1
|
Nuñez-Rios JD, Ulrich H, Díaz-Muñoz M, Lameu C, Vázquez-Cuevas FG. Purinergic system in cancer stem cells. Purinergic Signal 2025; 21:23-38. [PMID: 37966629 PMCID: PMC11904000 DOI: 10.1007/s11302-023-09976-5] [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: 06/10/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Accumulating evidence supports the idea that cancer stem cells (CSCs) are those with the capacity to initiate tumors, generate phenotypical diversity, sustain growth, confer drug resistance, and orchestrate the spread of tumor cells. It is still controversial whether CSCs originate from normal stem cells residing in the tissue or cancer cells from the tumor bulk that have dedifferentiated to acquire stem-like characteristics. Although CSCs have been pointed out as key drivers in cancer, knowledge regarding their physiology is still blurry; thus, research focusing on CSCs is essential to designing novel and more effective therapeutics. The purinergic system has emerged as an important autocrine-paracrine messenger system with a prominent role at multiple levels of the tumor microenvironment, where it regulates cellular aspects of the tumors themselves and the stromal and immune systems. Recent findings have shown that purinergic signaling also participates in regulating the CSC phenotype. Here, we discuss updated information regarding CSCs in the purinergic system and present evidence supporting the idea that elements of the purinergic system expressed by this subpopulation of the tumor represent attractive pharmacological targets for proposing innovative anti-cancer therapies.
Collapse
Affiliation(s)
- J D Nuñez-Rios
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - H Ulrich
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - C Lameu
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México.
| |
Collapse
|
2
|
Willmer WFDEA, Samonge EFN, Barcia Junior OE, Bogossian GM, Assumpção LR, Marques RG. Comparison between Glasgow prognostic criteria and O-POSSUM/ P-POSSUM physiological indices in patients undergoing gastrectomy for gastric adenocarcinoma and the occurrency of early postoperative complications. Rev Col Bras Cir 2024; 51:e20243662. [PMID: 38985034 PMCID: PMC11449513 DOI: 10.1590/0100-6991e-20243662-en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/12/2024] [Indexed: 07/11/2024] Open
Abstract
INTRODUCTION Gastric cancer is still the third cause of death worldwide due to malignant neoplasms. Its prognostic indices have not yet been well defined for surgical intervention in terms of stratifying the intensity of chronic inflammation. The Glasgow Prognostic Score (GPS) and O-POSSUM and P-POSSUM Indices may constitute these standardizations and were tested to assess the association between them and the prognosis after curative gastrectomy. METHOD Retrospective observational study, analysing medical records of patients with gastric adenocarcinoma who underwent gastrectomy, from 2015 to 2021, in two hospitals in Rio de Janeiro. Surgical extension, pre, peri and postoperative clinical and laboratory data were observed, up to 30 days after surgery. Patients were layered by GPS and compared according to the Clavien-Dindo (CD) classification. Logistic regression was performed to test the association between the outcome and independent variables. RESULTS Of the 48 patients, 56.25% were female. There was difference between the groups regarding surgical extension and GPS (both with p<0.001), while O-POSSUM, P-POSSUM and age showed no difference. Factors associated with CD ≥ III-a complication in the univariate analysis were GPS (OR: 85,261; CI: 24,909- 291,831) and P-POSSUM (OR: 1,211; CI:1,044-1,404). In the multivariate analysis, the independent factors associated with CD ≥ III-a were GPS (OR:114,865; CI: 15,430-855,086), P-POSSUM (OR: 1,133; CI: 1,086-1,181) and O-POSSUM (OR: 2,238; CI: 1,790-2,797). CONCLUSION In this model, GPS, P-POSSUM and O-POSSUM predicted serious surgical complications. There is a need for further studies to establish strategies to minimize the inflammatory response in the preoperative period.
