, Du J, Zuo Z, Fu Y, Chen A, Birnbaumer L, Yang Y, Dai B, Gao X. Chronic Stress Dampens Lactobacillus Johnsonii-Mediated Tumor Suppression to Enhance Colorectal Cancer Progression. Cancer Res 2024; 84:771-784. [PMID: 38190716 DOI: 10.1158/0008-5472.can-22-3705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 09/30/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Colorectal cancer development and outcome are impacted by modifiable risk factors, including psychologic stress. The gut microbiota has also been shown to be linked to psychologic factors. Here, we found a marked deteriorative effect of chronic stress in multiple colorectal cancer models, including chemically induced (AOM/DSS), genetically engineered (APCmin/+), and xenograft tumor mouse models. RNA sequencing data from colon tissues revealed that expression of stemness-related genes was upregulated in the stressed colorectal cancer group by activated β-catenin signaling, which was further confirmed by results from ex vivo organoid analyses as well as in vitro and in vivo cell tumorigenicity assays. 16S rRNA sequencing of the gut microbiota showed that chronic stress disrupted gut microbes, and antibiotic treatment and fecal microbiota transplantation abolished the stimulatory effects of chronic stress on colorectal cancer progression. Stressed colorectal cancer mice displayed a significant decrease in Lactobacillus johnsonii (L. johnsonii) abundance, which was inversely correlated with tumor load. Moreover, protocatechuic acid (PCA) was identified as a beneficial metabolite produced by L. johnsonii based on metabolome sequencing and LC/MS-MS analysis. Replenishment of L. johnsonii or PCA blocked chronic stress-induced colorectal cancer progression by decreasing β-catenin expression. Furthermore, PCA activated the cGMP pathway, and the cGMP agonist sildenafil abolished the effects of chronic stress on colorectal cancer. Altogether, these data identify that stress impacts the gut microbiome to support colorectal cancer progression. SIGNIFICANCE Chronic stress stimulates cancer stemness by reducing the intestinal abundance of L. johnsonii and its metabolite PCA to enhance β-catenin signaling, forming a basis for potential strategies to circumvent stress-induced cancer aggressiveness. See related commentary by McCollum and Shah, p. 645.
Collapse
Affiliation(s)
- Qiuhua Cao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Mingrui Zhao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yali Su
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Siliang Liu
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yanting Lin
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Huijuan Da
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Chongxiu Yue
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yiming Liu
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Dongquan Jing
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Qixiang Zhao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, P.R. China
| | - Juan Du
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, P.R. China
| | - Zhanjie Zuo
- Thoracic Cancer Treatment Center, Armed Police Beijing Corps Hospital, Beijing, P.R. China
| | - Yao Fu
- Department of Pathology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Anqi Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, P.R. China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Xinghua Gao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
19
|
Wang M, Tang L, Chen S, Wang L, Wu J, Zhong C, Li Y, Chen Y. ZNF217-activated Notch signaling mediates sulforaphane-suppressed stem cell properties in colorectal cancer. J Nutr Biochem 2024; 125:109551. [PMID: 38134973 DOI: 10.1016/j.jnutbio.2023.109551] [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: 05/13/2023] [Revised: 11/18/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Cancer stem cells (CSCs) are known to contribute to the progression of colorectal cancer (CRC). However, understanding of the molecular mechanisms and key factors involved in CRC is still insufficient to identify therapeutic targets against colorectal CSCs. In an effort to identify such mechanisms, we conducted bioinformatics analyses to evaluate the expression patterns in tumor and normal colorectal tissues, leading us to focus on the role of the ZNF217/Notch1 axis in mediating stem cell properties in CRC. Our findings revealed that ZNF217 overexpression activated self-renewal ability, expression of colorectal CSC markers, and Notch signaling in CRC. Dual-luciferase reporter assay suggested a role for ZNF217 in targeting Notch1 to activate Notch signaling. We observed that the promotional effects of Notch signaling, as well as CSC markers, under ZNF217 overexpression were attenuated after Notch1 knockdown. In addition to in vitro data, our in vivo results confirmed the inhibitory effect of sulforaphane on the tumorigenicity of CSCs, depicted the suppressive role of sulforaphane on colorectal CSCs mediated by the ZNF217/Notch1 axis, thereby providing new targetable vulnerabilities and therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Menghuan Wang
- Department of Children Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - LvYuwei Tang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Liudan Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Jinyi Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Yadong Li
- Department of Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yue Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Chen Y, Zhang YH, Li J, Shi L, Xie JC, Han X, Chen YT, Xiang M, Li BW, Xing HR, Wang JY. Novel lncRNA Gm33149 modulates metastatic heterogeneity in melanoma by regulating the miR-5623-3p/Wnt axis via exosomal transfer. Cancer Gene Ther 2024; 31:364-375. [PMID: 38072970 DOI: 10.1038/s41417-023-00707-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 03/16/2024]
Abstract
The high mortality rate associated with melanoma primarily results from metastasis and recurrence. However, the precise mechanisms driving these processes remain poorly understood. Intercellular communication between cancer cells and non-cancer cells significantly influences the tumor microenvironment and plays a crucial role in metastasis. Therefore, our current study aims to investigate the role and mechanism of long non-coding RNAs (lncRNAs) in regulating the interaction between melanoma cancer stem cells (CSCs) and non-CSCs during the metastatic colonization process. This study has characterized a novel lncRNA called Gm33149. Importantly, we provide evidence for the first time that Gm33149, originating from highly metastatic melanoma stem cells (OL-SD), can be packaged into exosomes and transferred to low-metastatic nonstem cells (OL). Once internalized by OL cells, Gm33149 exerts its function through a competitive endogenous RNA mechanism (ceRNA) involving miR-5623-3p. Specifically, Gm33149 competitively binds to miR-5623-3p, thereby activating the Wnt signaling pathway and promoting the acquisition of a more aggressive metastatic phenotype by OL cells. In summary, our findings suggest that targeting lncRNA Gm33149 within extracellular vesicles could potentially serve as a therapeutic strategy for the treatment of metastatic melanoma. Schematic representation of the mechanisms underlying the pro-metastatic activity of lncRNA Gm33149 mediated by exosomal transfer. The figure illustrates the key mechanisms involved in the pro-metastatic activity of lncRNA Gm33149 through exosomal transfer. Melanoma stem cells (OLSD) release exosomes containing lncRNA Gm33149. These exosomes are taken up by non-stem melanoma cells (OL), delivering lncRNA Gm33149 to the recipient cells. Within OL cells, lncRNA Gm33149 functions as a competitive endogenous RNA (ceRNA), sequestering miR-5623-3p. This sequestration prevents miR-5623-3p from binding to its target genes, thereby activating the Wnt signaling pathway. The activated Wnt signaling pathway enhances the migration, invasion, and metastatic colonization capabilities of OL cells. The transfer of lncRNA Gm33149 via exosomes contributes to OL cells acquiring "metastatic competency" while promoting their metastatic colonization. These findings underscore the importance of lncRNA Gm33149 in intercellular communication and the metastatic progression of melanoma.
Collapse
Affiliation(s)
- Yan Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yu-Han Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Lei Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jia-Cheng Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xue Han
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yu-Ting Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Meng Xiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Bo-Wen Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - H Rosie Xing
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| | - Jian-Yu Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
Jones J, Shi Q, Nath RR, Brito IL. Keystone pathobionts associated with colorectal cancer promote oncogenic reprograming. PLoS One 2024; 19:e0297897. [PMID: 38363784 PMCID: PMC10871517 DOI: 10.1371/journal.pone.0297897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/12/2024] [Indexed: 02/18/2024] Open
Abstract
Fusobacterium nucleatum (Fn) and enterotoxigenic Bacteroides fragilis (ETBF) are two pathobionts consistently enriched in the gut microbiomes of patients with colorectal cancer (CRC) compared to healthy counterparts and frequently observed for their direct association within tumors. Although several molecular mechanisms have been identified that directly link these organisms to features of CRC in specific cell types, their specific effects on the epithelium and local immune compartment are not well-understood. To fill this gap, we leveraged single-cell RNA sequencing (scRNA-seq) on wildtype mice and mouse model of CRC. We find that Fn and ETBF exacerbate cancer-like transcriptional phenotypes in transit-amplifying and mature enterocytes in a mouse model of CRC. We also observed increased T cells in the pathobiont-exposed mice, but these pathobiont-specific differences observed in wildtype mice were abrogated in the mouse model of CRC. Although there are similarities in the responses provoked by each organism, we find pathobiont-specific effects in Myc-signaling and fatty acid metabolism. These findings support a role for Fn and ETBF in potentiating tumorigenesis via the induction of a cancer stem cell-like transit-amplifying and enterocyte population and the disruption of CTL cytotoxic function.
