1
|
Glymenaki M, Curio S, Shrestha S, Zhong Q, Rushton L, Barry R, El-Bahrawy M, Marchesi JR, Wang Y, Gooderham NJ, Guerra N, Li JV. Roux-en-Y gastric bypass-associated fecal tyramine promotes colon cancer risk via increased DNA damage, cell proliferation, and inflammation. MICROBIOME 2025; 13:60. [PMID: 40022152 PMCID: PMC11869571 DOI: 10.1186/s40168-025-02049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Fecal abundances of Enterobacteriaceae and Enterococcaceae are elevated in patients following Roux-en-Y gastric bypass (RYGB) surgery. Concurrently, fecal concentrations of tyramine, derived from gut bacterial metabolism of tyrosine and/or food, increased post-RYGB. Furthermore, emerging evidence suggests that RYGB is associated with increased colorectal cancer (CRC) risk. However, the causal link between RYGB-associated microbial metabolites and CRC risk remains unclear. Hence, this study investigated the tyrosine metabolism of Enterobacteriaceae and Enterococcaceae strains isolated from patients post-RYGB and explored the causal effects of tyramine on the CRC risk and tumorigenesis using both human colonic cancer cell line (HCT 116) and wild-type and ApcMin/+ mice. RESULTS We isolated 31 bacterial isolates belonging to Enterobacteriaceae and Enterococcaceae families from the feces of patients with RYGB surgery. By culturing the isolates in tyrosine-supplemented medium, we found that Citrobacter produced phenol as a main product of tyrosine, whereas Enterobacter and Klebsiella produced 4-hydroxyphenylacetate, Escherichia produced 4-hydroxyphenyllactate and 4-hydroxyphenylpyruvate, and Enterococcus and two Klebsiella isolates produced tyramine. These observations suggested the gut bacterial contribution to increased fecal concentrations of tyramine post-RYGB. We subsequently evaluated the impact of tyramine on CRC risk and development. Tyramine induced necrosis and promoted cell proliferation and DNA damage of HCT 116 cells. Daily oral administration of tyramine for 49 days to wild-type mice resulted in visible adenomas in 5 out of 12 mice, accompanied by significantly enhanced DNA damage (γH2AX +) and an increased trend of cell proliferation (Ki67 +) in the ileum, along with an upregulated expression of the cell division cycle gene (Cdc34b) in the colon. To evaluate the impact of tyramine on intestinal tumor growth, we treated ApcMin/+ mice with the same doses of tyramine and duration. These mice showed larger colonic tumor size and increased intestinal cell proliferation and inflammation (e.g., increased mRNA expression of IL-17A and higher number of Ly6G + neutrophils) compared to water-treated ApcMin/+ control mice. CONCLUSIONS Our results collectively suggested that RYGB-associated fecal bacteria could contribute to tyramine production and tyramine increased CRC risk by increasing DNA damage, cell proliferation, and pro-inflammatory responses of the gut. Monitoring and modulating tyramine concentrations in high-risk individuals could aid CRC prognosis and management. Video Abstract.
Collapse
Affiliation(s)
- Maria Glymenaki
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sophie Curio
- Department of Life Sciences, Imperial College London, London, UK
- The University of Queensland Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Smeeta Shrestha
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Qi Zhong
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Laura Rushton
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department for Environment Food and Rural Affairs, London, UK
| | - Rachael Barry
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Mona El-Bahrawy
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Nigel J Gooderham
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, UK
| | - Jia V Li
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
2
|
Ambite I, Wan MLY, Tran HT, Nazari A, Chaudhuri A, Krintel C, Gomes I, Sabari S, Ahmadi S, Carneiro ANBM, Ishac R, Haq F, Svanborg C. Multitarget mechanism of MYC inhibition by the bacterial lon protease in disease. Sci Rep 2025; 15:6778. [PMID: 40000737 PMCID: PMC11861601 DOI: 10.1038/s41598-025-88093-2] [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: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Identifying specific inhibitors of the MYC oncogene has been challenging, due to off target effects associated with MYC inhibition. This study investigated how the recombinant Escherichia coli Lon protease (rLon), which targets MYC in human cells, inhibits MYC over-activation in models of infection and cancer. In silico predictions identified specific peptide domains of bacterial Lon that target MYC and the affinity of these peptides for MYC was investigated by surface plasmon resonance. The N-terminal domain of rLon was shown to interact with the C-terminal, leucine zipper domain of MYC and MAX and to prevent MYC/MAX dimerization. Furthermore, rLon targeted and degraded c-MYC in vitro and in cellular models, through the peptidase domain. In a model of kidney infection, rLon treatment prevented, c-MYC, N-MYC and L-MYC over-expression, MYC-dependent gene expression, specifically renal toxicity genes and pathology, suggesting that rLon recognizes and corrects MYC dysregulation in this disease. The findings describe a multitarget mechanism of MYC inhibition by rLon, and the combined effects achieved by the Lon domains, targeting different MYC epitopes and MYC-dependent functions, with no evidence of toxicity or detrimental effects on homeostatic MYC expression.
Collapse
Affiliation(s)
- Ines Ambite
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Murphy Lam Yim Wan
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Hien Thi Tran
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Atefeh Nazari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Arunima Chaudhuri
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Christian Krintel
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Inês Gomes
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Samudra Sabari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Shahram Ahmadi
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - António N B M Carneiro
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Rita Ishac
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Farhan Haq
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Catharina Svanborg
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden.
| |
Collapse
|
3
|
Chieppa M, De Santis S, Verna G. Winnie Mice: A Chronic and Progressive Model of Ulcerative Colitis. Inflamm Bowel Dis 2025:izaf006. [PMID: 39912845 DOI: 10.1093/ibd/izaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Recent trends show a continuous worldwide rise in the incidence of ulcerative colitis (UC), leading to increased interest in its etiology and pathogenesis, which is currently unknown. To gain a better mechanistic understanding of this disease, many mouse models have been developed over the last several years, with variations of dextran sodium sulfate administration representing the most widely employed. The Winnie mouse strain was created through elicited random mutations in Muc2, resulting in a progressive, chronic intestinal inflammation localized to the colon that worsens over time. Moreover, Winnie mice display immunologic and microbiota features that are similar to those that can be found in UC patients. Phenotypically, the presence, albeit rare, of rectal prolapse and other complications impacting quality of life can be observed in Winnie mice, as well as extraintestinal manifestations that are often associated with UC. While Winnie mice are currently less studied compared to other more established models of colitis, much has been discovered in the initial years of its use as a UC-like model. In summary, the use of Winnie mice adds to the growing armamentarium that is required to develop precision-based medicine for its future application in treating complex multifactorial diseases, such as UC.
Collapse
Affiliation(s)
- Marcello Chieppa
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy
| | - Stefania De Santis
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giulio Verna
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Luo LZ, Kim JH, Herrera I, Wu S, Wu X, Park SS, Cho J, Cope L, Xian L, West BE, Calderon-Espinosa J, Kim J, Thompson Z, Maloo I, Larman T, Reddy KL, Feng Y, Fearon ER, Sears CL, Resar L. HMGA1 acts as an epigenetic gatekeeper of ASCL2 and Wnt signaling during colon tumorigenesis. J Clin Invest 2025; 135:e184442. [PMID: 39895630 PMCID: PMC11785931 DOI: 10.1172/jci184442] [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: 07/11/2024] [Accepted: 11/27/2024] [Indexed: 02/04/2025] Open
Abstract
Mutated tumor cells undergo changes in chromatin accessibility and gene expression, resulting in aberrant proliferation and differentiation, although how this occurs is unclear. HMGA1 chromatin regulators are abundant in stem cells and oncogenic in diverse tissues; however, their role in colon tumorigenesis is only beginning to emerge. Here, we uncover a previously unknown epigenetic program whereby HMGA1 amplifies Wnt signaling during colon tumorigenesis driven by inflammatory microbiota and/or Adenomatous polyposis coli (Apc) inactivation. Mechanistically, HMGA1 "opens" chromatin to upregulate the stem cell regulator, Ascl2, and downstream Wnt effectors, promoting stem and Paneth-like cell states while depleting differentiated enterocytes. Loss of just one Hmga1 allele within colon epithelium restrains tumorigenesis and Wnt signaling driven by mutant Apc and inflammatory microbiota. However, HMGA1 deficiency has minimal effects in colon epithelium under homeostatic conditions. In human colon cancer cells, HMGA1 directly induces ASCL2 by recruiting activating histone marks. Silencing HMGA1 disrupts oncogenic properties, whereas reexpression of ASCL2 partially rescues these phenotypes. Further, HMGA1 and ASCL2 are coexpressed and upregulated in human colorectal cancer. Together, our results establish HMGA1 as an epigenetic gatekeeper of Wnt signals and cell state under conditions of APC inactivation, illuminating HMGA1 as a potential therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Li Z. Luo
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jung-Hyun Kim
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Iliana Herrera
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shaoguang Wu
- Division of Infectious Diseases, Department of Medicine
| | - Xinqun Wu
- Division of Infectious Diseases, Department of Medicine
| | - Seong-Sik Park
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Juyoung Cho
- Research Institute, National Cancer Center, Goyang-si, Gyeonggido, Republic of Korea
| | - Leslie Cope
- Sidney Kimmel Comprehensive Cancer Center, Division of Biostatistics
| | - Lingling Xian
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bailey E. West
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pathobiology Graduate Program, Department of Pathology, and
| | - Julian Calderon-Espinosa
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Human Genetics Graduate Program, Department of Genetics and Molecular Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph Kim
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zanshé Thompson
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Isha Maloo
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biochemistry and Molecular Biology Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Karen L. Reddy
- Department of Biological Chemistry, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ying Feng
- Department of Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric R. Fearon
- Department of Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cynthia L. Sears
- Division of Infectious Diseases, Department of Medicine
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and
- Molecular Immunology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda Resar
- Division of Hematology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Comprehensive Cancer Center, Division of Biostatistics
- Pathobiology Graduate Program, Department of Pathology, and
- Human Genetics Graduate Program, Department of Genetics and Molecular Medicine, the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biochemistry and Molecular Biology Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Pathology and
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and
| |
Collapse
|
5
|
Wang Y, Ybarra M, Wang Z. HMGA1 is a crucial mediator of colon tumorigenesis driven by the loss of APC. J Clin Invest 2025; 135:e187442. [PMID: 39895631 PMCID: PMC11785912 DOI: 10.1172/jci187442] [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] [Indexed: 02/04/2025] Open
Abstract
Colorectal cancer is the second leading cause of cancer death in the United States. The adenomatous polyposis coli (APC) pathway plays a critical role in colorectal tumorigenesis, but the mechanism is not fully understood. In this issue of the JCI, Luo and colleagues used genetically engineered mouse models to show that high mobility group A (HMGA1) is a critical mediator in the development of colon tumors driven by the loss of the Apc gene. HMGA1 activated the transcription of Achaete-Scute Family BHLH Transcription Factor 2 (ASCL2), which regulated intestinal stemness and promoted colon tumorigenesis.
Collapse
Affiliation(s)
- Yuxiang Wang
- Department of Genetics and Genome Sciences and
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mikayla Ybarra
- Department of Genetics and Genome Sciences and
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences and
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Gaynor L, Singh H, Tie G, Badarinath K, Madha S, Mancini A, Bhattacharya S, Hoshino M, de Sauvage FJ, Murata K, Jadhav U, Shivdasani RA. Crypt density and recruited enhancers underlie intestinal tumour initiation. Nature 2025:10.1038/s41586-024-08573-9. [PMID: 39778708 DOI: 10.1038/s41586-024-08573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Oncogenic mutations that drive colorectal cancer can be present in healthy intestines for long periods without overt consequence1,2. Mutation of Apc, the most common initiating event in conventional adenomas3, activates Wnt signalling, thus conferring fitness on mutant intestinal stem cells (ISCs)4,5. Apc mutations may occur in ISCs that arise by routine self-renewal or by dedifferentiation of their progeny. Although ISCs of these different origins are fundamentally similar6,7, it is unclear whether both generate tumours equally well in uninjured intestines. It is also unknown whether cis-regulatory elements are substantively modulated upon Wnt hyperactivation or as a feature of subsequent tumours. Here we show in two mouse models that adenomas are not an obligatory outcome of Apc deletion in either ISC source, but require proximity of mutant intestinal crypts. Reduced crypt density abrogates, and aggregation of mutant colonic crypts augments, adenoma formation. Moreover, adenoma-resident ISCs open chromatin at thousands of enhancers that are inaccessible in Apc-null ISCs that are not associated with adenomas. These cis elements explain adenoma-selective gene activity and persist, with little further expansion of the repertoire, as other oncogenic mutations accumulate. Thus, cooperativity between neighbouring mutant crypts and new accessibility at specific enhancers are key steps early in intestinal tumorigenesis.