Collapse
Affiliation(s)
- William Frederic DE Araújo Willmer
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| | | | | | | | - Lia Roque Assumpção
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| | - Ruy Garcia Marques
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| |
Collapse
|
3
|
Willmer WFDEA, Samonge EFN, Barcia Junior OE, Bogossian GM, Assumpção LR, Marques RG. Comparison between Glasgow prognostic criteria and O-POSSUM/ P-POSSUM physiological indices in patients undergoing gastrectomy for gastric adenocarcinoma and the occurrency of early postoperative complications. Rev Col Bras Cir 2024; 51:e20243662. [PMID: 38985034 DOI: 10.1590/0100-6991e-20243662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/12/2024] [Indexed: 01/03/2025] Open
Abstract
INTRODUCTION Gastric cancer is still the third cause of death worldwide due to malignant neoplasms. Its prognostic indices have not yet been well defined for surgical intervention in terms of stratifying the intensity of chronic inflammation. The Glasgow Prognostic Score (GPS) and O-POSSUM and P-POSSUM Indices may constitute these standardizations and were tested to assess the association between them and the prognosis after curative gastrectomy. METHOD Retrospective observational study, analysing medical records of patients with gastric adenocarcinoma who underwent gastrectomy, from 2015 to 2021, in two hospitals in Rio de Janeiro. Surgical extension, pre, peri and postoperative clinical and laboratory data were observed, up to 30 days after surgery. Patients were layered by GPS and compared according to the Clavien-Dindo (CD) classification. Logistic regression was performed to test the association between the outcome and independent variables. RESULTS Of the 48 patients, 56.25% were female. There was difference between the groups regarding surgical extension and GPS (both with p<0.001), while O-POSSUM, P-POSSUM and age showed no difference. Factors associated with CD ≥ III-a complication in the univariate analysis were GPS (OR: 85,261; CI: 24,909- 291,831) and P-POSSUM (OR: 1,211; CI:1,044-1,404). In the multivariate analysis, the independent factors associated with CD ≥ III-a were GPS (OR:114,865; CI: 15,430-855,086), P-POSSUM (OR: 1,133; CI: 1,086-1,181) and O-POSSUM (OR: 2,238; CI: 1,790-2,797). CONCLUSION In this model, GPS, P-POSSUM and O-POSSUM predicted serious surgical complications. There is a need for further studies to establish strategies to minimize the inflammatory response in the preoperative period.
Collapse
Affiliation(s)
- William Frederic DE Araújo Willmer
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| | | | | | | | - Lia Roque Assumpção
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| | - Ruy Garcia Marques
- - Hospital Universitário Pedro Ernesto/UERJ, Programa de Pós-graduação em Fisiopatologia e Ciências Cirúrgicas - Rio de Janeiro - RJ - Brasil
| |
Collapse
|
4
|
Tiwari M, Srivastava P, Abbas S, Jegatheesan J, Ranjan A, Sharma S, Maurya VP, Saxena AK, Sharma LK. Emerging Role of Autophagy in Governing Cellular Dormancy, Metabolic Functions, and Therapeutic Responses of Cancer Stem Cells. Cells 2024; 13:447. [PMID: 38474411 PMCID: PMC10930960 DOI: 10.3390/cells13050447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Tumors are composed of heterogeneous populations of dysregulated cells that grow in specialized niches that support their growth and maintain their properties. Tumor heterogeneity and metastasis are among the major hindrances that exist while treating cancer patients, leading to poor clinical outcomes. Although the factors that determine tumor complexity remain largely unknown, several genotypic and phenotypic changes, including DNA mutations and metabolic reprograming provide cancer cells with a survival advantage over host cells and resistance to therapeutics. Furthermore, the presence of a specific population of cells within the tumor mass, commonly known as cancer stem cells (CSCs), is thought to initiate tumor formation, maintenance, resistance, and recurrence. Therefore, these CSCs have been investigated in detail recently as potential targets to treat cancer and prevent recurrence. Understanding the molecular mechanisms involved in CSC proliferation, self-renewal, and dormancy may provide important clues for developing effective therapeutic strategies. Autophagy, a catabolic process, has long been