Collapse
Affiliation(s)
- Josh Jones
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Qiaojuan Shi
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Rahul R. Nath
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Ilana L. Brito
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
22
|
Li Y, Shi J, Liu Z, Lin Y, Xie A, Sun W, Liu J, Liang J. Regulation of the migration of colorectal cancer stem cells via the TLR4/MyD88 signaling pathway by the novel surface marker CD14 following LPS stimulation. Oncol Lett 2024; 27:60. [PMID: 38192670 PMCID: PMC10773188 DOI: 10.3892/ol.2023.14194] [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: 09/07/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
Cell surface markers are most widely used in the study of cancer stem cells (CSCs). However, cell surface markers that are safely and stably expressed in CSCs have yet to be identified. Colonic CSCs express leukocyte CD14. CD14 binding to the ligand lipopolysaccharide (LPS) is involved in the inflammatory response via the Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) signaling pathway. TLR4 and MyD88 have been reported to promote the proliferation, metastasis and tumorigenicity of colon cancer cells, which is consistent with the characteristics of CSCs. In the present study, the proposed experimental method to detect cell proliferation, metastasis and tumorigenesis was used to confirm that, under LPS stimulation, CD14 promoted the proliferation, migration and tumorigenesis of colonic CSCs via the TLR4/MyD88 signaling pathway. Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine assays were used to assess the proliferation and migration of the cells. Colony formation and nude mouse xenograft assays were used to assess the capacity of cells to form tumors. Using western blotting and reverse transcription-quantitative PCR, the mRNA and protein levels of CD14, TLR4 and MyD88 were examined. It was confirmed that CD14 promoted the proliferation, metastasis and tumorigenesis of colon CSCs in response to LPS stimulation via the TLR4/MyD88 signaling pathway, and CD14+ colon cancer cells were successfully isolated and sorted. According to the results of proliferation assay, it was determined that CD14 regulated the LPS-induced proliferation of colon CSCs. CD14, TLR4 and MyD88 protein and mRNA expression was upregulated in colon CSCs in response to LPS stimulation. This indicates a potential novel target for colon CSC-related studies.
Collapse
Affiliation(s)
- Yufei Li
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jiayi Shi
- School of Life Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Zhixin Liu
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yonggang Lin
- Department of Extracorporeal Circulation, Mudanjiang Cardiovascular Disease Hospital, Mudanjiang, Heilongjiang 157011, P.R. China
| | - An Xie
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Wenxiu Sun
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jiaqi Liu
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jun Liang
- Morphology Laboratory, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
23
|
Li D, Wang L, Jiang B, Jing Y, Li X. Improving cancer immunotherapy by preventing cancer stem cell and immune cell linking in the tumor microenvironment. Biomed Pharmacother 2024; 170:116043. [PMID: 38128186 DOI: 10.1016/j.biopha.2023.116043] [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: 10/18/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer stem cells are the key link between malignant tumor progression and drug resistance. This cell population has special properties that are different from those of conventional tumor cells, and the role of cancer stem cell-related exosomes in progression of tumor malignancy is becoming increasingly clear. Cancer stem cell-derived exosomes carry a variety of functional molecules involved in regulation of the microenvironment, especially with regard to immune cells, but how these exosomes exert their functions and the specific mechanisms need to be further clarified. Here, we summarize the role of cancer stem cell exosomes in regulating immune cells in detail, aiming to provide new insights for subsequent targeted drug development and clinical strategy formulation.
Collapse
Affiliation(s)
- Dongyu Li
- Department of General Surgery & VIP In-Patient Ward, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Bo Jiang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Yuchen Jing
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Xuan Li
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China.
| |
Collapse
|
24
|
Dalmasso G, Cougnoux A, Faïs T, Bonnin V, Mottet-Auselo B, Nguyen HTT, Sauvanet P, Barnich N, Jary M, Pezet D, Delmas J, Bonnet R. Colibactin-producing Escherichia coli enhance resistance to chemotherapeutic drugs by promoting epithelial to mesenchymal transition and cancer stem cell emergence. Gut Microbes 2024; 16:2310215. [PMID: 38374654 PMCID: PMC10880512 DOI: 10.1080/19490976.2024.2310215] [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: 05/21/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Human colorectal cancers (CRCs) are readily colonized by colibactin-producing E. coli (CoPEC). CoPEC induces DNA double-strand breaks, DNA mutations, genomic instability, and cellular senescence. Infected cells produce a senescence-associated secretory phenotype (SASP), which is involved in the increase in tumorigenesis observed in CRC mouse models infected with CoPEC. This study investigated whether CoPEC, and the SASP derived from CoPEC-infected cells, impacted chemotherapeutic resistance. Human intestinal epithelial cells were infected with the CoPEC clinical 11G5 strain or with its isogenic mutant, which is unable to produce colibactin. Chemotherapeutic resistance was assessed in vitro and in a xenograft mouse model. Expressions of cancer stem cell (CSC) markers in infected cells were investigated. Data were validated using a CRC mouse model and human clinical samples. Both 11G5-infected cells, and uninfected cells incubated with the SASP produced by 11G5-infected cells exhibited an increased resistance to chemotherapeutic drugs in vitro and in vivo. This finding correlated with the induction of the epithelial to mesenchymal transition (EMT), which led to the emergence of cells exhibiting CSC features. They grew on ultra-low attachment plates, formed colonies in soft agar, and overexpressed several CSC markers (e.g. CD133, OCT-3/4, and NANOG). In agreement with these results, murine and human CRC biopsies colonized with CoPEC exhibited higher expression levels of OCT-3/4 and NANOG than biopsies devoid of CoPEC. Conclusion: CoPEC might aggravate CRCs by inducing the emergence of cancer stem cells that are highly resistant to chemotherapy.
Collapse
Affiliation(s)
- Guillaume Dalmasso
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Antony Cougnoux
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Tiphanie Faïs
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Virginie Bonnin
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Benoit Mottet-Auselo
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Hang TT Nguyen
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Pierre Sauvanet
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Centre de référence de la résistance aux antibiotiques, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Nicolas Barnich
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Marine Jary
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Chirurgie Digestive, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Denis Pezet
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Chirurgie Digestive, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Julien Delmas
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Richard Bonnet
- Inserm U1071, USC-INRAe INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire, Clermont-Ferrand, France
- Centre de référence de la résistance aux antibiotiques, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| |
Collapse
|
25
|
Azarifar Z, Amini R, Tanzadehpanah H, Afshar S, Najafi R. In vitro co-delivery of 5-fluorouracil and all-trans retinoic acid by PEGylated liposomes for colorectal cancer treatment. Mol Biol Rep 2023; 50:10047-10059. [PMID: 37902908 DOI: 10.1007/s11033-023-08888-2] [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: 04/12/2023] [Accepted: 10/04/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Single-target inhibitors have not been successful in cancer treatment due to the development of drug resistance. Nevertheless, therapeutic agents capable of simultaneously inhibiting multiple targets have revealed encouraging results in inducing apoptosis and overcoming drug resistance in cancerous cells. Here, we designed a composite liposomal nano-carrier co-loading 5-Fluorouracil (5-FU) with all-trans retinoic acid (ATRA) to assess anticancer efficacy of the combined drugs in colorectal cancer (CRC). METHODS A PEGylated liposomal nano-carrier with phospholipid/cholesterol/DSPE-PEG (2000) was synthesized by the thin film hydration technique for co-delivery of ATRA and 5-FU. After characterizing, the role of 5-FU and ATRA co-loaded liposomal nano-carrier in proliferation, epithelial-mesenchymal transition (EMT), apoptosis, and cancer stem cells (CSCs) were investigated by using colony forming and MTT assay, RT-qPCR and Annexin V/PI kit. RESULTS The average size of liposomes (LPs) was < 150 nm with uniform size distribution. Drug release analyses indicated that both ATRA and 5-FU could simultaneously release from LPs in a sustained release manner. The synergistic inhibitory effects of ATRA and 5-FU loaded in LPs were verified with a combination index of 0.43. Dual drug LPs showed the highest cytotoxicity, enhanced inhibition of cell proliferation, increased apoptotic potential, decreased CSCs, and attenuated EMT-associated biomarkers. Also, dual drug LPs decreased β-catenin gene expression more than other liposomal formulations. CONCLUSION These findings suggest that using LPs to achieve a synergistic effect of ATRA and 5-FU is an effectual approach to increase the therapeutic effect of 5-FU toward CRC cells.