Collapse
Affiliation(s)
- Liam Gaynor
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Harshabad Singh
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Krithika Badarinath
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew Mancini
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Swarnabh Bhattacharya
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | - Kazutaka Murata
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Unmesh Jadhav
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
7
|
Husain K, Coppola D, Yang CS, Malafa MP. Effect of vitamin E δ-tocotrienol and aspirin on Wnt signaling in human colon cancer stem cells and in adenoma development in APCmin/+ mice. Carcinogenesis 2024; 45:881-892. [PMID: 38877828 DOI: 10.1093/carcin/bgae041] [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: 12/14/2023] [Revised: 04/17/2024] [Accepted: 06/14/2024] [Indexed: 06/16/2024] Open
Abstract
In this study, we evaluated the effects of vitamin E δ-tocotrienol (DT3) and aspirin on Wnt signaling in human colon cancer stem cells (CCSCs) and in the prevention of adenoma formation in APCmin/+ mice. We found that knockdown of the adenomatous polyposis coli (APC) gene led to subsequent activation of Wnt signaling in colon epithelial cells (NCM460-APCsiRNA) and induction of β-catenin and its downstream target proteins c-MYC, cyclin D1, and survivin. When aspirin and DT3 were combined, cell growth and survival were inhibited and apoptosis was induced in colon epithelial cells and CCSCs. However, DT3 and/or aspirin had little or no effect on the control of normal colon epithelial cells (NCM460-NCsiRNA). The induction of apoptosis was directly related to the activation of caspase 8 and cleavage of BH3-interacting-domain (BID) to truncated BID. In addition, DT3- and/or aspirin-induced apoptosis was associated with cleaved Poly (ADP-ribose) polymerase (PARP), elevated levels of cytosolic cytochrome c and BAX, and depletion of antiapoptotic protein BCl-2 in CCSCs. The combination of aspirin and DT3 inhibited the self-renewal capacity, Wnt/β-catenin receptor activity, and expression of β-catenin and its downstream targets c-MYC, cyclin D1, and survivin in CCSCs. We also found that treatment with DT3 alone or combined with aspirin significantly inhibited intestinal adenoma formation and Wnt/β-catenin signaling and induced apoptosis, compared with vehicle, in APCmin/+ mice. Our study demonstrated a rationale for further investigation of the combination of DT3 and aspirin for colorectal cancer prevention and therapy.
Collapse
Affiliation(s)
- Kazim Husain
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, Florida 33612, United States
| | - Domenico Coppola
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, Florida 33612, United States
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd, Piscataway, NJ 08854, United States
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, Florida 33612, United States
| |
Collapse
|
8
|
Wu L, Bai R, Zhang Y, Chen H, Wu J, Chen Z, Wang H, Zhao L. METTL3-VISTA axis-based combination immunotherapy for APC truncation colorectal cancer. J Immunother Cancer 2024; 12:e009865. [PMID: 39653551 PMCID: PMC11629097 DOI: 10.1136/jitc-2024-009865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE Although immune checkpoint blockade (ICB) therapy represents a bright spot in antitumor immunotherapy, its clinical benefits in colorectal cancer (CRC) are limited. Therefore, a new target for mediating CRC immunosuppression is urgently needed. Adenomatous polyposis coli (APC) mutations have been reported as early-stage characteristic events in CRC, but the role of truncated APC in the CRC immune microenvironment remains unclear and its clinical significance has yet to be explored. DESIGN Adenocarcinoma formation in the colon of the APCMin/+ mouse model, which displays features associated with the translation of truncated APC proteins, was induced by azoxymethane/dextran sodium sulfate. Multiplexed immunohistochemical consecutive staining on single slides and flow cytometry were used to explore the activation of immune cells and the expression of the immune checkpoint V-domain immunoglobulin suppressor of T-cell activation (VISTA) in the CRC tissues of APCWT and APCMin/+ mice. The construction of truncated APC vectors and an initial subserosal graft tumor mouse model was employed to mimic the tumor microenvironment (TME) during APC mutation. Methylated RNA immunoprecipitation-quantitative PCR assays were performed to investigate the N6-methyladenosine (m6A)-dependent transcriptional regulation of hypoxia-inducible factor-1 alpha (HIF1α) by methyltransferase-like protein 3 (METTL3). Mettl3fl/fl vil1-cre+/- mice were used to demonstrate that targeting METTL3 is a mediator that mitigates the deleterious effects of the APC978∆-HIF1α axis on antitumor immunity. A chimeric VISTA humanized mouse model was used to evaluate the drug efficacy of the VISTA-targeted compound onvatilimab. RESULTS We showed that APC978∆, a truncated APC protein, mediated overexpression of METTL3, resulting in m6A methylation of HIF1α messenger RNA and high expression of HIF1α. Furthermore, HIF1α promotes the migration of myeloid-derived suppressor cells to the TME by binding to the promoters of MCP-1 and MIF. In addition, HIF1α enhances the expression of the immune checkpoint VISTA on CRC cells, weakening tumor immune monitoring. CONCLUSIONS We elucidate that an underappreciated function of truncated APC in CRC is its ability to drive an immunosuppressive program that boosts tumor progression. Our work could provide a new perspective for the clinical application of immunotherapy in patients with CRC resistant to ICB therapy.
Collapse
Affiliation(s)
- Ling Wu
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Bai
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianghua Wu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zetao Chen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Medical Oncology, Guangzhou Medical University Affiliated Cancer Hospital, Guangzhou, Guangdong, China
| | - Liang Zhao
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Brandner S. Rodent models of tumours of the central nervous system. Mol Oncol 2024; 18:2842-2870. [PMID: 39324445 PMCID: PMC11619804 DOI: 10.1002/1878-0261.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 07/03/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
Modelling of human diseases is an essential component of biomedical research, to understand their pathogenesis and ultimately, develop therapeutic approaches. Here, we will describe models of tumours of the central nervous system, with focus on intrinsic CNS tumours. Model systems for brain tumours were established as early as the 1920s, using chemical carcinogenesis, and a systematic analysis of different carcinogens, with a more refined histological analysis followed in the 1950s and 1960s. Alternative approaches at the time used retroviral carcinogenesis, allowing a more topical, organ-centred delivery. Most of the neoplasms arising from this approach were high-grade gliomas. Whilst these experimental approaches did not directly demonstrate a cell of origin, the localisation and growth pattern of the tumours already pointed to an origin in the neurogenic zones of the brain. In the 1980s, expression of oncogenes in transgenic models allowed a more targeted approach by expressing the transgene under tissue-specific promoters, whilst the constitutive inactivation of tumour suppressor genes ('knock out')-often resulted in embryonic lethality. This limitation was elegantly solved by engineering the Cre-lox system, allowing for a promoter-specific, and often also time-controlled gene inactivation. More recently, the use of the CRISPR Cas9 technology has significantly increased experimental flexibility of gene expression or gene inactivation and thus added increased value of rodent models for the study of pathogenesis and establishing preclinical models.
Collapse
Affiliation(s)
- Sebastian Brandner
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals, NHS Foundation TrustLondonUK
| |
Collapse
|
10
|
Chieppa M, Kashyrina M, Miraglia A, Vardanyan D. Enhanced CRC Growth in Iron-Rich Environment, Facts and Speculations. Int J Mol Sci 2024; 25:12389. [PMID: 39596454 PMCID: PMC11594836 DOI: 10.3390/ijms252212389] [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: 10/04/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The contribution of nutritional factors to disease development has been demonstrated for several chronic conditions including obesity, type 2 diabetes, metabolic syndrome, and about 30 percent of cancers. Nutrients include macronutrients and micronutrients, which are required in large and trace quantities, respectively. Macronutrients, which include protein, carbohydrates, and lipids, are mainly involved in energy production and biomolecule synthesis; micronutrients include vitamins and minerals, which are mainly involved in immune functions, enzymatic reactions, blood clotting, and gene transcription. Among the numerous micronutrients potentially involved in disease development, the present review will focus on iron and its relation to tumor development. Recent advances in the understanding of iron-related proteins accumulating in the tumor microenvironment shed light on the pivotal role of iron availability in sustaining pathological tumor hallmarks, including cell cycle regulation, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Marcello Chieppa
- Department of Experimental Medicine, University of Salento Centro Ecotekne, S.P.6, 73100 Lecce, Italy; (M.K.); (D.V.)
| | - Marianna Kashyrina
- Department of Experimental Medicine, University of Salento Centro Ecotekne, S.P.6, 73100 Lecce, Italy; (M.K.); (D.V.)
| | - Alessandro Miraglia
- Institute of Science of Food Production, Unit of Lecce, C.N.R., 73100 Lecce, Italy;
| | - Diana Vardanyan
- Department of Experimental Medicine, University of Salento Centro Ecotekne, S.P.6, 73100 Lecce, Italy; (M.K.); (D.V.)
| |
Collapse
|
11
|
Shimizu Y, Hirano S, Salah M, Hoshi N, Yamashita Y, Fukumoto T, Mukumoto N, Nakaoka A, Ishihara T, Miyawaki D, Ashida H, Sasaki R. Black Soybean Seed Coat Extract Suppresses Gut Tumorigenesis by Augmenting the Production of Gut Microbiota-Derived Short-Chain Fatty Acids. Cancers (Basel) 2024; 16:3846. [PMID: 39594801 PMCID: PMC11592864 DOI: 10.3390/cancers16223846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Proanthocyanidins (PACs) from black soybean seed coat have antioxidant and anti-tumorigenic properties. We investigated the anti-tumor properties and mechanisms of action of PACs on colorectal cancer (CRC). METHODS We fed the APCmin/+ mice, which are highly susceptible to spontaneous intestinal adenoma formation, diets supplemented with or without PACs for 7 weeks and assessed adverse effects, the number and size of intestinal polyps, and the expression of pro- and anti-proliferative proteins in the intestine. The mouse gut microbiome composition was analyzed, and the concentrations of gut short-chain fatty acids (SCFAs) were quantified. We also compared CRC incidence in Tamba in Japan, where black soybean is consumed frequently, with that in the rest of Japan. RESULTS The number and size of intestinal polyps notably decreased in the PAC-fed mice. Compared with control mice, the PAC-fed mice showed lower expression of proliferation markers proliferating cell nuclear antigen and β catenin and a higher expression of the anti-inflammatory protein oligomeric mucus gel-forming. PAC supplementation increased the prevalence and concentrations of beneficial gut microbes and SCFAs, respectively. CONCLUSIONS Diet supplemented with black soybean-derived PACs could prevent CRC development in mice through gut microbiome remodeling. Regions consuming black soybeans have low CRC incidence. Notably, the incidence of CRC, breast cancer, and liver cancer was significantly lower in Tamba than in the rest of Hyogo Prefecture or Japan. Future studies should delineate the mechanisms underlying the CRC-protective effects of PACs. Nevertheless, our results demonstrate the potential of including PACs in dietary recommendations for cancer prevention.
Collapse
Affiliation(s)
- Yasuyuki Shimizu
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| | - Shunta Hirano
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
- Radiological Division, Osaka Metropolitan University Hospital, Osaka 545-8586, Japan
| | - Mohammed Salah
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
- Biochemistry Department, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan;
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-0013, Japan; (Y.Y.); (H.A.)
| | - Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan;
| | - Naritoshi Mukumoto
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| | - Ai Nakaoka
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| | - Takeaki Ishihara
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| | - Daisuke Miyawaki
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-0013, Japan; (Y.Y.); (H.A.)
- Faculty of Food Science and Nutrition, Mukogawa Women’s University, Nishinomiya 663-8558, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; (Y.S.); (S.H.); (N.M.); (A.N.); (T.I.); (D.M.)
| |
Collapse
|
12
|
Georgieva TG, Darmoul D, Chen H, Cui H, Rice PFS, Barton JK, Besselsen DG, Ignatenko NA. Kallikrein-Related Peptidase 6 Contributes to Murine Intestinal Tumorigenesis Driven by a Mutant Adenomatous polyposis coli Gene. Cancers (Basel) 2024; 16:3842. [PMID: 39594797 PMCID: PMC11592602 DOI: 10.3390/cancers16223842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The objective of this study was to assess the role of a secreted serine protease, kallikrein-related peptidase 6 (KLK6), during colorectal tumorigenesis driven by a mutant Adenomatous polyposis coli (APC) tumor suppressor gene. A first analysis of KLK6 expression in the intestinal tract of Apc-mutant multiple intestinal neoplasia (ApcMin/+) mice revealed up to four-fold induction of Klk6 mRNA levels in adenomas relative to its level in the adjacent mucosa. METHODS AND RESULTS The presence of KLK6 protein in the adenomatous areas was confirmed by immunohistochemistry and optical coherence tomography/laser-induced fluorescence (OCT/LIF) imaging. To assess the contribution of the KLK6 expression on the Apc-mutant intestinal and colon tumorigenesis, we engineered a mouse with floxed alleles of the Klk6 gene (Klk6lox/lox) and crossed it with a mouse expressing the truncated APC protein under control of the intestinal tract-specific human CDX2P9.5-NLS Cre transgene (CPC;Apcfl/fl;Klk6+/+). We found that CPC;Apcfl/fl mice with disrupted Klk6 gene expression (CPC;Apcfl/fl;Klk6fl/fl) had a significantly smaller average size of the small intestinal and colon crypts (p < 0.001 and p = 0.04, respectively) and developed a significantly fewer adenomas (p = 0.01). Moreover, a decrease in high-grade adenomas (p = 0.03) and adenomas with a diameter above 2 mm (p < 0.0001) was noted in CPC;Apcfl/fl;Klk6fl/fl mice. Further molecular analysis showed that Klk6 gene inactivation in the small intestine and colon tissues of CPC;Apcfl/fl;Klk6fl/fl mice resulted in a significant suppression of transforming growth factor β2 (TGF-β2) protein (p ≤ 0.02) and mitogen-activated protein kinase (MAPK) phosphorylation (p ≤ 0.01). CONCLUSIONS These findings demonstrate the oncogenic role of KLK6 in the mutant Apc-mediated intestinal tumorigenesis and suggest the utility of KLK6 for early diagnosis of colorectal tumors.