recognized to regulate various physiological and pathological processes. In addition to regulating cancer cells, recent studies have identified a critical role for autophagy in regulating CSC functions. Autophagy is activated under various adverse conditions and promotes cellular maintenance, survival, and even cell death. Thus, it is intriguing to address whether autophagy promotes or inhibits CSC functions and whether autophagy modulation can be used to regulate CSC functions, either alone or in combination. This review describes the roles of autophagy in the regulation of metabolic functions, proliferation and quiescence of CSCs, and its role during therapeutic stress. The review further highlights the autophagy-associated pathways that could be used to regulate CSCs. Overall, the present review will help to rationalize various translational approaches that involve autophagy-mediated modulation of CSCs in controlling cancer progression, metastasis, and recurrence.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Biochemistry, All India Institute of Medical Science, Patna 801507, India
| | - Pransu Srivastava
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Science, Lucknow 226014, India
| | - Sabiya Abbas
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Science, Lucknow 226014, India
| | - Janani Jegatheesan
- Department of Biochemistry, All India Institute of Medical Science, Patna 801507, India
| | - Ashish Ranjan
- Department of Biochemistry, All India Institute of Medical Science, Patna 801507, India
| | - Sadhana Sharma
- Department of Biochemistry, All India Institute of Medical Science, Patna 801507, India
| | - Ved Prakash Maurya
- Department of Neurosurgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Ajit Kumar Saxena
- Department of Pathology/Lab Medicine, All India Institute of Medical Science, Patna 801507, India
| | - Lokendra Kumar Sharma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Science, Lucknow 226014, India
| |
Collapse
|
5
|
Kasprzak A. Prognostic Biomarkers of Cell Proliferation in Colorectal Cancer (CRC): From Immunohistochemistry to Molecular Biology Techniques. Cancers (Basel) 2023; 15:4570. [PMID: 37760539 PMCID: PMC10526446 DOI: 10.3390/cancers15184570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/04/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and severe malignancies worldwide. Recent advances in diagnostic methods allow for more accurate identification and detection of several molecular biomarkers associated with this cancer. Nonetheless, non-invasive and effective prognostic and predictive testing in CRC patients remains challenging. Classical prognostic genetic markers comprise mutations in several genes (e.g., APC, KRAS/BRAF, TGF-β, and TP53). Furthermore, CIN and MSI serve as chromosomal markers, while epigenetic markers include CIMP and many other candidates such as SERP, p14, p16, LINE-1, and RASSF1A. The number of proliferation-related long non-coding RNAs (e.g., SNHG1, SNHG6, MALAT-1, CRNDE) and microRNAs (e.g., miR-20a, miR-21, miR-143, miR-145, miR-181a/b) that could serve as potential CRC markers has also steadily increased in recent years. Among the immunohistochemical (IHC) proliferative markers, the prognostic value regarding the patients' overall survival (OS) or disease-free survival (DFS) has been confirmed for thymidylate synthase (TS), cyclin B1, cyclin D1, proliferating cell nuclear antigen (PCNA), and Ki-67. In most cases, the overexpression of these markers in tissues was related to worse OS and DFS. However, slowly proliferating cells should also be considered in CRC therapy (especially radiotherapy) as they could represent a reservoir from which cells are recruited to replenish the rapidly proliferating population in response to cell-damaging factors. Considering the above, the aim of this article is to review the most common proliferative markers assessed using various methods including IHC and selected molecular biology techniques (e.g., qRT-PCR, in situ hybridization, RNA/DNA sequencing, next-generation sequencing) as prognostic and predictive markers in CRC.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Swiecicki Street 6, 60-781 Poznan, Poland
| |
Collapse
|
6
|