Collapse
Affiliation(s)
- Zahra Azarifar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
26
|
Zheng R, Xu Z, Zeng Y, Wang E, Li M. SPIDE: A single cell potency inference method based on the local cell-specific network entropy. Methods 2023; 220:90-97. [PMID: 37952704 DOI: 10.1016/j.ymeth.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023] Open
Abstract
For a given single cell RNA-seq data, it is critical to pinpoint key cellular stages and quantify cells' differentiation potency along a differentiation pathway in a time course manner. Currently, several methods based on the entropy of gene functions or PPI network have been proposed to solve the problem. Nevertheless, these methods still suffer from the inaccurate interactions and noises originating from scRNA-seq profile. In this study, we proposed a cell potency inference method based on cell-specific network entropy, called SPIDE. SPIDE introduces the local weighted cell-specific network for each cell to maintain cell heterogeneity and calculates the entropy by incorporating gene expression with network structure. In this study, we compared three cell entropy estimation models on eight scRNA-Seq datasets. The results show that SPIDE obtains consistent conclusions with real cell differentiation potency on most datasets. Moreover, SPIDE accurately recovers the continuous changes of potency during cell differentiation and significantly correlates with the stemness of tumor cells in Colorectal cancer. To conclude, our study provides a universal and accurate framework for cell entropy estimation, which deepens our understanding of cell differentiation, the development of diseases and other related biological research.
Collapse
Affiliation(s)
- Ruiqing Zheng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Ziwei Xu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yanping Zeng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Edwin Wang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Alberta, Canada
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
27
|
Chang Y, Chen L, Tang J, Chen G, Ji J, Xu M. USP7-mediated JUND suppresses RCAN2 transcription and elevates NFATC1 to enhance stem cell property in colorectal cancer. Cell Biol Toxicol 2023; 39:3121-3140. [PMID: 37535148 DOI: 10.1007/s10565-023-09822-9] [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: 03/06/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023]
Abstract
Cancer stem cells (CSCs) encompass a subset of highly aggressive tumor cells that are involved in tumor initiation and progression. This study investigates the function of regulator of calcineurin 2 (RCAN2) in the stem cell property in colorectal cancer (CRC). By analyzing four GEO datasets, we obtained RCAN2 as a stemness-related gene in CRC. RCAN2 was poorly expressed in CRC tissues and cells, especially in CSCs. RCAN2 restoration reduced calcineurin activity and promoted phosphorylation and degradation of nuclear factor of activated T cells 1 (NFATC1) protein, leading to reduced stemness of CSCs. JunD proto-oncogene (JUND), whose protein level was increased in CRC samples and CRC stem cells, bound to RCAN2 and suppressed its transcription. The abundant ubiquitin specific peptidase 7 (USP7) in CSCs enhanced JUND protein stability through deubiquitination modification. Lentivirus-mediated knockdown of USP7 or JUND also blocked the calcineurin-NFATC1 signaling and reduced the protein levels of stemness-related proteins. Moreover, the USP7 knockdown weakened the colony/sphere formation ability as well as the tumorigenicity of CSCs, and it reduced the CSC content in xenograft tumors. However, further restoration of JUND rescued the stemness of the CSCs. Overall, this study demonstrates that USP7-mediated JUND suppresses RCAN2 transcription and activates NFATC1 to enhance stem cell property in CRC. 1. RCAN2 is poorly expressed in CRC tissues and cells and especially in CSCs. 2. RCAN2 reduces stemness of CSCs by blocking calcineurin-NFATC1 signal transduction. 3. JUND binds to RCAN2 promoter to suppresses RCAN2 transcription. 4. USP7 enhances JUND protein stability via deubiquitination modification. 5. Downregulation of USP7 or JUND restores RCAN2 level and suppresses stemness of CSCs.
Collapse
Affiliation(s)
- Yunli Chang
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China
| | - Lingling Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China
| | - Jie Tang
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China
| | - Guoyu Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China
| | - Jieru Ji
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China
| | - Ming Xu
- Department of Gastroenterology, Pudong New Area People's Hospital, No. 490, Chuanhuan South Road, Pudong New Area, Shanghai, 201299, People's Republic of China.
| |
Collapse
|
28
|
Wardah ZH, Chaudhari HG, Prajapati V, Raol GG. Application of statistical methodology for the optimization of L-glutaminase enzyme production from Streptomyces pseudogriseolus ZHG20 under solid-state fermentation. J Genet Eng Biotechnol 2023; 21:138. [PMID: 37999820 PMCID: PMC10673782 DOI: 10.1186/s43141-023-00618-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Actinomycetes are excellent microbial sources for various chemical structures like enzymes, most of which are used in pharmaceutical and industrial products. Actinomycetes are preferred sources of enzymes due to their high ability to produce extracellular enzymes. L-glutaminase has proven its essential role as a pharmaceutical agent in cancer therapy and an economic agent in the food industry. The current study aimed to screen the potent L-glutaminase producer and optimize the production media for maximum enzyme yield using one factor at a time (OFAT) approach and statistical approaches under solid-state fermentation (SSF). RESULTS Out of 20 actinomycetes strains isolated from rhizosphere soil, 5 isolates produced extracellular L-glutaminase. One isolate was chosen as the most potent strain, and identified as Streptomyces pseudogriseolus ZHG20 based on 16S rRNA. The production and optimization process were carried out under SSF, after optimization using OFAT method, the enzyme production increased up to 884.61 U/gds. Further, statistical strategy, response surface methodology (RSM), and central composite design (CCD) were employed for the level optimization of significant media component (p < 0.05), i.e., wheat bran, sesame oil cake, and corn steep liquor which are leading to increase 3.21-fold L-glutaminase production as compared to unoptimized media. CONCLUSIONS The presented investigation reveals the optimization of various physicochemical parameters using OFAT and RSM-CCD. Statistical approaches proved to be an effective method for increasing the yield of extracellular L-glutaminase from S. pseudogriseolus ZHG20 where L-glutaminase activity increased up to 1297.87 U/gds which is 3.21-fold higher than the unoptimized medium using a mixture of two solid substrates (wheat bran and sesame oil cake) incubated at pH 7.0 for 6 days at 33 °C.
Collapse
Affiliation(s)
- Zuhour Hussein Wardah
- Department of Microbiology, Shri Alpesh N. Patel PG Institute of Science and Research, Sardar Patel University, Vallabh Vidyanagar, Anand, Gujarat, 388001, India
| | - Hiral G Chaudhari
- Department of Microbiology, Shri Alpesh N. Patel PG Institute of Science and Research, Sardar Patel University, Vallabh Vidyanagar, Anand, Gujarat, 388001, India
| | - Vimalkumar Prajapati
- Division of Microbial and Environmental Biotechnology, Aspee Shakilam Biotechnology Institute, Navsari Agricultural University, Athwa Farm, Ghod Dod Road, Surat, Gujarat, 395007, India.
| | - Gopalkumar G Raol
- Shri R. P. Arts, Shri K.B. Commerce and Smt, BCJ Science College, Khambhat, 388620, Gujarat, India.
| |
Collapse
|
29
|
Malkawi W, Lutfi A, Afghan MK, Shah LM, Costandy L, Ramirez AB, George TC, Toor F, Salem AK, Kasi PM. Circulating tumour cell enumeration, biomarker analyses, and kinetics in patients with colorectal cancer and other GI malignancies. Front Oncol 2023; 13:1305181. [PMID: 38044994 PMCID: PMC10693413 DOI: 10.3389/fonc.2023.1305181] [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: 09/30/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023] Open
Abstract
Objective Most of the work in terms of liquid biopsies in patients with solid tumors is focused on circulating tumor DNA (ctDNA). Our aim was to evaluate the feasibility of using circulating tumor cells (CTCs) in peripheral blood samples from patients with advanced or metastatic gastrointestinal (GI) cancers. Methods In this prospective study, blood samples were collected from each patient in 2 AccuCyte® blood collection tubes and each tube underwent CTC analysis performed utilizing the RareCyte® platform. The results from both tubes were averaged and a total of 150 draws were done, with 281 unique reported results. The cadence of sampling was based on convenience sampling and piggybacked onto days of actual clinical follow-ups and treatment visits. The CTC results were correlated with patient- and tumor-related variables. Results Data from a total of 59 unique patients were included in this study. Patients had a median age of 58 years, with males representing 69% of the study population. More than 57% had received treatment prior to taking blood samples. The type of GI malignancy varied, with more than half the patients having colorectal cancer (CRC, 54%) followed by esophageal/gastric cancer (17%). The least common cancer was cholangiocarcinoma (9%). The greatest number of CTCs were found in patients with colorectal cancer (Mean: 15.8 per 7.5 ml; Median: 7.5 per 7.5 ml). In comparison, patients with pancreatic cancer (PC) had considerably fewer CTCs (Mean: 4.2 per 7.5 ml; Median: 3 per 7.5 ml). Additionally, we found that patients receiving treatment had significantly fewer CTCs than patients who were not receiving treatment (Median 2.7 versus 0.7). CTC numbers showed noteworthy disparities between patients with responding/stable disease in comparison to those with untreated/progressive disease (Median of 2.7 versus 0). When CTCs were present, biomarker analyses of the four markers human epidermal growth factor receptor 2 (HER2)/programmed death-ligand 1 (PD-L1)/Kiel 67 (Ki-67)/epidermal growth factor receptor (EGFR) was feasible. Single cell sequencing confirmed the tumor of origin. Conclusion Our study is one of the first prospective real-time studies evaluating CTCs in patients with GI malignancies. While ctDNA-based analyses are more common in clinical trials and practice, CTC analysis provides complementary information from a liquid biopsy perspective that is of value and worthy of continued research.