Collapse
Affiliation(s)
- Teodora G. Georgieva
- Genetically Engineered Mouse Models Core, The University of Arizona Bio5 Institute, Tucson, AZ 85721-0240, USA;
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Dalila Darmoul
- Institut de Biologie Paris-Seine, Sorbonne Université, UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, 75005 Paris, France;
| | - Hwudaurw Chen
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Haiyan Cui
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Photini F. S. Rice
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721-0240, USA; (P.F.S.R.); (J.K.B.)
| | - Jennifer K. Barton
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721-0240, USA; (P.F.S.R.); (J.K.B.)
| | - David G. Besselsen
- University Animal Care, The University of Arizona, Tucson, AZ 85721-0101, USA;
| | - Natalia A. Ignatenko
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724-5024, USA
| |
Collapse
|
13
|
Shen C, Pandey A, Enosi Tuipulotu D, Mathur A, Liu L, Yang H, Adikari NK, Ngo C, Jing W, Feng S, Hao Y, Zhao A, Kirkby M, Kurera M, Zhang J, Venkataraman S, Liu C, Song R, Wong JJL, Schumann U, Natoli R, Wen J, Zhang L, Kaakoush NO, Man SM. Inflammasome protein scaffolds the DNA damage complex during tumor development. Nat Immunol 2024; 25:2085-2096. [PMID: 39402152 DOI: 10.1038/s41590-024-01988-6] [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: 02/26/2024] [Accepted: 09/13/2024] [Indexed: 10/30/2024]
Abstract
Inflammasome sensors activate cellular signaling machineries to drive inflammation and cell death processes. Inflammasomes also control the development of certain diseases independently of canonical functions. Here, we show that the inflammasome protein NLR family CARD domain-containing protein 4 (NLRC4) attenuated the development of tumors in the Apcmin/+ mouse model. This response was independent of inflammasome signaling by NLRP3, NLRP6, NLR family apoptosis inhibitory proteins, absent in melanoma 2, apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1 and caspase-11. NLRC4 interacted with the DNA-damage-sensing ataxia telangiectasia and Rad3-related (ATR)-ATR-interacting protein (ATRIP)-Ewing tumor-associated antigen 1 (ETAA1) complex to promote the recruitment of the checkpoint adapter protein claspin, licensing the activation of the kinase checkpoint kinase-1 (CHK1). Genotoxicity-induced activation of the NLRC4-ATR-ATRIP-ETAA1 complex drove the tumor-suppressing DNA damage response and CHK1 activation, and further attenuated the accumulation of DNA damage. These findings demonstrate a noninflammatory function of an inflammasome protein in promoting the DNA damage response and mediating protection against cancer.
Collapse
Affiliation(s)
- Cheng Shen
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Abhimanu Pandey
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Daniel Enosi Tuipulotu
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Anukriti Mathur
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Lixinyu Liu
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Canberra, Australian Capital Territory, Australia
| | - Haoyu Yang
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Canberra, Australian Capital Territory, Australia
| | - Nilanthi K Adikari
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Chinh Ngo
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Weidong Jing
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Shouya Feng
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yuwei Hao
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Anyang Zhao
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Max Kirkby
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Melan Kurera
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jing Zhang
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Shweta Venkataraman
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Cheng Liu
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Herston, Queensland, Australia
- Mater Pathology, Mater Hospital, South Brisbane, Queensland, Australia
| | - Renhua Song
- Epigenetics and RNA Biology Laboratory, The School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Justin J-L Wong
- Epigenetics and RNA Biology Laboratory, The School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Ulrike Schumann
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- The Shine Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- The Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- The Shine Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- School of Medicine and Psychology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jiayu Wen
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Canberra, Australian Capital Territory, Australia
| | - Liman Zhang
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Nadeem O Kaakoush
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Si Ming Man
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.
| |
Collapse
|
14
|
Li L, Wazir J, Huang Z, Wang Y, Wang H. A comprehensive review of animal models for cancer cachexia: Implications for translational research. Genes Dis 2024; 11:101080. [PMID: 39220755 PMCID: PMC11364047 DOI: 10.1016/j.gendis.2023.101080] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 09/04/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by progressive weight loss and a disease process that nutritional support cannot reverse. Although progress has been made in preclinical research, there is still a long way to go in translating research findings into clinical practice. One of the main reasons for this is that existing preclinical models do not fully replicate the conditions seen in clinical patients. Therefore, it is important to understand the characteristics of existing preclinical models of cancer cachexia and pay close attention to the latest developments in preclinical models. The main models of cancer cachexia used in current research are allogeneic and xenograft models, genetically engineered mouse models, chemotherapy drug-induced models, Chinese medicine spleen deficiency models, zebrafish and Drosophila models, and cellular models. This review aims to revisit and summarize the commonly used animal models of cancer cachexia by evaluating existing preclinical models, to provide tools and support for translational medicine research.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
15
|
Hua X, Zhao C, Tian J, Wang J, Miao X, Zheng G, Wu M, Ye M, Liu Y, Zhou Y. A Ctnnb1 enhancer transcriptionally regulates Wnt signaling dosage to balance homeostasis and tumorigenesis of intestinal epithelia. eLife 2024; 13:RP98238. [PMID: 39320349 PMCID: PMC11424096 DOI: 10.7554/elife.98238] [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: 09/26/2024] Open
Abstract
The β-catenin-dependent canonical Wnt signaling is pivotal in organ development, tissue homeostasis, and cancer. Here, we identified an upstream enhancer of Ctnnb1 - the coding gene for β-catenin, named ieCtnnb1 (intestinal enhancer of Ctnnb1), which is crucial for intestinal homeostasis. ieCtnnb1 is predominantly active in the base of small intestinal crypts and throughout the epithelia of large intestine. Knockout of ieCtnnb1 led to a reduction in Ctnnb1 transcription, compromising the canonical Wnt signaling in intestinal crypts. Single-cell sequencing revealed that ieCtnnb1 knockout altered epithelial compositions and potentially compromised functions of small intestinal crypts. While deletion of ieCtnnb1 hampered epithelial turnovers in physiologic conditions, it prevented occurrence and progression of Wnt/β-catenin-driven colorectal cancers. Human ieCTNNB1 drove reporter gene expression in a pattern highly similar to mouse ieCtnnb1. ieCTNNB1 contains a single-nucleotide polymorphism associated with CTNNB1 expression levels in human gastrointestinal epithelia. The enhancer activity of ieCTNNB1 in colorectal cancer tissues was stronger than that in adjacent normal tissues. HNF4α and phosphorylated CREB1 were identified as key trans-factors binding to ieCTNNB1 and regulating CTNNB1 transcription. Together, these findings unveil an enhancer-dependent mechanism controlling the dosage of Wnt signaling and homeostasis in intestinal epithelia.
Collapse
Affiliation(s)
- Xiaojiao Hua
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Chen Zhao
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Junbao Wang
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Gen Zheng
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Mei Ye
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Liu
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Yan Zhou
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Sasaki T, Ota Y, Takikawa Y, Terrooatea T, Kanaya T, Takahashi M, Taguchi-Atarashi N, Tachibana N, Yabukami H, Surh CD, Minoda A, Kim KS, Ohno H. Food antigens suppress small intestinal tumorigenesis. Front Immunol 2024; 15:1373766. [PMID: 39359724 PMCID: PMC11445177 DOI: 10.3389/fimmu.2024.1373766] [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: 01/20/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024] Open
Abstract
Food components suppressing small intestinal tumorigenesis are not well-defined partly because of the rarity of this tumor type compared to colorectal tumors. Using Apcmin/+ mice, a mouse model for intestinal tumorigenesis, and antigen-free diet, we report here that food antigens serve this function in the small intestine. By depleting Peyer's patches (PPs), immune inductive sites in the small intestine, we found that PPs have a role in the suppression of small intestinal tumors and are important for the induction of small intestinal T cells by food antigens. On the follicle-associated epithelium (FAE) of PPs, microfold (M) cells pass food antigens from lumen to the dendritic cells to induce T cells. Single-cell RNA-seq (scRNA-seq) analysis of immune cells in PPs revealed a significant impact of food antigens on the induction of the PP T cells and the antigen presentation capacity of dendritic cells. These data demonstrate the role of food antigens in the suppression of small intestinal tumorigenesis by PP-mediated immune cell induction.
Collapse
Affiliation(s)
- Takaharu Sasaki
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuna Ota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Yui Takikawa
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Tommy Terrooatea
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takashi Kanaya
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Masumi Takahashi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoko Taguchi-Atarashi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoko Tachibana
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Haruka Yabukami
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Charles D. Surh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Aki Minoda
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, Netherlands
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
17
|
Arteaga-Blanco LA, Evans AE, Dixon DA. Plasma-Derived Extracellular Vesicles and Non-Extracellular Vesicle Components from APC Min/+ Mice Promote Pro-Tumorigenic Activities and Activate Human Colonic Fibroblasts via the NF-κB Signaling Pathway. Cells 2024; 13:1195. [PMID: 39056778 PMCID: PMC11274984 DOI: 10.3390/cells13141195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer worldwide. Current studies have demonstrated that tumor-derived extracellular vesicles (EVs) from different cancer cell types modulate the fibroblast microenvironment to contribute to cancer development and progression. Here, we isolated and characterized circulating large EVs (LEVs), small EVs (SEVs) and non-EV entities released in the plasma from wild-type (WT) mice and the APCMin/+ CRC mice model. Our results showed that human colon fibroblasts exposed from APC-EVs, but not from WT-EVs, exhibited the phenotypes of cancer-associated fibroblasts (CAFs) through EV-mediated NF-κB pathway activation. Cytokine array analysis on secreted proteins revealed elevated levels of inflammatory cytokine implicated in cancer growth and metastasis. Finally, non-activated cells co-cultured with supernatant from fibroblasts treated with APC-EVs showed increased mRNA expressions of CAFs markers, the ECM, inflammatory cytokines, as well as the expression of genes controlled by NF-κB. Altogether, our work suggests that EVs and non-EV components from APCMin/+ mice are endowed with pro-tumorigenic activities and promoted inflammation and a CAF-like state by triggering NF-κB signaling in fibroblasts to support CRC growth and progression. These findings provide insight into the interaction between plasma-derived EVs and human cells and can be used to design new CRC diagnosis and prognosis tools.
Collapse
Affiliation(s)
| | - Andrew E. Evans
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Dan A. Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
- University of Kansas Comprehensive Cancer Center, Kansas City, KS 66103, USA
| |
Collapse
|
18
|
Guo B, Zheng Y, Fan Y, Yang Y, Wang Y, Qin L, An Y, Xu X, Zhang X, Sun G, Dou H, Shao C, Gong Y, Jiang B, Hu H. Enhanced Apc Min/+ adenoma formation after epithelial CUL4B deletion by recruitment of myeloid-derived suppressor cells. Neoplasia 2024; 53:101005. [PMID: 38761506 PMCID: PMC11127156 DOI: 10.1016/j.neo.2024.101005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/10/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Colorectal cancer (CRC) stands as a prevalent malignancy globally. A pivotal event in CRC pathogenesis involves the loss-of-function mutation in the APC gene, leading to the formation of benign polyps. Despite the well-established role of APC, the contribution of CUL4B to CRC initiation in the pre-tumorous stage remains poorly understood. In this investigation, we generated a murine model by crossing ApcMin/+ mice with Cul4bΔIEC mice to achieve specific deletion of Cul4b in the gut epithelium against an ApcMin/+ background. By employing histological methods, RNA-sequencing (RNA-seq), and flow cytometry, we assessed alterations and characterized the immune microenvironment. Our results unveiled that CUL4B deficiency in gut epithelium expedited ApcMin/+ adenoma formation. Notably, CUL4B in adenomas restrained the accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). In vivo inhibition of MDSCs significantly delayed the growth of CUL4B deleted ApcMin/+ adenomas. Furthermore, the addition of MDSCs to in vitro cultured ApcMin/+; Cul4bΔIEC adenoma organoids mitigated their alterations. Mechanistically, CUL4B directly interacted with the promoter of Csf3, the gene encoding granulocyte-colony stimulating factor (G-CSF) by coordinating with PRC2. Inhibiting CUL4B epigenetically activated the expression of G-CSF, promoting the recruitment of MDSCs. These findings offer novel insights into the tumor suppressor-like roles of CUL4B in regulating ApcMin/+ adenomas, suggesting a potential therapeutic strategy for CRC initiation and progression in the context of activated Wnt signaling.