Jennelle LT, Dampier CH, Tring S, Powell S, Casey G. Colon Crypts of Subjects With Familial Adenomatous Polyposis Show an Increased Number of LGR5+ Ectopic Stem Cells. Clin Transl Gastroenterol 2021; 12:e00353. [PMID: 33999013 PMCID: PMC8133103 DOI: 10.14309/ctg.0000000000000353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 03/29/2021] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Familial adenomatous polyposis (FAP) is a hereditary colorectal cancer (CRC) syndrome characterized by accelerated adenoma development due to inherited (or de novo) mutations in the APC regulator of WNT signaling pathway (APC) gene. The mechanism underlying this accelerated polyp development in subjects with FAP has not been defined. Given that LGR5+ stem cells drive crypt cell proliferation, we hypothesized that FAP crypts would demonstrate aberrant leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) staining patterns. METHODS Biopsies were taken from 11 healthy subjects, 7 subjects with Lynch syndrome, 4 subjects with FAP, and 1 subject with MUTYH-associated polyposis syndrome during routine screening or surveillance colonoscopy. Crypt staining was evaluated by immunohistochemistry of paraffin-embedded tissue sections. Stem cell numbers were estimated by immunofluorescence staining of isolated crypts using antibodies against LGR5 and other proteins. RESULTS Subjects with FAP exhibited a greater number of LGR5+ stem cells in their crypts than healthy subjects and subjects with Lynch syndrome and MUTYH-associated polyposis syndrome. Most crypts of subjects with FAP harbored LGR5+ cells located above the lower third of the crypts. DISCUSSION These findings support a model in which inactivation of one copy of APC leads to increased numbers of LGR5+ stem cells, many of which are ectopic, in colon crypts of subjects with FAP. Overabundant and ectopic LGR5+ stem cells could lead to an expanded proliferative zone of dividing cells more likely to develop mutations that would contribute to the accelerated adenoma development observed in FAP.
Collapse
Affiliation(s)
- Lucas T. Jennelle
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Christopher H. Dampier
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
- Department of General Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Stephanie Tring
- USC Genomics Core, University of Southern California, Los Angeles, California, USA
| | - Steven Powell
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Graham Casey
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
7
|
Uchuya-Castillo J, Aznar N, Frau C, Martinez P, Le Nevé C, Marisa L, Penalva LOF, Laurent-Puig P, Puisieux A, Scoazec JY, Samarut J, Ansieau S, Plateroti M. Increased expression of the thyroid hormone nuclear receptor TRα1 characterizes intestinal tumors with high Wnt activity. Oncotarget 2018; 9:30979-30996. [PMID: 30123421 PMCID: PMC6089551 DOI: 10.18632/oncotarget.25741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/12/2018] [Indexed: 01/10/2023] Open
Abstract
Our previous work demonstrated a key function of the thyroid hormone nuclear receptor TRα1, a T3-modulated transcription factor, in controlling intestinal development and homeostasis via the Wnt and Notch pathways. Importantly, increased expression of TRα1 in the intestinal epithelium in a mutated Apc genetic background (vil-TRα1/Apc+/1638N mice) accelerated tumorigenesis and contributed to a more aggressive tumor phenotype compared to that of the Apc mutants alone. Therefore, the aim of this study was to determine the relevance of this synergistic effect in human colorectal cancers and to gain insights into the mechanisms involved. We analyzed cohorts of patients by in silico and experimental approaches and observed increased TRα1 expression and a significant correlation between TRα1 levels and Wnt activity. TRα1 loss-of-function and gain-of-function in Caco2 cell lines not only confirmed that TRα1 levels control Wnt activity but also demonstrated the role of TRα1 in regulating cell proliferation and migration. Finally, upon investigation of the molecular mechanisms responsible for the Wnt-TRα1 association, we described the repression by TRα1 of several Wnt inhibitors, including Frzb, Sox17 and Wif1. In conclusion, our results underline an important functional interplay between the thyroid hormone nuclear receptor TRα1 and the canonical Wnt pathway in intestinal cancer initiation and progression. More importantly, we show for the first time that the expression of TRα1 is induced in human colorectal cancers.