Collapse
Affiliation(s)
- Walla Malkawi
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa, Iowa, IA, United States
| | - Areeb Lutfi
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Maaz Khan Afghan
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Lamisha Mashiyat Shah
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| | | | | | | | - Fatima Toor
- Experimental Therapeutics Program, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa, IA, United States
- Department of Electrical and Computer Engineering, University of Iowa, Iowa, IA, United States
| | - Aliasger K. Salem
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa, Iowa, IA, United States
- Experimental Therapeutics Program, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa, IA, United States
| | - Pashtoon Murtaza Kasi
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
30
|
Rengganaten V, Huang CJ, Wang ML, Chien Y, Tsai PH, Lan YT, Ong HT, Chiou SH, Choo KB. Circular RNA ZNF800 (hsa_circ_0082096) regulates cancer stem cell properties and tumor growth in colorectal cancer. BMC Cancer 2023; 23:1088. [PMID: 37950151 PMCID: PMC10636831 DOI: 10.1186/s12885-023-11571-1] [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: 04/06/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Cancer stem cells form a rare cell population in tumors that contributes to metastasis, recurrence and chemoresistance in cancer patients. Circular RNAs (circRNAs) are post-transcriptional regulators of gene expression that sponge targeted microRNA (miRNAs) to affect a multitude of downstream cellular processes. We previously showed in an expression profiling study that circZNF800 (hsa_circ_0082096) was up-regulated in cancer stem cell-enriched spheroids derived from colorectal cancer (CRC) cell lines. METHODS Spheroids were generated in suspension spheroidal culture. The ZNF800 mRNA, pluripotency stem cell markers and circZNF800 levels were determined by quantitative RT-PCR. CircZNF800-miRNA interactions were shown in RNA pulldown assays and the miRNA levels determined by stem-loop qRT-PCR. The effects of circZNF800 on cell proliferation were tested by EdU staining followed by flowcytometry. Expression of stem cell markers CD44/CD133, Lgr5 and SOX9 was demonstrated in immunofluorescence microscopy. To manipulate the cellular levels of circZNF800, circZNF800 over-expression was achieved via transfection of in vitro synthesized and circularized circZNF800, and knockdown attained using a CRISPR-Cas13d-circZNF800 vector system. Xenografted nude mice were used to demonstrate effects of circZNF800 over-expression and knockdown on tumor growth in vivo. RESULTS CircZNF800 was shown to be over-expressed in late-stage tumor tissues of CRC patients. Data showed that circZNF800 impeded expression of miR-140-3p, miR-382-5p and miR-579-3p while promoted the mRNA levels of ALK/ACVR1C, FZD3 and WNT5A targeted by the miRNAs, as supported by alignments of seed sequences between the circZNF800-miRNA, and miRNA-mRNA paired interactions. Analysis in CRC cells and biopsied tissues showed that circZNF800 positively regulated the expression of intestinal stem cell, pluripotency and cancer stem cell markers, and promoted CRC cell proliferation, spheroid and colony formation in vitro, all of which are cancer stem cell properties. In xenografted mice, circZNF800 over-expression promoted tumor growth, while circZNF800 knockdown via administration of CRISPR Cas13d-circZNF800 viral particles at the CRC tumor sites impeded tumor growth. CONCLUSIONS CircZNF800 is an oncogenic factor that regulate cancer stem cell properties to lead colorectal tumorigenesis, and may be used as a predictive marker for tumor progression and the CRISPR Cas13d-circZNF800 knockdown strategy for therapeutic intervention of colorectal cancer.
Collapse
Affiliation(s)
- Vimalan Rengganaten
- Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, 43000, Kajang, Selangor, Malaysia
- Postgraduate Program, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000, Kajang, Malaysia
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Chiu-Jung Huang
- Department of Animal Science & Graduate Institute of Biotechnology, Chinese Culture University, Taipei, 11221, Taiwan
| | - Mong-Lien Wang
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Yuan-Tzu Lan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, 11221, Taiwan
| | - Hooi Tin Ong
- Centre for Cancer Research, Universiti Tunku Abdul Rahman, 43000, Kajang, Selangor, Malaysia
- Department of Preclinical Sciences, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sg Long, 43000, Kajang, Selangor, Malaysia
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11221, Taiwan.
| | - Kong Bung Choo
- Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, 43000, Kajang, Selangor, Malaysia.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11221, Taiwan.
- Department of Preclinical Sciences, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sg Long, 43000, Kajang, Selangor, Malaysia.
| |
Collapse
|
31
|
Islam MS, Gopalan V, Lam AK, Shiddiky MJA. Current advances in detecting genetic and epigenetic biomarkers of colorectal cancer. Biosens Bioelectron 2023; 239:115611. [PMID: 37619478 DOI: 10.1016/j.bios.2023.115611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Colorectal carcinoma (CRC) is the third most common cancer in terms of diagnosis and the second in terms of mortality. Recent studies have shown that various proteins, extracellular vesicles (i.e., exosomes), specific genetic variants, gene transcripts, cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and altered epigenetic patterns, can be used to detect, and assess the prognosis of CRC. Over the last decade, a plethora of conventional methodologies (e.g., polymerase chain reaction [PCR], direct sequencing, enzyme-linked immunosorbent assay [ELISA], microarray, in situ hybridization) as well as advanced analytical methodologies (e.g., microfluidics, electrochemical biosensors, surface-enhanced Raman spectroscopy [SERS]) have been developed for analyzing genetic and epigenetic biomarkers using both optical and non-optical tools. Despite these methodologies, no gold standard detection method has yet been implemented that can analyze CRC with high specificity and sensitivity in an inexpensive, simple, and time-efficient manner. Moreover, until now, no study has critically reviewed the advantages and limitations of these methodologies. Here, an overview of the most used genetic and epigenetic biomarkers for CRC and their detection methods are discussed. Furthermore, a summary of the major biological, technical, and clinical challenges and advantages/limitations of existing techniques is also presented.
Collapse
Affiliation(s)
- Md Sajedul Islam
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast Campus, Southport, QLD, 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast Campus, Southport, QLD, 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia.
| | - Alfred K Lam
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast Campus, Southport, QLD, 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia; Pathology Queensland, Gold Coast University Hospital, Southport, QLD, 4215, Australia
| | - Muhammad J A Shiddiky
- Rural Health Research Institute, Charles Sturt University, Orange, NSW, 2800, Australia.
| |
Collapse
|
32
|
Osei GY, Adu-Amankwaah J, Koomson S, Beletaa S, Ahmad MK, Asiamah EA, Smith-Togobo C, Abdul Razak SR. Revolutionizing colorectal cancer treatment: unleashing the potential of miRNAs in targeting cancer stem cells. Future Oncol 2023; 19:2369-2382. [PMID: 37970643 DOI: 10.2217/fon-2023-0426] [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] [Indexed: 11/17/2023] Open
Abstract
Colorectal cancer (CRC) is a significant contributor to cancer mortality worldwide, and the presence of cancer stem cells (CSC) represents a major challenge for achieving effective treatment. miRNAs have emerged as critical regulators of gene expression, and recent studies have highlighted their role in regulating stemness and therapeutic resistance in CRC stem cells. This review highlights the mechanisms of CSC development, therapy resistance and the potential of miRNAs as therapeutic targets for CRC. It emphasizes the promise of miRNAs as a novel approach to CRC treatment and calls for further research to explore effective miRNA-based therapies and strategies for delivering miRNAs to CSCs in vivo.