Collapse
Affiliation(s)
- Beibei Guo
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yawen Zheng
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China; Department of Obstetrics & Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yujia Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yang Yang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuxing Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Liping Qin
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yachun An
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaoran Xu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiyu Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Histoembryology, Shandong University Cheeloo Medical College, Shandong University School of Medicine, Jinan, China
| | - Hao Dou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
19
|
Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, Dong Q, Wei X. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol 2024; 17:46. [PMID: 38886806 PMCID: PMC11184729 DOI: 10.1186/s13045-024-01563-4] [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: 03/03/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
The Wnt/β-catenin signaling pathway plays a crucial role in various physiological processes, encompassing development, tissue homeostasis, and cell proliferation. Under normal physiological conditions, the Wnt/β-catenin signaling pathway is meticulously regulated. However, aberrant activation of this pathway and downstream target genes can occur due to mutations in key components of the Wnt/β-catenin pathway, epigenetic modifications, and crosstalk with other signaling pathways. Consequently, these dysregulations contribute significantly to tumor initiation and progression. Therapies targeting the Wnt/β-catenin signaling transduction have exhibited promising prospects and potential for tumor treatment. An increasing number of medications targeting this pathway are continuously being developed and validated. This comprehensive review aims to summarize the latest advances in our understanding of the role played by the Wnt/β-catenin signaling pathway in carcinogenesis and targeted therapy, providing valuable insights into acknowledging current opportunities and challenges associated with targeting this signaling pathway in cancer research and treatment.
Collapse
Affiliation(s)
- Pan Song
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Zirui Gao
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yige Bao
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuhe Huang
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yanyan Liu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Qiang Dong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
20
|
Suzuki K, Tsuruoka C, Morioka T, Seo H, Ogawa M, Kambe R, Imaoka T, Kakinuma S, Takahashi A. Combined effects of radiation and simulated microgravity on intestinal tumorigenesis in C3B6F1 Apc Min/+ mice. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:202-209. [PMID: 38670648 DOI: 10.1016/j.lssr.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Explorations of the Moon and Mars are planned as future manned space missions, during which humans will be exposed to both radiation and microgravity. We do not, however, know the health effects for such combined exposures. In a ground-based experiment, we evaluated the combined effects of radiation and simulated microgravity on tumorigenesis by performing X-irradiation and tail suspension in C3B6F1 ApcMin/+ mice, a well-established model for intestinal tumorigenesis. Mice were irradiated at 2 weeks of age and underwent tail suspension for 3 or 11 weeks using a special device that avoids damage to the tail. The tail suspension treatment significantly reduced the thymus weight after 3 weeks but not 11 weeks, suggesting a transient stress response. The combination of irradiation and tail suspension significantly increased the number of small intestinal tumors less than 2 mm in diameter as compared with either treatment alone. The combined treatment also increased the fraction of malignant tumors among all small intestinal tumors as compared with the radiation-only treatment. Thus, the C3B6F1 ApcMin/+ mouse is a useful model for assessing cancer risk in a simulated space environment, in which simulated microgravity accelerates tumor progression when combined with radiation exposure.
Collapse
Affiliation(s)
- Kenshi Suzuki
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hitomi Seo
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Mari Ogawa
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Ryosuke Kambe
- Gunma University Heavy Ion Medical Center, Gunma, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| | | |
Collapse
|
21
|
Ren X, Liu Q, Zhou P, Zhou T, Wang D, Mei Q, Flavell RA, Liu Z, Li M, Pan W, Zhu S. DHX9 maintains epithelial homeostasis by restraining R-loop-mediated genomic instability in intestinal stem cells. Nat Commun 2024; 15:3080. [PMID: 38594251 PMCID: PMC11004185 DOI: 10.1038/s41467-024-47235-2] [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: 06/20/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
Epithelial barrier dysfunction and crypt destruction are hallmarks of inflammatory bowel disease (IBD). Intestinal stem cells (ISCs) residing in the crypts play a crucial role in the continuous self-renewal and rapid recovery of intestinal epithelial cells (IECs). However, how ISCs are dysregulated in IBD remains poorly understood. Here, we observe reduced DHX9 protein levels in IBD patients, and mice with conditional DHX9 depletion in the intestinal epithelium (Dhx9ΔIEC) exhibit an increased susceptibility to experimental colitis. Notably, Dhx9ΔIEC mice display a significant reduction in the numbers of ISCs and Paneth cells. Further investigation using ISC-specific or Paneth cell-specific Dhx9-deficient mice demonstrates the involvement of ISC-expressed DHX9 in maintaining epithelial homeostasis. Mechanistically, DHX9 deficiency leads to abnormal R-loop accumulation, resulting in genomic instability and the cGAS-STING-mediated inflammatory response, which together impair ISC function and contribute to the pathogenesis of IBD. Collectively, our findings highlight R-loop-mediated genomic instability in ISCs as a risk factor in IBD.
Collapse
Affiliation(s)
- Xingxing Ren
- Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, 510145, Guangzhou, China
| | - Qiuyuan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Peirong Zhou
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, 510145, Guangzhou, China
| | - Tingyue Zhou
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Decai Wang
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhanju Liu
- Center for IBD Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, 510145, Guangzhou, China.
| | - Wen Pan
- Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Shu Zhu
- Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
- Key Laboratory of immune response and immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Data Science, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
22
|
Murphy KC, Ruscetti M. Advances in Making Cancer Mouse Models More Accessible and Informative through Non-Germline Genetic Engineering. Cold Spring Harb Perspect Med 2024; 14:a041348. [PMID: 37277206 PMCID: PMC10982712 DOI: 10.1101/cshperspect.a041348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Genetically engineered mouse models (GEMMs) allow for modeling of spontaneous tumorigenesis within its native microenvironment in mice and have provided invaluable insights into mechanisms of tumorigenesis and therapeutic strategies to treat human disease. However, as their generation requires germline manipulation and extensive animal breeding that is time-, labor-, and cost-intensive, traditional GEMMs are not accessible to most researchers, and fail to model the full breadth of cancer-associated genetic alterations and therapeutic targets. Recent advances in genome-editing technologies and their implementation in somatic tissues of mice have ushered in a new class of mouse models: non-germline GEMMs (nGEMMs). nGEMM approaches can be leveraged to generate somatic tumors de novo harboring virtually any individual or group of genetic alterations found in human cancer in a mouse through simple procedures that do not require breeding, greatly increasing the accessibility and speed and scale on which GEMMs can be produced. Here we describe the technologies and delivery systems used to create nGEMMs and highlight new biological insights derived from these models that have rapidly informed functional cancer genomics, precision medicine, and immune oncology.
Collapse
Affiliation(s)
- Katherine C Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA;
- Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
23
|
Bonnet V, Maikranz E, Madec M, Vertti-Quintero N, Cuche C, Mastrogiovanni M, Alcover A, Di Bartolo V, Baroud CN. Cancer-on-a-chip model shows that the adenomatous polyposis coli mutation impairs T cell engagement and killing of cancer spheroids. Proc Natl Acad Sci U S A 2024; 121:e2316500121. [PMID: 38442157 PMCID: PMC10945811 DOI: 10.1073/pnas.2316500121] [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: 09/22/2023] [Accepted: 01/25/2024] [Indexed: 03/07/2024] Open
Abstract
Evaluating the ability of cytotoxic T lymphocytes (CTLs) to eliminate tumor cells is crucial, for instance, to predict the efficiency of cell therapy in personalized medicine. However, the destruction of a tumor by CTLs involves CTL migration in the extra-tumoral environment, accumulation on the tumor, antigen recognition, and cooperation in killing the cancer cells. Therefore, identifying the limiting steps in this complex process requires spatio-temporal measurements of different cellular events over long periods. Here, we use a cancer-on-a-chip platform to evaluate the impact of adenomatous polyposis coli (APC) mutation on CTL migration and cytotoxicity against 3D tumor spheroids. The APC mutated CTLs are found to have a reduced ability to destroy tumor spheroids compared with control cells, even though APC mutants migrate in the extra-tumoral space and accumulate on the spheroids as efficiently as control cells. Once in contact with the tumor however, mutated CTLs display reduced engagement with the cancer cells, as measured by a metric that distinguishes different modes of CTL migration. Realigning the CTL trajectories around localized killing cascades reveals that all CTLs transition to high engagement in the 2 h preceding the cascades, which confirms that the low engagement is the cause of reduced cytotoxicity. Beyond the study of APC mutations, this platform offers a robust way to compare cytotoxic cell efficiency of even closely related cell types, by relying on a multiscale cytometry approach to disentangle complex interactions and to identify the steps that limit the tumor destruction.
Collapse
Affiliation(s)
- Valentin Bonnet
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Erik Maikranz
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Marianne Madec
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Faculty of Medicine, Department of Pathology and Immunology, University of Geneva, Geneva 4CH-1211, Switzerland
| | - Nadia Vertti-Quintero
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
| | - Céline Cuche
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Marta Mastrogiovanni
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Albert Einstein College of Medicine, Department of Developmental and Molecular Biology, New York, NY10461
| | - Andrés Alcover
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Vincenzo Di Bartolo
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Charles N. Baroud
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| |
Collapse
|
24
|
Tran HT, Wan MLY, Ambite I, Cavalera M, Grossi M, Háček J, Esmaeili P, Carneiro ANBM, Chaudhuri A, Ahmadi S, Svanborg C. BAMLET administration via drinking water inhibits intestinal tumor development and promotes long-term health. Sci Rep 2024; 14:3838. [PMID: 38360830 PMCID: PMC10869698 DOI: 10.1038/s41598-024-54040-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
Though new targeted therapies for colorectal cancer, which progresses from local intestinal tumors to metastatic disease, are being developed, tumor specificity remains an important problem, and side effects a major concern. Here, we show that the protein-fatty acid complex BAMLET (bovine alpha-lactalbumin made lethal to tumor cells) can act as a peroral treatment for colorectal cancer. ApcMin/+ mice, which carry mutations relevant to hereditary and sporadic human colorectal cancer, that received BAMLET in the drinking water showed long-term protection against tumor development and decreased expression of tumor growth-, migration-, metastasis- and angiogenesis-related genes. BAMLET treatment via drinking water inhibited the Wnt/β-catenin and PD-1 signaling pathways and prolonged survival without evidence of toxicity. Systemic disease in the lungs, livers, spleens, and kidneys, which accompanied tumor progression, was inhibited by BAMLET treatment. The metabolic response to BAMLET included carbohydrate and lipid metabolism, which were inhibited in tumor prone ApcMin/+ mice and weakly regulated in C57BL/6 mice, suggesting potential health benefits of peroral BAMLET administration in addition to the potent antitumor effects. Together, these findings suggest that BAMLET administration in the drinking water maintains antitumor pressure by removing emergent cancer cells and reprogramming gene expression in intestinal and extra-intestinal tissues.
Collapse
Affiliation(s)
- Hien Thi Tran
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Murphy Lam Yim Wan
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Ines Ambite
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Michele Cavalera
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Mario Grossi
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Jaromir Háček
- Department of Pathology and Molecular Medicine, Motol University Hospital, 2nd Faculty of Medicine, Charles University Praha, 150 06, Prague, Czech Republic
| | - Parisa Esmaeili
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - António N B M Carneiro
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Arunima Chaudhuri
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Shahram Ahmadi
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden.
| |
Collapse
|
25
|
Ferrer-Mayorga G, Muñoz A, González-Sancho JM. Vitamin D and colorectal cancer. FELDMAN AND PIKE'S VITAMIN D 2024:859-899. [DOI: 10.1016/b978-0-323-91338-6.00039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
26
|
Yakou MH, Afshar-Sterle S, Ernst M, Mielke LA. Orthotopic MC-38 Allograft as a Robust Preclinical Model of Colorectal Carcinoma. Methods Mol Biol 2024; 2806:197-207. [PMID: 38676804 DOI: 10.1007/978-1-0716-3858-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Colorectal cancer (CRC) is a significant global health concern, requiring effective preclinical models for studying its development and testing therapies. Mouse models have been used, including spontaneous tumors, carcinogen exposure, and tumor cell implantation as xenografts or at orthotopic sites. Here, we describe an orthotopic preclinical model of CRC, which provides a valuable tool for studying tumor growth and the tumor microenvironment, offering a more accurate representation of human CRC compared to xenograft models.