Collapse
Affiliation(s)
- Joel Uchuya-Castillo
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Nicolas Aznar
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Carla Frau
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Pierre Martinez
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Clementine Le Nevé
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Laetitia Marisa
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre le Cancer, Paris 75000, France
| | - Luiz O F Penalva
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | | | - Alain Puisieux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | | | - Jacques Samarut
- Institute de Génomique Fonctionnelle de Lyon, ENS de Lyon, Lyon 69342, France
| | - Stephane Ansieau
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| | - Michelina Plateroti
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Département de la recherche, Lyon 69000, France
| |
Collapse
|
8
|
Sonoda J, Seki Y, Hakura A, Hosokawa S. Time course of the incidence/multiplicity and histopathological features of murine colonic dysplasia, adenoma and adenocarcinoma induced by benzo[a]pyrene and dextran sulfate sodium. J Toxicol Pathol 2015; 28:109-20. [PMID: 26028820 PMCID: PMC4444509 DOI: 10.1293/tox.2014-0061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/05/2015] [Indexed: 12/29/2022] Open
Abstract
Benzo[a]pyrene (BP) is mutagenic but noncarcinogenic in the murine colon. Recently, we reported rapid induction of colonic tumors by treatment of CD2F1 mice with BP (125 mg/kg for 5 days) followed by a colitis inducer, dextran sulfate sodium (DSS) (4% in drinking water for 1 or 2 weeks). However, there are no reports on detailed time course and histopathological features of colonic proliferative lesions in this model. Here, we show the detailed time course of colonic dysplasia, adenoma and adenocarcinoma induced by treatment with BP, DSS, and a combination of the two (BP/DSS). In the colon of mice exposed to BP/DSS, 14.6 dysplastic foci per mouse were present one week after DSS treatment (week 4). The number of dysplastic foci decreased with time to 3.1 at week 9 and thereafter remained almost constant. At week 4, 1.5 adenocarcinomas were also observed, with a marked increase in numbers with time, reaching 29.3 at week 14. In contrast, the number of dysplastic foci induced by DSS alone showed a time course similar to that following BP/DSS treatment; however, only a few tumors appeared. Neither dysplastic foci nor neoplastic lesions were induced by BP only. In mice exposed to BP/DSS, β-catenin was demonstrated immunohistochemically in the nucleus and/or cytoplasm of the tumor cells, and this translocation from the cell membrane was evident in subsets of dysplastic foci. In dysplastic foci induced by DSS alone, β-catenin was absent in the nucleus/cytoplasm. These finding suggest that aberrant β-catenin accumulation in dysplastic foci is associated with tumor progression in this BP/DSS model.