Collapse
Affiliation(s)
- George Yiadom Osei
- Department of Biomedical Sciences, Advanced Medical & Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Malaysia
- Department of Medical Laboratory Sciences, University of Health & Allied Sciences, PMB 31, Ho, Ghana
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Selina Koomson
- Department of Medical Laboratory Sciences, University of Health & Allied Sciences, PMB 31, Ho, Ghana
| | - Solomon Beletaa
- Department of Medical Laboratory Sciences, University of Health & Allied Sciences, PMB 31, Ho, Ghana
| | - Muhammad Khairi Ahmad
- Department of Biomedical Sciences, Advanced Medical & Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Emmanuel Akomanin Asiamah
- Department of Medical Laboratory Sciences, University of Health & Allied Sciences, PMB 31, Ho, Ghana
- Discipline of Public Health Medicine, School of Nursing & Public Health, University of KwaZulu-Natal, Durban, 4001, South Africa
- Cancer & Infectious Diseases Epidemiology Research Unit (CIDERU), College of Health Sciences, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Cecilia Smith-Togobo
- Department of Medical Laboratory Sciences, University of Health & Allied Sciences, PMB 31, Ho, Ghana
| | - Siti Razila Abdul Razak
- Department of Biomedical Sciences, Advanced Medical & Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Malaysia
| |
Collapse
|
33
|
Nagae A, Miyoshi N, Fujino S, Horie M, Yachida S, Sasaki M, Sekido Y, Hata T, Hamabe A, Ogino T, Takahashi H, Uemura M, Yamamoto H, Doki Y, Eguchi H. Cancer Stem Cells Persist Despite Cellular Damage, Emergence of the Refractory Cell Population. Ann Surg Oncol 2023; 30:6913-6924. [PMID: 37523119 PMCID: PMC10506944 DOI: 10.1245/s10434-023-13849-x] [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: 04/27/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE Cancer stem cells (CSCs) are responsible for chemotherapy resistance and have unique properties that protect them from chemotherapy. Investigating CSCs may help to identify the population that is more resistant to treatments, leading to recurrence. We evaluated persisting CSCs, emerging after chemotherapy that cause tumor recurrence. METHODS Using human colorectal cancer organoids prepared from surgical specimens, we looked at changes in CSCs, the emergence and changes in the original population, which single-cell analysis identified. RESULTS With regards to changes in cancer stem cell markers, CD44 showed low levels after 5-fluorouracil administration. Once the CD44-ve population was sorted and cultured, the CD44+ve population gradually emerged, and the CD44-ve population decreased. Compared with the CD44-ve population of an organoid parent, the CD44-ve population proliferated after chemotherapeutic agent stimulation. The CD44-ve population was derived from the CD44+ve population before chemotherapeutic agents. In addition, when the CD44 variants were evaluated, the CD44v9 population remained. In single-cell analysis, we found that POU5F1 was highly expressed in the CD44low population. Velocity analysis showed that the CD44-ve population was induced after chemotherapy and expressed POU5F1. POU5F1-EGFP-Casp9 transfected organoids resulted in the appearance of a CD44-ve population after administration of a chemotherapeutic reagent. Both in vivo and in vitro, the dimerizer administration inhibited tumor growth significantly. CONCLUSIONS POU5F1 is involved in chemotherapy resistance in relation to stemness. For the treatment against refractory tumors, such as the recurrence after chemotherapy, the treatment should target the emerging specific population such as CD44 (or CD44v9) and proliferative cancer cells.
Collapse
Affiliation(s)
- Ayumi Nagae
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka, Japan
| | - Norikatsu Miyoshi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka, Japan.
| | - Shiki Fujino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka, Japan
| | - Masafumi Horie
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan
| | - Masaru Sasaki
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuki Sekido
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tsuyoshi Hata
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Hamabe
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
34
|
Izadpanah A, Mohammadkhani N, Masoudnia M, Ghasemzad M, Saeedian A, Mehdizadeh H, Poorebrahim M, Ebrahimi M. Update on immune-based therapy strategies targeting cancer stem cells. Cancer Med 2023; 12:18960-18980. [PMID: 37698048 PMCID: PMC10557910 DOI: 10.1002/cam4.6520] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
Accumulating data reveals that tumors possess a specialized subset of cancer cells named cancer stem cells (CSCs), responsible for metastasis and recurrence of malignancies, with various properties such as self-renewal, heterogenicity, and capacity for drug resistance. Some signaling pathways or processes like Notch, epithelial to mesenchymal transition (EMT), Hedgehog (Hh), and Wnt, as well as CSCs' surface markers such as CD44, CD123, CD133, and epithelial cell adhesion molecule (EpCAM) have pivotal roles in acquiring CSCs properties. Therefore, targeting CSC-related signaling pathways and surface markers might effectively eradicate tumors and pave the way for cancer survival. Since current treatments such as chemotherapy and radiation therapy cannot eradicate all of the CSCs and tumor relapse may happen following temporary recovery, improving novel and more efficient therapeutic options to combine with current treatments is required. Immunotherapy strategies are the new therapeutic modalities with promising results in targeting CSCs. Here, we review the targeting of CSCs by immunotherapy strategies such as dendritic cell (DC) vaccines, chimeric antigen receptors (CAR)-engineered immune cells, natural killer-cell (NK-cell) therapy, monoclonal antibodies (mAbs), checkpoint inhibitors, and the use of oncolytic viruses (OVs) in pre-clinical and clinical studies. This review will mainly focus on blood malignancies but also describe solid cancers.
Collapse
Affiliation(s)
- Amirhossein Izadpanah
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Niloufar Mohammadkhani
- Department of Clinical BiochemistrySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Mina Masoudnia
- Department of ImmunologySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Mahsa Ghasemzad
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Molecular Cell Biology‐Genetics, Faculty of Basic Sciences and Advanced Technologies in BiologyUniversity of Science and CultureTehranIran
| | - Arefeh Saeedian
- Radiation Oncology Research CenterCancer Research Institute, Tehran University of Medical SciencesTehranIran
- Department of Radiation OncologyCancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Hamid Mehdizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Mansour Poorebrahim
- Arnie Charbonneau Cancer Research Institute, University of CalgaryAlbertaCalgaryCanada
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of regenerative medicineCell Science research Center, Royan Institute for stem cell biology and technology, ACECRTehranIran
| |
Collapse
|
35
|
Saoudi González N, Salvà F, Ros J, Baraibar I, Rodríguez-Castells M, García A, Alcaráz A, Vega S, Bueno S, Tabernero J, Elez E. Unravelling the Complexity of Colorectal Cancer: Heterogeneity, Clonal Evolution, and Clinical Implications. Cancers (Basel) 2023; 15:4020. [PMID: 37627048 PMCID: PMC10452468 DOI: 10.3390/cancers15164020] [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: 07/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Colorectal cancer (CRC) is a global health concern and a leading cause of death worldwide. The disease's course and response to treatment are significantly influenced by its heterogeneity, both within a single lesion and between primary and metastatic sites. Biomarkers, such as mutations in KRAS, NRAS, and BRAF, provide valuable guidance for treatment decisions in patients with metastatic CRC. While high concordance exists between mutational status in primary and metastatic lesions, some heterogeneity may be present. Circulating tumor DNA (ctDNA) analysis has proven invaluable in identifying genetic heterogeneity and predicting prognosis in RAS-mutated metastatic CRC patients. Tumor heterogeneity can arise from genetic and non-genetic factors, affecting tumor development and response to therapy. To comprehend and address clonal evolution and intratumoral heterogeneity, comprehensive genomic studies employing techniques such as next-generation sequencing and computational analysis are essential. Liquid biopsy, notably through analysis of ctDNA, enables real-time clonal evolution and treatment response monitoring. However, challenges remain in standardizing procedures and accurately characterizing tumor subpopulations. Various models elucidate the origin of CRC heterogeneity, highlighting the intricate molecular pathways involved. This review focuses on intrapatient cancer heterogeneity and genetic clonal evolution in metastatic CRC, with an emphasis on clinical applications.
Collapse
Affiliation(s)
- Nadia Saoudi González
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Francesc Salvà
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Javier Ros
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Iosune Baraibar
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Marta Rodríguez-Castells
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Ariadna García
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
| | - Adriana Alcaráz
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Sharela Vega
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Sergio Bueno
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Josep Tabernero
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Elena Elez
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| |
Collapse
|
36
|
Wang Y, Yang Z, Zhu W, Chen Y, He X, Li J, Han Z, Yang Y, Liu W, Zhang K. Dihydroartemisinin inhibited stem cell-like properties and enhanced oxaliplatin sensitivity of colorectal cancer via AKT/mTOR signaling. Drug Dev Res 2023; 84:988-998. [PMID: 37132439 DOI: 10.1002/ddr.22067] [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: 02/16/2023] [Revised: 03/26/2023] [Accepted: 04/11/2023] [Indexed: 05/04/2023]
Abstract
Colorectal cancer (CRC) is a common tumor with high morbidity and mortality. The use of oxaliplatin (L-OHP) as a first-line treatment for CRC is limited due to chemoresistance. Growing evidence have revealed that the existence of cancer stem-like cells (CSLCs) is one of the important reasons for drug resistance and recurrence of cancers. Dihydroartemisinin (DHA), a derivative of artemisinin, has showed anticancer effects on a variety of malignancies, in addition to its antimalarial effects. However, the effect and mechanism of DHA on CSLCs and chemosensitivity in CRC cells remains unclear. In this study, we found that DHA inhibited cell viability in HCT116 and SW620 cells. Moreover, DHA decreased cell clonogenicity, and improved L-OHP sensitivity. Furthermore, DHA treatment attenuated tumor sphere formation, and the expressions of stem cell surface marker (CD133 and CD44) and stemness-associated transcription factor (Nanog, c-Myc, and OCT4). Mechanistically, the present findings showed that DHA inhibited of AKT/mTOR signaling pathway. The activation of AKT/mTOR signaling reversed DHA-decreased cell viability, clonogenicity, L-OHP resistance, tumor sphere, and expressions of stemness-associated protein in CRC. The inhibitory effect of DHA on tumorigenicity of CRC cells has also been demonstrated in BALB/c nude mice. In conclusion, this study revealed that DHA inhibited CSLCs properties in CRC via AKT/mTOR signaling, suggesting that DHA may be used as a potential therapeutic agent for CRC.