Collapse
Affiliation(s)
- Marina H Yakou
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia.
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia.
| | - Lisa A Mielke
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia.
| |
Collapse
|
27
|
Ben-Shahar Y, Vasserman V, Pollak Y, Kremer K, Sukhotnik I. The mechanism of intestinal stem cells differentiation after ischemia-reperfusion injury in a rat model. Pediatr Surg Int 2023; 40:23. [PMID: 38108924 DOI: 10.1007/s00383-023-05610-y] [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] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Notch and Wnt/β-catenin signaling are responsible for regulation of intestinal stem cells (ISCs) proliferation and differentiation. The purpose of the study was to evaluate Wnt/β-catenin and Notch signaling roles in regulation of ISC differentiation following ischemia-reperfusion (IR) injury in a rat. METHODS Rats were assigned into two groups: Sham rats underwent laparotomy without vascular intervention and IR rats underwent occlusion of SMA and portal vein for 20 min followed by 48 h of reperfusion. Wnt/β-catenin and Notch-related gene expression were determined using Real-Time PCR. Enterocyte proliferation, differentiation and Wnt-related proteins were determined by immunohistochemistry. RESULTS IR rats demonstrated a significant decrease in β-catenin gene expression, a decrease in cyclin D1 and β-catenin positive cells in jejunum and ileum compared to Sham rats. IR rats demonstrated a significant increase in Notch-related gene expression in jejunum and ileum compared to Sham rats. The number of secretory cells was higher mainly in the jejunum and number of absorptive cells was significantly lower in jejunum and lower in ileum in IR rats compared to Sham rats. CONCLUSIONS Intestinal stem-cell differentiation is toward secretory cells 48 h after IR injury; however, Wnt/β-catenin pathway inhibition and Notch-related gene expression stimulation suggest crosstalk between pathways.
Collapse
Affiliation(s)
- Yoav Ben-Shahar
- Laboratory of Intestinal Adaptation and Recovery, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel.
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel.
| | - Victoria Vasserman
- Laboratory of Intestinal Adaptation and Recovery, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yulia Pollak
- Laboratory of Intestinal Adaptation and Recovery, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Keren Kremer
- Laboratory of Intestinal Adaptation and Recovery, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Igor Sukhotnik
- Laboratory of Intestinal Adaptation and Recovery, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| |
Collapse
|
28
|
Ahmed AU, Almasabi S, Firestein R, Williams BRG. Integrin-linked kinase expression in myeloid cells promotes colon tumorigenesis. Front Immunol 2023; 14:1270194. [PMID: 38077324 PMCID: PMC10710162 DOI: 10.3389/fimmu.2023.1270194] [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: 07/31/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common forms of cancer worldwide and treatment options for advanced CRC, which has a low 5-year survival rate, remain limited. Integrin-linked kinase (ILK), a multifunctional, scaffolding, pseudo-kinase regulating many integrin-mediated cellular processes, is highly expressed in many cancers. However, the role of ILK in cancer progression is yet to be fully understood. We have previously uncovered a pro-inflammatory role for myeloid-specific ILK in dextran sodium sulfate (DSS)-induced colitis. To establish a correlation between chronic intestinal inflammation and colorectal cancer (CRC), we investigated the role of myeloid-ILK in mouse models of CRC. When myeloid-ILK deficient mice along with the WT control mice were subjected to colitis-associated and APCmin/+-driven CRC, tumour burden was reduced by myeloid-ILK deficiency in both models. The tumour-promoting phenotype of macrophages, M2 polarization, in vitro was impaired by the ILK deficiency and the number of M2-specific marker CD206-expressing tumour-associated macrophages (TAMs) in vivo were significantly diminished in myeloid-ILK deficient mice. Myeloid-ILK deficient mice showed enhanced tumour infiltration of CD8+ T cells and reduced tumour infiltration of FOXP3+ T cells in colitis-associated and APCmin/+-driven CRC, respectively, with an overall elevated CD8+/FOXP3+ ratio suggesting an anti-tumour immune phenotypes. In patient CRC tissue microarrays we observed elevated ILK+ myeloid (ILK+ CD11b+) cells in tumour sections compared to adjacent normal tissues, suggesting a conserved role for myeloid-ILK in CRC development in both human and animal models. This study identifies myeloid-specific ILK expression as novel driver of CRC, which could be targeted as a potential therapeutic option for advanced disease.
Collapse
Affiliation(s)
- Afsar U Ahmed
- Centre for Cancer Research, Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Saleh Almasabi
- Centre for Cancer Research, Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Ron Firestein
- Centre for Cancer Research, Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Bryan R G Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| |
Collapse
|
29
|
Chiriac MT, Hracsko Z, Becker C, Neurath MF. STAT2 Controls Colorectal Tumorigenesis and Resistance to Anti-Cancer Drugs. Cancers (Basel) 2023; 15:5423. [PMID: 38001683 PMCID: PMC10670206 DOI: 10.3390/cancers15225423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Colorectal cancer (CRC) is a significant socioeconomic burden in modern society and is accountable for millions of premature deaths each year. The role of signal transducer and activator of transcription 2 (STAT2)-dependent signaling in this context is not yet fully understood, and no therapies targeting this pathway are currently being pursued. We investigated the role of STAT2 in CRC using experimental mouse models coupled with RNA-sequencing (RNA-Seq) data and functional assays with anti-cancer agents in three-dimensional tumoroids. Stat2-/- mice showed greater resistance to the development of CRC in both inflammation-driven and inflammation-independent experimental CRC models. In ex vivo studies, tumoroids derived from Stat2-/- mice with the multiple intestinal neoplasia (Min) mutant allele of the adenomatous polyposis coli (Apc) locus exhibited delayed growth, were overall smaller and more differentiated as compared with tumoroids from ApcMin/+ wildtype (WT) mice. Notably, tumoroids from ApcMin/+ Stat2-/- mice were more susceptible to anti-cancer agents inducing cell death by different mechanisms. Our findings clearly indicated that STAT2 promotes CRC and suggested that interventions targeting STAT2-dependent signals might become an attractive therapeutic option for patients with CRC.
Collapse
Affiliation(s)
- Mircea T. Chiriac
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Zsuzsanna Hracsko
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
- Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
30
|
Li W, Nakano H, Fan W, Li Y, Sil P, Nakano K, Zhao F, Karmaus PW, Grimm SA, Shi M, Xu X, Mizuta R, Kitamura D, Wan Y, Fessler MB, Cook DN, Shats I, Li X, Li L. DNASE1L3 enhances antitumor immunity and suppresses tumor progression in colon cancer. JCI Insight 2023; 8:e168161. [PMID: 37581941 PMCID: PMC10544201 DOI: 10.1172/jci.insight.168161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/02/2023] [Indexed: 08/17/2023] Open
Abstract
DNASE1L3, an enzyme highly expressed in DCs, is functionally important for regulating autoimmune responses to self-DNA and chromatin. Deficiency of DNASE1L3 leads to development of autoimmune diseases in both humans and mice. However, despite the well-established causal relationship between DNASE1L3 and immunity, little is known about the involvement of DNASE1L3 in regulation of antitumor immunity, the foundation of modern antitumor immunotherapy. In this study, we identify DNASE1L3 as a potentially new regulator of antitumor immunity and a tumor suppressor in colon cancer. In humans, DNASE1L3 is downregulated in tumor-infiltrating DCs, and this downregulation is associated with poor patient prognosis and reduced tumor immune cell infiltration in many cancer types. In mice, Dnase1l3 deficiency in the tumor microenvironment enhances tumor formation and growth in several colon cancer models. Notably, the increased tumor formation and growth in Dnase1l3-deficient mice are associated with impaired antitumor immunity, as evidenced by a substantial reduction of cytotoxic T cells and a unique subset of DCs. Consistently, Dnase1l3-deficient DCs directly modulate cytotoxic T cells in vitro. To our knowledge, our study unveils a previously unknown link between DNASE1L3 and antitumor immunity and further suggests that restoration of DNASE1L3 activity may represent a potential therapeutic approach for anticancer therapy.
Collapse
Affiliation(s)
- Wenling Li
- Biostatistics and Computational Biology Branch
- Signal Transduction Laboratory
| | | | - Wei Fan
- Biostatistics and Computational Biology Branch
- Signal Transduction Laboratory
| | - Yuanyuan Li
- Biostatistics and Computational Biology Branch
| | - Payel Sil
- Biostatistics and Computational Biology Branch
| | | | - Fei Zhao
- Immunity, Inflammation, and Disease Laboratory
| | | | | | - Min Shi
- Biostatistics and Computational Biology Branch
| | - Xin Xu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina, USA
| | - Ryushin Mizuta
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Daisuke Kitamura
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yisong Wan
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, North Carolina, USA
| | | | | | | | | | - Leping Li
- Biostatistics and Computational Biology Branch
| |
Collapse
|
31
|
Ji M, Xu X, Xu Q, Hsiao YC, Martin C, Ukraintseva S, Popov V, Arbeev KG, Randall TA, Wu X, Garcia-Peterson LM, Liu J, Xu X, Andrea Azcarate-Peril M, Wan Y, Yashin AI, Anantharaman K, Lu K, Li JL, Shats I, Li X. Methionine restriction-induced sulfur deficiency impairs antitumour immunity partially through gut microbiota. Nat Metab 2023; 5:1526-1543. [PMID: 37537369 PMCID: PMC10513933 DOI: 10.1038/s42255-023-00854-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023]
Abstract
Restriction of methionine (MR), a sulfur-containing essential amino acid, has been reported to repress cancer growth and improve therapeutic responses in several preclinical settings. However, how MR impacts cancer progression in the context of the intact immune system is unknown. Here we report that while inhibiting cancer growth in immunocompromised mice, MR reduces T cell abundance, exacerbates tumour growth and impairs tumour response to immunotherapy in immunocompetent male and female mice. Mechanistically, MR reduces microbial production of hydrogen sulfide, which is critical for immune cell survival/activation. Dietary supplementation of a hydrogen sulfide donor or a precursor, or methionine, stimulates antitumour immunity and suppresses tumour progression. Our findings reveal an unexpected negative interaction between MR, sulfur deficiency and antitumour immunity and further uncover a vital role of gut microbiota in mediating this interaction. Our study suggests that any possible anticancer benefits of MR require careful consideration of both the microbiota and the immune system.
Collapse
Affiliation(s)
- Ming Ji
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Qing Xu
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cody Martin
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Svetlana Ukraintseva
- Social Science Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Vladimir Popov
- Social Science Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Konstantin G Arbeev
- Social Science Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Tom A Randall
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xiaoyue Wu
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Liz M Garcia-Peterson
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Xin Xu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology and Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yisong Wan
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anatoliy I Yashin
- Social Science Research Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Kun Lu
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Igor Shats
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
32
|
Cunningham P, Unger CA, Patton EA, Aiken A, Browne A, James E, Aladhami AK, Hope 3rd MC, VanderVeen BN, Cardaci TD, Murphy EA, Enos RT, Velázquez KT. Platelet status in cancer cachexia progression in Apc Min/+ mice. Front Immunol 2023; 14:1253587. [PMID: 37701438 PMCID: PMC10493779 DOI: 10.3389/fimmu.2023.1253587] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/10/2023] [Indexed: 09/14/2023] Open
Abstract
Cachexia, a complex wasting syndrome, significantly affects the quality of life and treatment options for cancer patients. Studies have reported a strong correlation between high platelet count and decreased survival in cachectic individuals. Therefore, this study aimed to investigate the immunopathogenesis of cancer cachexia using the ApcMin/+ mouse model of spontaneous colorectal cancer. The research focused on identifying cellular elements in the blood at different stages of cancer cachexia, assessing inflammatory markers and fibrogenic factors in the skeletal muscle, and studying the behavioral and metabolic phenotype of ApcMin/+ mice at the pre-cachectic and severely cachectic stages. Platelet measurements were also obtained from other animal models of cancer cachexia - Lewis Lung Carcinoma and Colon 26 adenocarcinoma. Our study revealed that platelet number is elevated prior to cachexia development in ApcMin/+ mice and can become activated during its progression. We also observed increased expression of TGFβ2, TGFβ3, and SMAD3 in the skeletal muscle of pre-cachectic ApcMin/+ mice. In severely cachectic mice, we observed an increase in Ly6g, CD206, and IL-10 mRNA. Meanwhile, IL-1β gene expression was elevated in the pre-cachectic stage. Our behavioral and metabolic phenotyping results indicate that pre-cachectic ApcMin/+ mice exhibit decreased physical activity. Additionally, we found an increase in anemia at pre-cachectic and severely cachectic stages. These findings highlight the altered platelet status during early and late stages of cachexia and provide a basis for further investigation of platelets in the field of cancer cachexia.