Collapse
Affiliation(s)
- Yujun Wang
- School of Pharmacy, Chengdu Medical College, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Zhirong Yang
- Pathology Department, Deyang People's Hospital, Deyang, China
| | - Wanglong Zhu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Yuzhuo Chen
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xingqiang He
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Jiaofeng Li
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Zhengyu Han
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Yuhan Yang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Wei Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Kun Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
37
|
Quarshie JT, Fosu K, Offei NA, Sobo AK, Quaye O, Aikins AR. Cryptolepine Suppresses Colorectal Cancer Cell Proliferation, Stemness, and Metastatic Processes by Inhibiting WNT/β-Catenin Signaling. Pharmaceuticals (Basel) 2023; 16:1026. [PMID: 37513937 PMCID: PMC10383422 DOI: 10.3390/ph16071026] [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: 05/31/2023] [Revised: 06/25/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent cancer and the second leading cause of cancer-related deaths globally. Evidence shows that over 90% of CRC cases are initiated by a deregulated Wingless Integrated Type-1 (WNT)/β-catenin signaling pathway. The WNT/β-catenin pathway also promotes CRC cell proliferation, stemness, and metastasis. Therefore, modulators of the WNT/β-catenin pathway may serve as promising regimens for CRC. This study investigated the effect of cryptolepine-a plant-derived compound-on the WNT/β-catenin pathway in CRC. Two CRC cell lines, COLO205 and DLD1, were treated with cryptolepine or XAV 939 (a WNT inhibitor) in the presence or absence of WNT3a (a WNT activator). Using a tetrazolium-based assay, cryptolepine was found to reduce cell viability in a dose- and time-dependent manner and was a more potent inhibitor of viability than XAV 939. RT-qPCR analyses showed that cryptolepine reverses WNT3a-induced expression of β-catenin, c-MYC, and WISP1, suggesting that cryptolepine inhibits WNT3a-mediated activation of WNT/β-catenin signaling. Cryptolepine also repressed WNT3a-induced OCT4 and CD133 expression and suppressed colony formation of the cells, indicating that cryptolepine inhibits the stemness of CRC cells. Additionally, cryptolepine inhibited WNT3a-induced epithelial-to-mesenchymal transition by reducing the expression of SNAI1 and TWIST1 genes. In a wound healing assay, cryptolepine was found to suppress cell migration under unstimulated and WNT3a-stimulated conditions. Moreover, cryptolepine downregulated WNT3a-induced expression of MMP2 and MMP9 genes, which are involved in cancer cell invasion. Altogether, cryptolepine suppresses CRC cell proliferation, stemness, and metastatic properties by inhibiting WNT3a-mediated activation of the WNT/β-catenin signaling pathway. These findings provide a rationale for considering cryptolepine as a potential WNT inhibitor in CRC.
Collapse
Affiliation(s)
- Jude Tetteh Quarshie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Kwadwo Fosu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Nicholas Awuku Offei
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Augustine Kojo Sobo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Anastasia Rosebud Aikins
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| |
Collapse
|
38
|
Cao T, Wang S, Qian L, Wu C, Huang T, Wang Y, Li Q, Wang J, Xia Y, Xu L, Wang L, Huang X. NPRA promotes fatty acid metabolism and proliferation of gastric cancer cells by binding to PPARα. Transl Oncol 2023; 35:101734. [PMID: 37418841 DOI: 10.1016/j.tranon.2023.101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/26/2023] [Accepted: 06/22/2023] [Indexed: 07/09/2023] Open
Abstract
Among cancers, gastric cancer (GC) ranks third globally in morbidity and mortality, particularly in East Asia. Natriuretic peptide receptor A (NPRA), a receptor for guanylate cyclase, plays important roles in regulating water and sodium balance. Recent studies have suggested that NPRA is involved in tumorigenesis, but its role in GC development remains unclear. Herein, we showed that the expression level of NPRA was positively correlated with gastric tumor size and clinical stage. Patients with high NPRA expression had a lower five-year survival rate than those with low expression, and NPRA was identified as an independent predictor of GC prognosis. NPRA knockdown suppressed GC cell proliferation, migration and invasion. NPRA overexpression enhanced cell malignant behavior. Immunohistochemistry of collected tumor samples showed that tumors with high NPRA expression had higher peroxisome proliferator-activated receptor α (PPARα) levels. In vivo and in vitro studies showed that NPRA promotes fatty acid oxidation and tumor cell metastasis. Co-IP showed that NPRA binds to PPARα and prevents PPARα degradation. PPARα upregulation under NPRA protection activates arnitine palmitoyl transferase 1B (CPT1B) to promote fatty acid oxidation. In this study, new mechanisms by which NPRA promotes the development of GC and new regulatory mechanisms of PPARα were identified.
Collapse
Affiliation(s)
- Tingting Cao
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Song Wang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Long Qian
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China; General Surgery Department, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, Anhui, China
| | - Chengwei Wu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Tao Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Ye Wang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Qian Li
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Jiawei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Yabin Xia
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Li Xu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, No.2, Zheshan West Road, Wuhu, Anhui 241001, China.
| |
Collapse
|
39
|
Suman S, Hota SK, Misra P, Sahu N, Sahu S. Immunohistochemical Expression of the Stem Cell Marker CD133 in Colorectal Carcinoma. Cureus 2023; 15:e41242. [PMID: 37529823 PMCID: PMC10387822 DOI: 10.7759/cureus.41242] [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: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Background Colorectal carcinoma (CRC) is the second-leading cause of cancer-related death. Despite the combined (surgery, chemotherapy, radiotherapy, and immunotherapy) modalities of treatment, the prognosis remains poor, mostly because of recurrence and distant metastasis. Cancer stem cells (CSC) are thought to be responsible for the development and spread of tumors. Hence, targeted therapy against these cells hopes to reduce the chance of recurrence and metastasis and improve the prognosis. Many immune markers have been identified to detect CSC in CRC. Here, we tried to assess the immunohistochemical expression of the stem cell marker CD133 in colorectal carcinoma and its correlation with various pathological parameters. Methodology A total of 51 cases of CRC were analyzed. Immunohistochemistry for CD133 was done after standardization in our laboratory. Expression status was decided based on the total score obtained by multiplying the intensity score by the percentage score. CD133 expression was correlated with the age and gender of the patient, tumor location, histological grade, extent of invasion, lymphovascular invasion (LVI), perineural invasion (PNI), and nodal status. Results High CD133 expression was seen in 21 (41.17%) cases. There was no significant association between CD133 expression and the pathological parameters except the tumor site. CD133 expression was significantly higher as we moved from the proximal colon to the rectum. Conclusions CD133 expression was significantly higher in the distal part of the large intestine as compared to the proximal part. But there was no linear correlation between CD133 expression and histological grade, extent of invasion, or nodal status.
Collapse
Affiliation(s)
- Sweta Suman
- Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | | | - Pranati Misra
- Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | - Nageswar Sahu
- Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| | - Subrat Sahu
- Surgery, Kalinga Institute of Medical Sciences, Bhubaneswar, Bhubaneswar, IND
| |
Collapse
|
40
|
Bartolomé RA, Casal JI. Proteomic profiling and network biology of colorectal cancer liver metastasis. Expert Rev Proteomics 2023; 20:357-370. [PMID: 37874121 DOI: 10.1080/14789450.2023.2275681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/23/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Tissue-based proteomic studies of colorectal cancer (CRC) metastasis have delivered fragmented results, with very few therapeutic targets and prognostic biomarkers moving beyond the discovery phase. This situation is likely due to the difficulties in obtaining and analyzing large numbers of patient-derived metastatic samples, the own heterogeneity of CRC, and technical limitations in proteomics discovery. As an alternative, metastatic CRC cell lines provide a flexible framework to investigate the underlying mechanisms and network biology of metastasis for target discovery. AREAS COVERED In this perspective, we comment on different in-depth proteomic studies of metastatic versus non-metastatic CRC cell lines. Identified metastasis-related proteins are introduced and discussed according to the spatial location in different cellular fractions, with special emphasis on membrane/adhesion proteins, secreted proteins, and nuclear factors, including miRNAs associated with liver metastasis. Moreover, we analyze the biological significance and potential therapeutic applications of the identified liver metastasis-related proteins. EXPERT OPINION The combination of protein discovery and functional analysis is the only way to accelerate the progress to clinical translation of the proteomic-derived findings in a relatively fast pace. Patient-derived organoids represent a promising alternative to patient tissues and cell lines, but further optimizations are still required for achieving solid and reproducible results.
Collapse
Affiliation(s)
- Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| |
Collapse
|
41
|
Razi S, Haghparast A, Chodari Khameneh S, Ebrahimi Sadrabadi A, Aziziyan F, Bakhtiyari M, Nabi-Afjadi M, Tarhriz V, Jalili A, Zalpoor H. The role of tumor microenvironment on cancer stem cell fate in solid tumors. Cell Commun Signal 2023; 21:143. [PMID: 37328876 PMCID: PMC10273768 DOI: 10.1186/s12964-023-01129-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 06/18/2023] Open
Abstract
In the last few decades, the role of cancer stem cells in initiating tumors, metastasis, invasion, and resistance to therapies has been recognized as a potential target for tumor therapy. Understanding the mechanisms by which CSCs contribute to cancer progression can help to provide novel therapeutic approaches against solid tumors. In this line, the effects of mechanical forces on CSCs such as epithelial-mesenchymal transition, cellular plasticity, etc., the metabolism pathways of CSCs, players of the tumor microenvironment, and their influence on the regulating of CSCs can lead to cancer progression. This review focused on some of these mechanisms of CSCs, paving the way for a better understanding of their regulatory mechanisms and developing platforms for targeted therapies. While progress has been made in research, more studies will be required in the future to explore more aspects of how CSCs contribute to cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Sara Razi
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
- Cytotech and Bioinformatics Research Group, Tehran, Iran
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahideh Tarhriz
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, P.O. Box 5163639888, Tabriz, Iran.
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran.
- Parvaz Research Ideas Supporter Institute, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
42
|
Luo Q, Liu P, Yu P, Qin T. Cancer Stem Cells are Actually Stem Cells with Disordered Differentiation: the Monophyletic Origin of Cancer. Stem Cell Rev Rep 2023; 19:827-838. [PMID: 36648606 PMCID: PMC10185654 DOI: 10.1007/s12015-023-10508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Cancer stem cells (CSCs) play an important role in cancer development. Based on advancements in CSC research, we propose a monophyletic model of cancer. This model is based on the idea that CSCs are stem cells with disordered differentiation whose original purpose was to repair damaged tissues. Inflammatory responses and damage repair signals are crucial for the creation and maintenance of CSCs. Normal quiescent stem cells are activated by environmental stimulation, such as an inflammatory response, and undergo cell division and differentiation. In the initial stage of cancer development, stem cell differentiation leads to heteromorphism due to the accumulation of gene mutations, resulting in the development of metaplasia or precancerosis. In the second stage, accumulated mutations induce poor differentiation and lead to cancer development. The monophyletic model illustrates the evolution, biological behavior, and hallmarks of CSCs, proposes a concise understanding of the origin of cancer, and may encourage a novel therapeutic approach.
Collapse
Affiliation(s)
- Qiankun Luo
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pan Liu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pengfei Yu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Tao Qin
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China.
| |
Collapse
|
43
|
Saha N, Baek DS, Mendoza RP, Robev D, Xu Y, Goldgur Y, De La Cruz MJ, de Stanchina E, Janes PW, Xu K, Dimitrov DS, Nikolov DB. Fully human monoclonal antibody targeting activated ADAM10 on colorectal cancer cells. Biomed Pharmacother 2023; 161:114494. [PMID: 36917886 PMCID: PMC10499537 DOI: 10.1016/j.biopha.2023.114494] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Metastasis and chemoresistance in colorectal cancer are mediated by certain poorly differentiated cancer cells, known as cancer stem cells, that are maintained by Notch downstream signaling initiated upon Notch cleavage by the metalloprotease ADAM10. It has been shown that ADAM10 overexpression correlates with aberrant signaling from Notch, erbBs, and other receptors, as well as a more aggressive metastatic phenotype, in a range of cancers including colon, gastric, prostate, breast, ovarian, uterine, and leukemia. ADAM10 inhibition, therefore, stands out as an important and new approach to deter the progression of advanced CRC. For targeting the ADAM10 substrate-binding region, which is located outside of the catalytic domain of the protease, we generated a human anti-ADAM10 monoclonal antibody named 1H5. Structural and functional characterization of 1H5 reveals that it binds to the substrate-binding cysteine-rich domain and recognizes an activated ADAM10 conformation present on tumor cells. The mAb inhibits Notch cleavage and proliferation of colon cancer cell lines in vitro and in mouse models. Consistent with its binding to activated ADAM10, the mAb augments the catalytic activity of ADAM10 towards small peptide substrates in vitro. Most importantly, in a mouse model of colon cancer, when administered in combination with the therapeutic agent Irinotecan, 1H5 causes highly effective tumor growth inhibition without any discernible toxicity effects. Our singular approach to target the ADAM10 substrate-binding region with therapeutic antibodies could overcome the shortcomings of previous intervention strategies of targeting the protease active site with small molecule inhibitors that exhibit musculoskeletal toxicity.
Collapse
Affiliation(s)
- Nayanendu Saha
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Du-San Baek
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Rachelle P Mendoza
- Department of Pathology, University of Chicago, Chicago, IL 60637, United States
| | - Dorothea Robev
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yan Xu
- Department of Veterinary Biosciences, Ohio State University, Columbus, OH 43210, United States
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - M Jason De La Cruz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Elisa de Stanchina
- Antitumor Assessment Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Peter W Janes
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Victoria 3084, Australia
| | - Kai Xu
- Department of Veterinary Biosciences, Ohio State University, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH 43210, United States
| | - Dimiter S Dimitrov
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Dimitar B Nikolov
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
44
|
Lee E, Cheung J, Bialkowska AB. Krüppel-like Factors 4 and 5 in Colorectal Tumorigenesis. Cancers (Basel) 2023; 15:cancers15092430. [PMID: 37173904 PMCID: PMC10177156 DOI: 10.3390/cancers15092430] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Krüppel-like factors (KLFs) are transcription factors regulating various biological processes such as proliferation, differentiation, migration, invasion, and homeostasis. Importantly, they participate in disease development and progression. KLFs are expressed in multiple tissues, and their role is tissue- and context-dependent. KLF4 and KLF5 are two fascinating members of this family that regulate crucial stages of cellular identity from embryogenesis through differentiation and, finally, during tumorigenesis. They maintain homeostasis of various tissues and regulate inflammation, response to injury, regeneration, and development and progression of multiple cancers such as colorectal, breast, ovarian, pancreatic, lung, and prostate, to name a few. Recent studies broaden our understanding of their function and demonstrate their opposing roles in regulating gene expression, cellular function, and tumorigenesis. This review will focus on the roles KLF4 and KLF5 play in colorectal cancer. Understanding the context-dependent functions of KLF4 and KLF5 and the mechanisms through which they exert their effects will be extremely helpful in developing targeted cancer therapy.
Collapse
Affiliation(s)
- Esther Lee
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jacky Cheung
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
45
|
Babazadeh M, Zamani M, Mehrbod P, Mokarram P. Stemness targeting of colorectal cell lines mediated by BAMLET and 5-Flourouracil. Biochem Biophys Res Commun 2023; 664:136-141. [PMID: 37167706 DOI: 10.1016/j.bbrc.2023.04.010] [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: 03/16/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 05/13/2023]
Abstract
PURPOSE Stemness is the potential for self-renewal and repopulation causing the relapse, progression, and drug resistance of colorectal cancer. We investigated the effects of bovine alpha-lactalbumin made lethal to tumor cells and 5-Flourouracil consisting of bovine α-lactalbumin protein and oleic acid, on colorectal cancer cells on stemness. METHODS The quantitative real-time polymerase chain reaction assessed the expression levels of stemness-related genes (c-myc, Lgr5, OCT4). Expression of stemness-related surface markers (CD44 and CD24) was also measured by the flow cytometry technique following the treatments. RESULTS Our results indicated decreased expression levels of C-Myc, Lgr5, oct4 as the stemness-related genes (P < 0.0001), and reduced population of CD44+ as the stemness-related cell surface marker upon treatment with BAMLET and 5-Flourouracil. BAMLET inhibited the stemness more effectively than 5-Flourouracil (P < 0.0001). CONCLUSION Based on the result, inhibition of the Stemness related-genes (C-Myc, Lgr5, Oct4) and the surface markers (CD 24+ and CD44+) is a promising therapeutic approach using BAMLET.