Collapse
Affiliation(s)
- Patrice Cunningham
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Christian A. Unger
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Emma A. Patton
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Akyla Aiken
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- Columbia Department of Veterans Affairs Health Care System, Columbia, SC, United States
| | - Alea Browne
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Ella James
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Ahmed K. Aladhami
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Marion C. Hope 3rd
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- Columbia Department of Veterans Affairs Health Care System, Columbia, SC, United States
| |
Collapse
|
33
|
Vande Voorde J, Steven RT, Najumudeen AK, Ford CA, Dexter A, Gonzalez-Fernandez A, Nikula CJ, Xiang Y, Ford L, Maneta Stavrakaki S, Gilroy K, Zeiger LB, Pennel K, Hatthakarnkul P, Elia EA, Nasif A, Murta T, Manoli E, Mason S, Gillespie M, Lannagan TRM, Vlahov N, Ridgway RA, Nixon C, Raven A, Mills M, Athineos D, Kanellos G, Nourse C, Gay DM, Hughes M, Burton A, Yan B, Sellers K, Wu V, De Ridder K, Shokry E, Huerta Uribe A, Clark W, Clark G, Kirschner K, Thienpont B, Li VSW, Maddocks ODK, Barry ST, Goodwin RJA, Kinross J, Edwards J, Yuneva MO, Sumpton D, Takats Z, Campbell AD, Bunch J, Sansom OJ. Metabolic profiling stratifies colorectal cancer and reveals adenosylhomocysteinase as a therapeutic target. Nat Metab 2023; 5:1303-1318. [PMID: 37580540 PMCID: PMC10447251 DOI: 10.1038/s42255-023-00857-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/06/2023] [Indexed: 08/16/2023]
Abstract
The genomic landscape of colorectal cancer (CRC) is shaped by inactivating mutations in tumour suppressors such as APC, and oncogenic mutations such as mutant KRAS. Here we used genetically engineered mouse models, and multimodal mass spectrometry-based metabolomics to study the impact of common genetic drivers of CRC on the metabolic landscape of the intestine. We show that untargeted metabolic profiling can be applied to stratify intestinal tissues according to underlying genetic alterations, and use mass spectrometry imaging to identify tumour, stromal and normal adjacent tissues. By identifying ions that drive variation between normal and transformed tissues, we found dysregulation of the methionine cycle to be a hallmark of APC-deficient CRC. Loss of Apc in the mouse intestine was found to be sufficient to drive expression of one of its enzymes, adenosylhomocysteinase (AHCY), which was also found to be transcriptionally upregulated in human CRC. Targeting of AHCY function impaired growth of APC-deficient organoids in vitro, and prevented the characteristic hyperproliferative/crypt progenitor phenotype driven by acute deletion of Apc in vivo, even in the context of mutant Kras. Finally, pharmacological inhibition of AHCY reduced intestinal tumour burden in ApcMin/+ mice indicating its potential as a metabolic drug target in CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yuchen Xiang
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Lauren Ford
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Stefania Maneta Stavrakaki
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | | | - Lucas B Zeiger
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Kathryn Pennel
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | - Eftychios Manoli
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Sam Mason
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Michael Gillespie
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Megan Mills
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | | | - Craig Nourse
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - David M Gay
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Københavns Universitet, BRIC, Copenhagen, Denmark
| | - Mark Hughes
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Amy Burton
- National Physical Laboratory, London, UK
| | - Bin Yan
- National Physical Laboratory, London, UK
| | - Katherine Sellers
- The Francis Crick Institute, London, UK
- Rheos Medicines, Cambridge, MA, USA
| | - Vincen Wu
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Kobe De Ridder
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Engy Shokry
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | | | - Graeme Clark
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Bernard Thienpont
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | | | | | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Richard J A Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - James Kinross
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Zoltan Takats
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Biological Mass Spectrometry, Rosalind Franklin Institute, Didcot, UK
| | | | - Josephine Bunch
- National Physical Laboratory, London, UK
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Biological Mass Spectrometry, Rosalind Franklin Institute, Didcot, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
34
|
Keller MP, Hudkins KL, Shalev A, Bhatnagar S, Kebede MA, Merrins MJ, Davis DB, Alpers CE, Kimple ME, Attie AD. What the BTBR/J mouse has taught us about diabetes and diabetic complications. iScience 2023; 26:107036. [PMID: 37360692 PMCID: PMC10285641 DOI: 10.1016/j.isci.2023.107036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Human and mouse genetics have delivered numerous diabetogenic loci, but it is mainly through the use of animal models that the pathophysiological basis for their contribution to diabetes has been investigated. More than 20 years ago, we serendipidously identified a mouse strain that could serve as a model of obesity-prone type 2 diabetes, the BTBR (Black and Tan Brachyury) mouse (BTBR T+ Itpr3tf/J, 2018) carrying the Lepob mutation. We went on to discover that the BTBR-Lepob mouse is an excellent model of diabetic nephropathy and is now widely used by nephrologists in academia and the pharmaceutical industry. In this review, we describe the motivation for developing this animal model, the many genes identified and the insights about diabetes and diabetes complications derived from >100 studies conducted in this remarkable animal model.
Collapse
Affiliation(s)
- Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Anath Shalev
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Melkam A. Kebede
- School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, Sydney, NSW 2006, Australia
| | - Matthew J. Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Charles E. Alpers
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Michelle E. Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
35
|
Le Naour J, Montégut L, Pan Y, Scuderi SA, Cordier P, Joseph A, Sauvat A, Iebba V, Paillet J, Ferrere G, Brechard L, Mulot C, Dubourg G, Zitvogel L, Pol JG, Vacchelli E, Puig PL, Kroemer G. Formyl peptide receptor-1 (FPR1) represses intestinal oncogenesis. Oncoimmunology 2023; 12:2237354. [PMID: 37492227 PMCID: PMC10364666 DOI: 10.1080/2162402x.2023.2237354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Abstract
Formyl peptide receptor-1 (FPR1) is a pattern recognition receptor that is mostly expressed by myeloid cells. In patients with colorectal cancer (CRC), a loss-of-function polymorphism (rs867228) in the gene coding for FPR1 has been associated with reduced responses to chemotherapy or chemoradiotherapy. Moreover, rs867228 is associated with accelerated esophageal and colorectal carcinogenesis. Here, we show that dendritic cells from Fpr1-/- mice exhibit reduced migration in response to chemotherapy-treated CRC cells. Moreover, Fpr1-/- mice are particularly susceptible to chronic ulcerative colitis and colorectal oncogenesis induced by the mutagen azoxymethane followed by oral dextran sodium sulfate, a detergent that induces colitis. These experiments were performed after initial co-housing of Fpr1-/- mice and wild-type controls, precluding major Fpr1-driven differences in the microbiota. Pharmacological inhibition of Fpr1 by cyclosporin H also tended to increase intestinal oncogenesis in mice bearing the ApcMin mutation, and this effect was reversed by the anti-inflammatory drug sulindac. We conclude that defective FPR1 signaling favors intestinal tumorigenesis through the modulation of the innate inflammatory/immune response.
Collapse
Affiliation(s)
- Julie Le Naour
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Yuhong Pan
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Sarah Adriana Scuderi
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Pierre Cordier
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Valerio Iebba
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Juliette Paillet
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Gladys Ferrere
- Institut National de la Santé Et de la Recherche Medicale (INSERM) U1015 and Equipe Labellisée–Ligue Nationale Contre le Cancer, Villejuif, France
| | - Ludivine Brechard
- Aix Marseille Univ, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, Equipe Labélisée Ligue Contre le Cancer, Sorbonne Université, Université Paris Cité, INSERM, Paris, France
| | - Grégory Dubourg
- Aix Marseille Univ, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Laurence Zitvogel
- Faculty of Medicine Kremlin Bicêtre, Université Paris Saclay, Le Kremlin Bicêtre, France
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
| | - Jonathan G. Pol
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Erika Vacchelli
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
| | - Pierre-Laurent Puig
- Centre de Recherche des Cordeliers, Equipe Labélisée Ligue Contre le Cancer, Sorbonne Université, Université Paris Cité, INSERM, Paris, France
- Institut du Cancer Paris CARPEM, APHP. Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Villejuif, France
- Institut du Cancer Paris CARPEM, APHP. Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
36
|
Semba R, Morioka T, Yanagihara H, Suzuki K, Tachibana H, Hamoya T, Horimoto Y, Imaoka T, Saito M, Kakinuma S, Arai M. Azithromycin induces read-through of the nonsense Apc allele and prevents intestinal tumorigenesis in C3B6F1 Apc Min/+ mice. Biomed Pharmacother 2023; 164:114968. [PMID: 37276642 DOI: 10.1016/j.biopha.2023.114968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/07/2023] Open
Abstract
Therapeutic strategies that promote read-through of a mutant gene have proved effective for certain non-neoplastic diseases. However, the efficacy of this approach is unproven regarding neoplastic diseases with germline nonsense mutations, including familial adenomatous polyposis. Here we examined the cancer-preventive efficacy of the macrolide antibiotic azithromycin, with a reported read-through effect, on intestinal tumorigenesis in C3B6F1 ApcMin/+ mice harboring a nonsense Apc mutation resulting in a truncated Apc protein. Mice were given drinking water lacking azithromycin or containing 0.0125-0.2 mg/mL azithromycin from 3 weeks of age. The small intestine and cecum were analyzed for pathological changes and alterations of intestinal flora. Azithromycin suppressed the number of tumors and the proportion of adenocarcinomas, with the most effective drinking-water concentration being 0.0125 mg/mL. Furthermore, azithromycin recovered the cellular level of full-length Apc, resulting in downregulation of β-catenin and cyclin D1. Conversely, the effect of azithromycin on the diversity of the intestinal microbiota depended on the drinking-water concentration. These results suggest that the balance between azithromycin-mediate read-through of mutant Apc mRNA and antibacterial effects influences intestinal tumorigenesis. Thus, azithromycin is a potential anticancer agent for familial adenomatous polyposis patients harboring nonsense mutations.
Collapse
Affiliation(s)
- Ryoko Semba
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan; Department of Breast Oncology, Juntendo University School of Medicine, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Hiromi Yanagihara
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Kenshi Suzuki
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Hirotaka Tachibana
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Takahiro Hamoya
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University School of Medicine, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University School of Medicine, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology (NIRS/QST), Japan.
| | - Masami Arai
- Department of Clinical Genetics, Juntendo University School of Medicine, Japan
| |
Collapse
|
37
|
Yanagihara H, Morioka T, Yamazaki S, Yamada Y, Tachibana H, Daino K, Tsuruoka C, Amasaki Y, Kaminishi M, Imaoka T, Kakinuma S. Interstitial deletion of the Apc locus in β-catenin-overexpressing cells is a signature of radiation-induced intestinal tumors in C3B6F1 ApcMin/+ mice†. JOURNAL OF RADIATION RESEARCH 2023; 64:622-631. [PMID: 37117033 DOI: 10.1093/jrr/rrad021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/13/2023] [Indexed: 05/27/2023]
Abstract
Recent studies have identified interstitial deletions in the cancer genome as a radiation-related mutational signature, although most of them do not fall on cancer driver genes. Pioneering studies in the field have indicated the presence of loss of heterozygosity (LOH) spanning Apc in a subset of sporadic and radiation-induced intestinal tumors of ApcMin/+ mice, albeit with a substantial subset in which LOH was not detected; whether copy number losses accompany such LOH has also been unclear. Herein, we analyzed intestinal tumors of C3B6F1 ApcMin/+ mice that were either left untreated or irradiated with 2 Gy of γ-rays. We observed intratumor mosaicism with respect to the nuclear/cytoplasmic accumulation of immunohistochemically detectable β-catenin, which is a hallmark of Apc+ allele loss. An immunoguided laser microdissection approach enabled the detection of LOH involving the Apc+ allele in β-catenin-overexpressing cells; in contrast, the LOH was not observed in the non-overexpressing cells. With this improvement, LOH involving Apc+ was detected in all 22 tumors analyzed, in contrast to what has been reported previously. The use of a formalin-free fixative facilitated the LOH and microarray-based DNA copy number analyses, enabling the classification of the aberrations as nondisjunction/mitotic recombination type or interstitial deletion type. Of note, the latter was observed only in radiation-induced tumors (nonirradiated, 0 of 8; irradiated, 11 of 14). Thus, an analysis considering intratumor heterogeneity identifies interstitial deletion involving the Apc+ allele as a causative radiation-related event in intestinal tumors of ApcMin/+ mice, providing an accurate approach for attributing individual tumors to radiation exposure.