Collapse
Affiliation(s)
- Marziyeh Babazadeh
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran, Iran
| | - Pooneh Mokarram
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
46
|
Jones J, Shi Q, Nath RR, Brito IL. Keystone pathobionts associated with colorectal cancer promote oncogenic reprograming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535410. [PMID: 37066368 PMCID: PMC10103987 DOI: 10.1101/2023.04.03.535410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Fusobacterium nucleatum (Fn) and enterotoxigenic Bacteroides fragilis (ETBF) are two pathobionts consistently enriched in the gut microbiomes of patients with colorectal cancer (CRC) compared to healthy counterparts and frequently observed for their direct association within tumors. Although several molecular mechanisms have been identified that directly link these organisms to features of CRC in specific cell types, their specific effects on the epithelium and local immune compartment are not well-understood. To fill this gap, we leveraged single-cell RNA sequencing (scRNA-seq) on wildtype mice and mouse model of CRC. We find that Fn and ETBF exacerbate cancer-like transcriptional phenotypes in transit-amplifying and mature enterocytes in a mouse model of CRC. We also observed increased T cells in the pathobiont-exposed mice, but these pathobiont-specific differences observed in wildtype mice were abrogated in the mouse model of CRC. Although there are similarities in the responses provoked by each organism, we find pathobiont-specific effects in Myc-signaling and fatty acid metabolism. These findings support a role for Fn and ETBF in potentiating tumorigenesis via the induction of a cancer stem cell-like transit-amplifying and enterocyte population and the disruption of CTL cytotoxic function.
Collapse
Affiliation(s)
- Josh Jones
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Qiaojuan Shi
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Rahul R. Nath
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| | - Ilana L. Brito
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
47
|
Stage IV Colorectal Cancer Management and Treatment. J Clin Med 2023; 12:jcm12052072. [PMID: 36902858 PMCID: PMC10004676 DOI: 10.3390/jcm12052072] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
(1) Background: Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related mortality worldwide. Up to 50% of patients with CRC develop metastatic CRC (mCRC). Surgical and systemic therapy advances can now offer significant survival advantages. Understanding the evolving treatment options is essential for decreasing mCRC mortality. We aim to summarize current evidence and guidelines regarding the management of mCRC to provide utility when making a treatment plan for the heterogenous spectrum of mCRC. (2) Methods: A comprehensive literature search of PubMed and current guidelines written by major cancer and surgical societies were reviewed. The references of the included studies were screened to identify additional studies that were incorporated as appropriate. (3) Results: The standard of care for mCRC primarily consists of surgical resection and systemic therapy. Complete resection of liver, lung, and peritoneal metastases is associated with better disease control and survival. Systemic therapy now includes chemotherapy, targeted therapy, and immunotherapy options that can be tailored by molecular profiling. Differences between colon and rectal metastasis management exist between major guidelines. (4) Conclusions: With the advances in surgical and systemic therapy, as well as a better understanding of tumor biology and the importance of molecular profiling, more patients can anticipate prolonged survival. We provide a summary of available evidence for the management of mCRC, highlighting the similarities and presenting the difference in available literature. Ultimately, a multidisciplinary evaluation of patients with mCRC is crucial to selecting the appropriate pathway.
Collapse
|
48
|
Karami Fath M, Garousi S, Mottahedi M, Ghasemzadeh N, Salmani K, Olfati F, Beit Saeed M, Sotoudeh S, Barati G. The role of hypoxia-inducible factors in breast cancer stem cell specification. Pathol Res Pract 2023; 243:154349. [PMID: 36791562 DOI: 10.1016/j.prp.2023.154349] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
Breast tumor is heterogeneous cancer with high morbidity and mortality rates, particularly in developing countries. Despite new efforts to reduce the breast cancer implications, the number of newly diagnosed cases is increasing worldwide. It is believed that cancer stem cells (CSCs) are responsible for the implication of cancers including breast cancer. Although CSCs compose a small population in tumor bulks, they play a crucial role in tumor initiation, progression, metastasis, and chemotherapeutic resistance. These events are mediated by the hypoxia-inducible factor (HIF) pathway which regulates the transcription of genes involved in CSC maintenance and tumorigenesis. In this review, we discussed the mechanisms by which hypoxia- or chemotherapy-induced HIFs promote breast CSC specification.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Setareh Garousi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Mottahedi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Kiana Salmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Olfati
- Department of Reproductive Health, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | - Miad Beit Saeed
- Faculty of Nursing and Midwifery, Abadan Islamic Azad University, Abadan, Iran
| | - Sina Sotoudeh
- Faculty of Nursing and Midwifery, Guilan University of Medical Sciences, Guilan, Iran
| | | |
Collapse
|
49
|
D'Antonio L, Fieni C, Ciummo SL, Vespa S, Lotti L, Sorrentino C, Di Carlo E. Inactivation of interleukin-30 in colon cancer stem cells via CRISPR/Cas9 genome editing inhibits their oncogenicity and improves host survival. J Immunother Cancer 2023; 11:jitc-2022-006056. [PMID: 36927528 PMCID: PMC10030651 DOI: 10.1136/jitc-2022-006056] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Progression of colorectal cancer (CRC), a leading cause of cancer-related death worldwide, is driven by colorectal cancer stem cells (CR-CSCs), which are regulated by endogenous and microenvironmental signals. Interleukin (IL)-30 has proven to be crucial for CSC viability and tumor progression. Whether it is involved in CRC tumorigenesis and impacts clinical behavior is unknown. METHODS IL30 production and functions, in stem and non-stem CRC cells, were determined by western blot, immunoelectron microscopy, flow cytometry, cell viability and sphere formation assays. CRISPR/Cas9-mediated deletion of the IL30 gene, RNA-Seq and implantation of IL30 gene transfected or deleted CR-CSCs in NSG mice allowed to investigate IL30's role in CRC oncogenesis. Bioinformatics and immunopathology of CRC samples highlighted the clinical implications. RESULTS We demonstrated that both CR-CSCs and CRC cells express membrane-anchored IL30 that regulates their self-renewal, via WNT5A and RAB33A, and/or proliferation and migration, primarily by upregulating CXCR4 via STAT3, which are suppressed by IL30 gene deletion, along with WNT and RAS pathways. Deletion of IL30 gene downregulates the expression of proteases, such as MMP2 and MMP13, chemokine receptors, mostly CCR7, CCR3 and CXCR4, and growth and inflammatory mediators, including ANGPT2, CXCL10, EPO, IGF1 and EGF. These factors contribute to IL30-driven CR-CSC and CRC cell expansion, which is abrogated by their selective blockade. IL30 gene deleted CR-CSCs displayed reduced tumorigenicity and gave rise to slow-growing and low metastatic tumors in 80% of mice, which survived much longer than controls. Bioinformatics and CIBERSORTx of the 'Colorectal Adenocarcinoma TCGA Nature 2012' collection, and morphometric assessment of IL30 expression in clinical CRC samples revealed that the lack of IL30 in CRC and infiltrating leucocytes correlates with prolonged overall survival. CONCLUSIONS IL30 is a new CRC driver, since its inactivation, which disables oncogenic pathways and multiple autocrine loops, inhibits CR-CSC tumorigenicity and metastatic ability. The development of CRISPR/Cas9-mediated targeting of IL30 could improve the current therapeutic landscape of CRC.
Collapse
Affiliation(s)
- Luigi D'Antonio
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Cristiano Fieni
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Stefania Livia Ciummo
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Simone Vespa
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Lavinia Lotti
- Department of Experimental Medicine, University of Rome La Sapienza, Rome, Italy
| | - Carlo Sorrentino
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| | - Emma Di Carlo
- Dipartimento di Medicina e Scienze dell'Invecchiamento, Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Università degli Studi "G. d'Annunzio" di Chieti-Pescara, Chieti, Italy
| |
Collapse
|
50
|
Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 2023; 22:38. [PMID: 36810098 PMCID: PMC9942413 DOI: 10.1186/s12943-023-01748-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Ongoing research has revealed that the existence of cancer stem cells (CSCs) is one of the biggest obstacles in the current cancer therapy. CSCs make an influential function in tumor progression, recurrence and chemoresistance due to their typical stemness characteristics. CSCs are preferentially distributed in niches, and those niche sites exhibit characteristics typical of the tumor microenvironment (TME). The complex interactions between CSCs and TME illustrate these synergistic effects. The phenotypic heterogeneity within CSCs and the spatial interactions with the surrounding tumor microenvironment led to increased therapeutic challenges. CSCs interact with immune cells to protect themselves against immune clearance by exploiting the immunosuppressive function of multiple immune checkpoint molecules. CSCs also can protect themselves against immune surveillance by excreting extracellular vesicles (EVs), growth factors, metabolites and cytokines into the TME, thereby modulating the composition of the TME. Therefore, these interactions are also being considered for the therapeutic development of anti-tumor agents. We discuss here the immune molecular mechanisms of CSCs and comprehensively review the interplay between CSCs and the immune system. Thus, studies on this topic seem to provide novel ideas for reinvigorating therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Bo Wu
- grid.459742.90000 0004 1798 5889Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Xiang Shi
- grid.459742.90000 0004 1798 5889Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Meixi Jiang
- grid.412644.10000 0004 5909 0696Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032 China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|