Collapse
Affiliation(s)
- Hiromi Yanagihara
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Shunsuke Yamazaki
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yutaka Yamada
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hirotaka Tachibana
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Japan
| | - Kazuhiro Daino
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Japan
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yoshiko Amasaki
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Mutsumi Kaminishi
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
38
|
Lanas A, Tacconelli S, Contursi A, Piazuelo E, Bruno A, Ronci M, Marcone S, Dovizio M, Sopeña F, Falcone L, Milillo C, Mucci M, Ballerini P, Patrignani P. Biomarkers of Response to Low-Dose Aspirin in Familial Adenomatous Polyposis Patients. Cancers (Basel) 2023; 15:cancers15092457. [PMID: 37173923 PMCID: PMC10177499 DOI: 10.3390/cancers15092457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND The results of Aspirin prevention of colorectal adenomas in patients with familial adenomatous polyposis (FAP) are controversial. METHODS We conducted a biomarker-based clinical study in eight FAP patients treated with enteric-coated low-dose Aspirin (100 mg daily for three months) to explore whether the drug targets mainly platelet cyclooxygenase (COX)-1 or affects extraplatelet cellular sources expressing COX-isozymes and/or off-target effects in colorectal adenomas. RESULTS In FAP patients, low-dose Aspirin-acetylated platelet COX-1 at Serine529 (>70%) was associated with an almost complete inhibition of platelet thromboxane (TX) B2 generation ex vivo (serum TXB2). However, enhanced residual urinary 11-dehydro-TXB2 and urinary PGEM, primary metabolites of TXA2 and prostaglandin (PG)E2, respectively, were detected in association with incomplete acetylation of COX-1 in normal colorectal biopsies and adenomas. Proteomics of adenomas showed that Aspirin significantly modulated only eight proteins. The upregulation of vimentin and downregulation of HBB (hemoglobin subunit beta) distinguished two groups with high vs. low residual 11-dehydro-TXB2 levels, possibly identifying the nonresponders and responders to Aspirin. CONCLUSIONS Although low-dose Aspirin appropriately inhibited the platelet, persistently high systemic TXA2 and PGE2 biosynthesis were found, plausibly for a marginal inhibitory effect on prostanoid biosynthesis in the colorectum. Novel chemotherapeutic strategies in FAP can involve blocking the effects of TXA2 and PGE2 signaling with receptor antagonists.
Collapse
Affiliation(s)
- Angel Lanas
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, 50009 Zaragoza, Spain
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Elena Piazuelo
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | - Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Maurizio Ronci
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Simone Marcone
- Trinity Translational Medicine Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Melania Dovizio
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Federico Sopeña
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, 50009 Zaragoza, Spain
| | - Lorenza Falcone
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Cristina Milillo
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Matteo Mucci
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, 66100 Chieti, Italy
| |
Collapse
|
39
|
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: 1.5] [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
|
40
|
Abbott J, Näthke IS. The adenomatous polyposis coli protein 30 years on. Semin Cell Dev Biol 2023:S1084-9521(23)00093-9. [PMID: 37095033 DOI: 10.1016/j.semcdb.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 04/26/2023]
Abstract
Mutations in the gene encoding the Adenomatous polyposis coli protein (APC) were discovered as driver mutations in colorectal cancers almost 30 years ago. Since then, the importance of APC in normal tissue homeostasis has been confirmed in a plethora of other (model) organisms spanning a large evolutionary space. APC is a multifunctional protein, with roles as a key scaffold protein in complexes involved in diverse signalling pathways, most prominently the Wnt signalling pathway. APC is also a cytoskeletal regulator with direct and indirect links to and impacts on all three major cytoskeletal networks. Correspondingly, a wide range of APC binding partners have been identified. Mutations in APC are extremely strongly associated with colorectal cancers, particularly those that result in the production of truncated proteins and the loss of significant regions from the remaining protein. Understanding the complement of its role in health and disease requires knowing the relationship between and regulation of its diverse functions and interactions. This in turn requires understanding its structural and biochemical features. Here we set out to provide a brief overview of the roles and function of APC and then explore its conservation and structure using the extensive sequence data, which is now available, and spans a broad range of taxonomy. This revealed conservation of APC across taxonomy and new relationships between different APC protein families.
Collapse
Affiliation(s)
- James Abbott
- Division of Computational Biology & D'Arcy Thompson Unit, University of Dundee, Dow Street, Dundee, DD2 1 EH, United Kingdom.
| | - Inke S Näthke
- Division of Molecular Cell and Developmental Biology, University of Dundee, Dow Street, Dundee DD2 1EH, United Kingdom.
| |
Collapse
|
41
|
Domènech-Moreno E, Brandt A, Lemmetyinen TT, Wartiovaara L, Mäkelä TP, Ollila S. Tellu - an object-detector algorithm for automatic classification of intestinal organoids. Dis Model Mech 2023; 16:297124. [PMID: 36804687 PMCID: PMC10067441 DOI: 10.1242/dmm.049756] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Intestinal epithelial organoids recapitulate many of the in vivo features of the intestinal epithelium, thus representing excellent research models. Morphology of the organoids based on light-microscopy images is used as a proxy to assess the biological state of the intestinal epithelium. Currently, organoid classification is manual and, therefore, subjective and time consuming, hampering large-scale quantitative analyses. Here, we describe Tellu, an object-detector algorithm trained to classify cultured intestinal organoids. Tellu was trained by manual annotation of >20,000 intestinal organoids to identify cystic non-budding organoids, early organoids, late organoids and spheroids. Tellu can also be used to quantify the relative organoid size, and can classify intestinal organoids into these four subclasses with accuracy comparable to that of trained scientists but is significantly faster and without bias. Tellu is provided as an open, user-friendly online tool to benefit the increasing number of investigations using organoids through fast and unbiased organoid morphology and size analysis.
Collapse
Affiliation(s)
- Eva Domènech-Moreno
- HiLIFE-Helsinki Institute of Life Science, Yliopistonkatu 4, 00014 University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Stenbäckinkatu 9 Hallintokeskus, University of Helsinki, 00290 Helsinki, Finland
| | - Anders Brandt
- HiLIFE-Helsinki Institute of Life Science, Yliopistonkatu 4, 00014 University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Stenbäckinkatu 9 Hallintokeskus, University of Helsinki, 00290 Helsinki, Finland
| | - Toni T Lemmetyinen
- Translational Cancer Medicine Program, University of Helsinki, 00100 Helsinki, Finland
| | - Linnea Wartiovaara
- Translational Cancer Medicine Program, University of Helsinki, 00100 Helsinki, Finland
| | - Tomi P Mäkelä
- HiLIFE-Helsinki Institute of Life Science, Yliopistonkatu 4, 00014 University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Stenbäckinkatu 9 Hallintokeskus, University of Helsinki, 00290 Helsinki, Finland
| | - Saara Ollila
- Translational Cancer Medicine Program, University of Helsinki, 00100 Helsinki, Finland
| |
Collapse
|
42
|
Novellasdemunt L, Kucharska A, Baulies A, Hutton C, Vlachogiannis G, Repana D, Rowan A, Suárez-Bonnet A, Ciccarelli F, Valeri N, Li VSW. USP7 inactivation suppresses APC-mutant intestinal hyperproliferation and tumor development. Stem Cell Reports 2023; 18:570-584. [PMID: 36669491 PMCID: PMC9968985 DOI: 10.1016/j.stemcr.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023] Open
Abstract
Adenomatous polyposis coli (APC) mutation is the hallmark of colorectal cancer (CRC), resulting in constitutive WNT activation. Despite decades of research, targeting WNT signaling in cancer remains challenging due to its on-target toxicity. We have previously shown that the deubiquitinating enzyme USP7 is a tumor-specific WNT activator in APC-truncated cells by deubiquitinating and stabilizing β-catenin, but its role in gut tumorigenesis is unknown. Here, we show in vivo that deletion of Usp7 in Apc-truncated mice inhibits crypt hyperproliferation and intestinal tumor development. Loss of Usp7 prolongs the survival of the sporadic intestinal tumor model. Genetic deletion, but not pharmacological inhibition, of Usp7 in Apc+/- intestine induces colitis and enteritis. USP7 inhibitor treatment suppresses growth of patient-derived cancer organoids carrying APC truncations in vitro and in xenografts. Our findings provide direct evidence that USP7 inhibition may offer a safe and efficacious tumor-specific therapy for both sporadic and germline APC-mutated CRC.
Collapse
Affiliation(s)
- Laura Novellasdemunt
- Stem Cell and Cancer Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Anna Kucharska
- Stem Cell and Cancer Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Anna Baulies
- Stem Cell and Cancer Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Colin Hutton
- Stem Cell and Cancer Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Georgios Vlachogiannis
- Centre for Molecular Pathology, the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Dimitra Repana
- Cancer Systems Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - A Suárez-Bonnet
- Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK; Experimental Histopathology, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Francesca Ciccarelli
- Cancer Systems Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicola Valeri
- Centre for Molecular Pathology, the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, the Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
43
|
Garside GB, Sandoval M, Beronja S, Rudolph KL. Lentiviral in situ targeting of stem cells in unperturbed intestinal epithelium. BMC Biol 2023; 21:6. [PMID: 36627630 PMCID: PMC9832770 DOI: 10.1186/s12915-022-01466-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/16/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Methods for the long-term in situ transduction of the unperturbed murine intestinal epithelium have not been developed in past research. Such a method could speed up functional studies and screens to identify genetic factors influencing intestinal epithelium biology. Here, we developed an efficient method achieving this long-sought goal. RESULTS We used ultrasound-guided microinjections to transduce the embryonic endoderm at day 8 (E8.0) in utero. The injection procedure can be completed in 20 min and had a 100% survival rate. By injecting a small volume (0.1-0.2 μl) of concentrated virus, single shRNA constructs as well as lentiviral libraries can successfully be transduced. The new method stably and reproducibly targets adult intestinal epithelium, as well as other endoderm-derived organs such as the lungs, pancreas, liver, stomach, and bladder. Postnatal analysis of young adult mice indicates that single transduced cells at E8.0 gave rise to crypt fields that were comprised of 20-30 neighbouring crypts per crypt-field at 90 days after birth. Lentiviral targeting of ApcMin/+ mutant and wildtype mice revealed that heterozygous loss of Apc function suppresses the developmental normal growth pattern of intestinal crypt fields. This suppression of crypt field sizes did not involve a reduction of the crypt number per field, indicating that heterozygous Apc loss impaired the growth of individual crypts within the fields. Lentiviral-mediated shRNA knockdown of p53 led to an approximately 20% increase of individual crypts per field in both Apc+/+ and ApcMin/+ mice, associating with an increase in crypt size in ApcMin/+ mice but a slight reduction in crypt size in Apc+/+ mice. Overall, p53 knockdown rescued the reduction in crypt field size in Apc-mutant mice but had no effect on crypt field size in wildtype mice. CONCLUSIONS This study develops a novel technique enabling robust and reproducible in vivo targeting of intestinal stem cells in situ in the unperturbed intestinal epithelium across different regions of the intestine. In vivo somatic gene editing and genetic screening of lentiviral libraries has the potential to speed up discoveries and mechanistic understanding of genetic pathways controlling the biology of the intestinal epithelium during development and postnatal life. The here developed method enables such approaches.
Collapse
Affiliation(s)
- George B. Garside
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
- National Center for Tumor Diseases, 01307 Dresden, Germany
| | - Madeline Sandoval
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98109 USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98109 USA
| | - K. Lenhard Rudolph
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| |
Collapse
|
44
|
WU X, XU J, WANG D, YANG X, XU X. A new anti-colon cancer tumor pathway of Phenyllactic acid by reducing adhesion of Fusobacterium nucleatum. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.123222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Xinyu WU
- Northeast Agricultural University, China; Northeast Agricultural University, China
| | - Jinzhao XU
- Northeast Agricultural University, China; Northeast Agricultural University, China
| | - Danping WANG
- Northeast Agricultural University, China; Northeast Agricultural University, China
| | - Xiaoying YANG
- Northeast Agricultural University, China; Northeast Agricultural University, China
| | - Xiaoxi XU
- Northeast Agricultural University, China; Northeast Agricultural University, China
| |
Collapse
|
45
|
Teixeira SA, Luzzi MDC, Martin ACBM, Duarte TT, Leal MDO, Teixeira GR, Reis MT, Junior CRA, Santos K, Melendez ME, da Silva DDCSA, Bernécule PN, Firmino HVL, Alves ALV, Guimarães DP, Borduqui JV, Laus AC, Mançano BM, Reis RM. The Barretos Cancer Hospital Animal Facility: Implementation and Results of a Dedicated Platform for Preclinical Oncology Models. Vet Sci 2022; 9:636. [PMID: 36423085 PMCID: PMC9699001 DOI: 10.3390/vetsci9110636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 03/17/2025] Open
Abstract
The Barretos Cancer Hospital Animal Facility (BCHAF) is a unique facility in Brazil exclusively dedicated to working with animal models for cancer research. In this article, we briefly present our modern facility and the main experiments performed, focusing on mutant strains of mice (PTCH-knockout and ApcMin mice), xenograft models, and patient-derived xenografts (PDXs). Our results show the progress and challenges in establishing these models and the need for having an appropriate representation of our cancer population to better understand tumor biology and to identify cancer biomarkers, which could be putatively targeted, allowing for personalized therapy.
Collapse
Affiliation(s)
- Silvia A. Teixeira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Mayara de Cassia Luzzi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | | | - Terence Teixeira Duarte
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Mônica de Oliveira Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Gustavo Ramos Teixeira
- Department of Pathology, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata—FACISB, Barretos 14785-002, São Paulo, Brazil
| | - Monise Tadin Reis
- Department of Pathology, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | | | - Karina Santos
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Department of Radiology, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Matias Eliseo Melendez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Molecular Carcinogenesis Program, National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | | | - Priscila Neves Bernécule
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata—FACISB, Barretos 14785-002, São Paulo, Brazil
| | - Higor Vinicius Lourenço Firmino
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata—FACISB, Barretos 14785-002, São Paulo, Brazil
| | - Ana Laura Vieira Alves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Department of Endoscopy, Barretos Cancer Hospital, Barretos 14780-000, São Paulo, Brazil
| | - João Vitor Borduqui
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata—FACISB, Barretos 14785-002, São Paulo, Brazil
| | - Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Bruna Minniti Mançano
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Department of Neurosurgery, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| |
Collapse
|
46
|
Bruno A, Contursi A, Tacconelli S, Sacco A, Hofling U, Mucci M, Lamolinara A, Del Pizzo F, Ballerini P, Di Gregorio P, Yu Y, Patrignani P. The specific deletion of cyclooxygenase-1 in megakaryocytes/platelets reduces intestinal polyposis in Apc Min/+ mice. Pharmacol Res 2022; 185:106506. [PMID: 36241001 DOI: 10.1016/j.phrs.2022.106506] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/26/2022] [Accepted: 10/09/2022] [Indexed: 10/31/2022]
Abstract
Clinical and experimental evidence sustain the role of cyclooxygenase (COX)-1 in intestinal tumorigenesis. However, the cell type expressing the enzyme involved and molecular mechanism(s) have not been clarified yet. We aimed to elucidate the role of platelet COX-1 (the target of low-dose aspirin in humans) in intestinal tumorigenesis of ApcMin/+ mice, considered a clinically relevant model. To realize this objective, we generated an ApcMin/+ mouse with a specific deletion of Ptgs1(COX-1 gene name) in megakaryocytes/platelets (ApcMin/+;pPtgs1-/-mice) characterized by profound inhibition of thromboxane(TX)A2 biosynthesis ex vivo (serum TXB2; by 99%) and in vivo [urinary 2,3-dinor-TXB2(TXM), by 79%]. ApcMin/+ mice with the deletion of platelet COX-1 showed a significantly reduced number (67%) and size (32%) of tumors in the small intestine. The intestinal adenomas of these mice had decreased proliferative index associated with reduced COX-2 expression and systemic prostaglandin(PG)E2 biosynthesis (urinary PGEM) vs. ApcMin/+mice. Extravasated platelets were detected in the intestine of ApcMin/+mice. Thus, we explored their contribution to COX-2 induction in fibroblasts, considered the primary polyp cell type expressing the protein. In the coculture of human platelets and myofibroblasts, platelet-derived TXA2 was involved in the induction of COX-2-dependent PGE2 in myofibroblasts since it was prevented by the selective inhibition of platelet COX-1 by aspirin or by a specific antagonist of TXA2 receptors. In conclusion, our results support the platelet hypothesis of intestinal tumorigenesis and provide experimental evidence that selective inhibition of platelet COX-1 can mitigate early events of intestinal tumorigenesis by restraining COX-2 induction.
Collapse
Affiliation(s)
- Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Angela Sacco
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Ulrika Hofling
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Matteo Mucci
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Alessia Lamolinara
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Francesco Del Pizzo
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Patrizia Di Gregorio
- Institute of Transfusion Medicine, "Ss. Annunziata" Hospital, 66100 Chieti, Italy
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy.
| |
Collapse
|
47
|
Tomaz LB, Liu BA, Meroshini M, Ong SLM, Tan EK, Tolwinski NS, Williams CS, Gingras AC, Leushacke M, Dunn NR. MCC is a centrosomal protein that relocalizes to non-centrosomal apical sites during intestinal cell differentiation. J Cell Sci 2022; 135:jcs259272. [PMID: 36217793 PMCID: PMC10658790 DOI: 10.1242/jcs.259272] [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: 08/13/2021] [Accepted: 09/27/2022] [Indexed: 11/20/2022] Open
Abstract
The gene mutated in colorectal cancer (MCC) encodes a coiled-coil protein implicated, as its name suggests, in the pathogenesis of hereditary human colon cancer. To date, however, the contributions of MCC to intestinal homeostasis and disease remain unclear. Here, we examine the subcellular localization of MCC, both at the mRNA and protein levels, in the adult intestinal epithelium. Our findings reveal that Mcc transcripts are restricted to proliferating crypt cells, including Lgr5+ stem cells, where the Mcc protein is distinctly associated with the centrosome. Upon intestinal cellular differentiation, Mcc is redeployed to the apical domain of polarized villus cells where non-centrosomal microtubule organizing centers (ncMTOCs) are positioned. Using intestinal organoids, we show that the shuttling of the Mcc protein depends on phosphorylation by casein kinases 1δ and ε, which are critical modulators of WNT signaling. Together, our findings support a role for MCC in establishing and maintaining the cellular architecture of the intestinal epithelium as a component of both the centrosome and ncMTOC.
Collapse
Affiliation(s)
- Lucian B. Tomaz
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Bernard A. Liu
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Meroshini M
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Sheena L. M. Ong
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Ee Kim Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | | | | | - Anne-Claude Gingras
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Marc Leushacke
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| | - N. Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| |
Collapse
|
48
|
Colozza G, Park SY, Koo BK. Clone wars: From molecules to cell competition in intestinal stem cell homeostasis and disease. Exp Mol Med 2022; 54:1367-1378. [PMID: 36117218 PMCID: PMC9534868 DOI: 10.1038/s12276-022-00854-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/27/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
The small intestine is among the fastest self-renewing tissues in adult mammals. This rapid turnover is fueled by the intestinal stem cells residing in the intestinal crypt. Wnt signaling plays a pivotal role in regulating intestinal stem cell renewal and differentiation, and the dysregulation of this pathway leads to cancer formation. Several studies demonstrate that intestinal stem cells follow neutral drift dynamics, as they divide symmetrically to generate other equipotent stem cells. Competition for niche space and extrinsic signals in the intestinal crypt is the governing mechanism that regulates stemness versus cell differentiation, but the underlying molecular mechanisms are still poorly understood, and it is not yet clear how this process changes during disease. In this review, we highlight the mechanisms that regulate stem cell homeostasis in the small intestine, focusing on Wnt signaling and its regulation by RNF43 and ZNRF3, key inhibitors of the Wnt pathway. Furthermore, we summarize the evidence supporting the current model of intestinal stem cell regulation, highlighting the principles of neutral drift at the basis of intestinal stem cell homeostasis. Finally, we discuss recent studies showing how cancer cells bypass this mechanism to gain a competitive advantage against neighboring normal cells. Stem cells in the gut rapidly renew themselves through processes that cancer cells co-opt to trigger tumor development. Gabriele Colozza from the Institute of Molecular Biotechnology in Vienna, Austria, and colleagues review how a network of critical molecular signals and competition for limited space help to regulate the dynamics of stem cells in the intestines. The correct balance between self-renewal and differentiation is tightly controlled by the so-called Wnt signaling pathway and its inhibitors. Competition between dividing cells in the intestinal crypts, the locations between finger-like protrusions in the gut where stem cells are found, provides another protective mechanism against runaway stem cell growth. However, intestinal cancer cells, thanks to their activating mutations, bypass these safeguards to gain a survival advantage. Drugs that target these ‘super-competitive’ behaviors could therefore help combat tumor proliferation.
Collapse
|
49
|
Yu H, Chen C, Han F, Tang J, Deng M, Niu Y, Lai M, Zhang H. Long Noncoding RNA MIR4435-2HG Suppresses Colorectal Cancer Initiation and Progression By Reprogramming Neutrophils. Cancer Immunol Res 2022; 10:1095-1110. [PMID: 35862232 PMCID: PMC9433964 DOI: 10.1158/2326-6066.cir-21-1011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/31/2022] [Accepted: 07/15/2022] [Indexed: 01/07/2023]
Abstract
MIR4435-2HG, also known as LINC00978, has previously been described as an oncogenic long noncoding RNA (lncRNA). However, we show here that Mir4435-2hg depletion promoted colorectal tumorigenesis and progression in in vivo models of colitis-associated colorectal cancer, spontaneous intestinal adenomatous polyposis, and subcutaneous tumors. Alteration of MIR4435-2HG in colorectal cancer cells did not change the potential for cell proliferation, migration, or invasion in vitro. RNAscope assays showed that most MIR4435-2HG was located in the tumor stroma, which caused high expression of MIR4435-2HG in colorectal cancer tumor tissue. Transcriptome analysis of colorectal cancer tissues from wild-type and Mir4435-2hg-deficient mice revealed Mir4435-2hg as a tumor suppressor gene that regulated the immune microenvironment. Loss of Mir4435-2hg led to a decline in neutrophils and elevation of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC). In tissue-specific Mir4435-2hg knockout mice, we confirmed that Mir4435-2hg depletion in neutrophils, but not in intestinal epithelial cells, promoted colorectal cancer progression. Mechanistically, Mir4435-2hg depletion enhanced the immunosuppressive ability of PMN-MDSCs by disturbing their fatty acid metabolism. These findings suggest that MIR4435-2HG is a tumor-suppressing lncRNA whose deficiency could increase tumor-infiltrating PMN-MDSCs and enhance the immunosuppressive potential of PMN-MDSCs to promote colorectal cancer development. This provides a theoretical basis for further illustrating the pathogenesis of colorectal cancer and a potential antitumor immunotherapy target.
Collapse
Affiliation(s)
- Hongfei Yu
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Chaoyi Chen
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Fengyan Han
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Jinlong Tang
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Mengli Deng
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Yumiao Niu
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Maode Lai
- Department of Pathology, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China.,Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Corresponding Authors: Honghe Zhang, Department of Pathology, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Yuhangtang Road 866#, Hangzhou, Zhejiang 310058, China. E-mails: ; and Maode Lai,
| | - Honghe Zhang
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China.,Corresponding Authors: Honghe Zhang, Department of Pathology, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Yuhangtang Road 866#, Hangzhou, Zhejiang 310058, China. E-mails: ; and Maode Lai,
| |
Collapse
|
50
|
Krieg C, Weber LM, Fosso B, Marzano M, Hardiman G, Olcina MM, Domingo E, El Aidy S, Mallah K, Robinson MD, Guglietta S. Complement downregulation promotes an inflammatory signature that renders colorectal cancer susceptible to immunotherapy. J Immunother Cancer 2022; 10:e004717. [PMID: 36137652 PMCID: PMC9511657 DOI: 10.1136/jitc-2022-004717] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIMS The role of inflammatory immune responses in colorectal cancer (CRC) development and response to therapy is a matter of intense debate. While inflammation is a known driver of CRC, inflammatory immune infiltrates are a positive prognostic factor in CRC and predispose to response to immune checkpoint blockade (ICB) therapy. Unfortunately, over 85% of CRC cases are primarily unresponsive to ICB due to the absence of an immune infiltrate, and even the cases that show an initial immune infiltration can become refractory to ICB. The identification of therapy supportive immune responses in the field has been partially hindered by the sparsity of suitable mouse models to recapitulate the human disease. In this study, we aimed to understand how the dysregulation of the complement anaphylatoxin C3a receptor (C3aR), observed in subsets of patients with CRC, affects the immune responses, the development of CRC, and response to ICB therapy. METHODS We use a comprehensive approach encompassing analysis of publicly available human CRC datasets, inflammation-driven and newly generated spontaneous mouse models of CRC, and multiplatform high-dimensional analysis of immune responses using microbiota sequencing, RNA sequencing, and mass cytometry. RESULTS We found that patients' regulation of the complement C3aR is associated with epigenetic modifications. Specifically, downregulation of C3ar1 in human CRC promotes a tumor microenvironment characterized by the accumulation of innate and adaptive immune cells that support antitumor immunity. In addition, in vivo studies in our newly generated mouse model revealed that the lack of C3a in the colon activates a microbiota-mediated proinflammatory program which promotes the development of tumors with an immune signature that renders them responsive to the ICB therapy. CONCLUSIONS Our findings reveal that C3aR may act as a previously unrecognized checkpoint to enhance antitumor immunity in CRC. C3aR can thus be exploited to overcome ICB resistance in a larger group of patients with CRC.
Collapse
Affiliation(s)
- Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lukas M Weber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Bruno Fosso
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University of Belfast, Belfast, UK
| | - Monica M Olcina
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Enric Domingo
- Institute of Radiation Oncology, Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - Sahar El Aidy
- Host-microbe Metabolic Interactions, Microbiology, University of Groningen, Groningen, The Netherlands
| | - Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Silvia Guglietta
- Hollings Cancer Center Charleston, Medical University of South Carolina, Charleston, South Carolina, USA
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|