Collapse
Affiliation(s)
- Jiro Sonoda
- Tsukuba Drug Safety, Global Drug Safety, Biopharmaceutical Assessments Core Function Unit, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Yuki Seki
- Tsukuba Drug Safety, Global Drug Safety, Biopharmaceutical Assessments Core Function Unit, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Atsushi Hakura
- Tsukuba Drug Safety, Global Drug Safety, Biopharmaceutical Assessments Core Function Unit, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Satoru Hosokawa
- Tsukuba Drug Safety, Global Drug Safety, Biopharmaceutical Assessments Core Function Unit, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| |
Collapse
|
9
|
Oshima CTF, Landman G, Paiotti APR, Artigiani Neto R, Silva RM, Campanholo VMDLP, Gollucke APB, Ribeiro DA, Forones NM. Grape juice concentrate (G8000™) modulates apoptosis but not oxidative stress following rat colon carcinogenesis induced by azoxymethane. Toxicol Mech Methods 2014; 25:91-7. [PMID: 25421551 DOI: 10.3109/15376516.2014.989556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate if grape juice concentrate is able to protect against experimental colon carcinogenesis. MATERIAL AND METHODS For this purpose, a total of 35 male Wistar rats were randomly distributed into seven groups: G1: SHAM animals receiving only saline; G2: animals receiving 15 mg/kg azoxymethane (AOM); G3: animals receiving 1% grape juice concentrate 2 weeks before the administration of AOM; G4: animals receiving 2% grape juice concentrate 2 weeks before the administration of AOM; G5: animals receiving 1% grape juice concentrate 4 weeks after the last administration of AOM; G6: animals receiving 2% grape juice concentrate 4 weeks after the last administration of AOM; G7: animals receiving only 2% grape juice concentrate. RESULTS The group that received 2% grape juice concentrate before induction with AOM showed the decreased expression of Bcl-2 compared to those animals that were induced by AOM (positive control). Regarding Bax, animals that received grape juice at 2% decreased Bax immunoexpression when compared to AOM group. Furthermore, animals that intake grape juice at 1% after induced by AOM decreased Bax immunoexpression as well. 8-OHdGLI did not show significant statistically differences (p > 0.05) among groups. CONCLUSION In summary, our results demonstrate that grape juice is able to modulate rat colon carcinogenesis as a result of induction of apoptosis.
Collapse
|
10
|
Azcárate-Peril MA, Sikes M, Bruno-Bárcena JM. The intestinal microbiota, gastrointestinal environment and colorectal cancer: a putative role for probiotics in prevention of colorectal cancer? Am J Physiol Gastrointest Liver Physiol 2011; 301:G401-24. [PMID: 21700901 PMCID: PMC3774253 DOI: 10.1152/ajpgi.00110.2011] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer in the United States, and, even though 5-15% of the total CRC cases can be attributed to individual genetic predisposition, environmental factors could be considered major factors in susceptibility to CRC. Lifestyle factors increasing the risks of CRC include elevated body mass index, obesity, and reduced physical activity. Additionally, a number of dietary elements have been associated with higher or lower incidence of CRC. In this context, it has been suggested that diets high in fruit and low in meat might have a protective effect, reducing the incidence of colorectal adenomas by modulating the composition of the normal nonpathogenic commensal microbiota. In addition, it has been demonstrated that changes in abundance of taxonomic groups have a profound impact on the gastrointestinal physiology, and an increasing number of studies are proposing that the microbiota mediates the generation of dietary factors triggering colon cancer. High-throughput sequencing and molecular taxonomic technologies are rapidly filling the knowledge gaps left by conventional microbiology techniques to obtain a comprehensive catalog of the human intestinal microbiota and their associated metabolic repertoire. The information provided by these studies will be essential to identify agents capable of modulating the massive amount of gut bacteria in safe noninvasive manners to prevent CRC. Probiotics, defined as "live microorganisms which, when administered in adequate amounts, confer a health benefit on the host" (219), are capable of transient modulation of the microbiota, and their beneficial effects include reinforcement of the natural defense mechanisms and protection against gastrointestinal disorders. Probiotics have been successfully used to manage infant diarrhea, food allergies, and inflammatory bowel disease; hence, the purpose of this review was to examine probiotic metabolic activities that may have an effect on the prevention of CRC by scavenging toxic compounds or preventing their generation in situ. Additionally, a brief consideration is given to safety evaluation and production methods in the context of probiotics efficacy.
Collapse
Affiliation(s)
- M. Andrea Azcárate-Peril
- 1Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill;
| | - Michael Sikes
- 2Department of Microbiology, North Carolina State University, Raleigh, North Carolina
| | - José M. Bruno-Bárcena
- 2Department of Microbiology, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
11
|
Will OCC, Deheragoda M, Phillips RKS, Clark SK, Tomlinson IPM. The role of cell proliferation and crypt fission in adenoma aggressiveness: a comparison of ileoanal pouch and rectal adenomas in familial adenomatous polyposis. Colorectal Dis 2011; 13:387-392. [PMID: 20002690 PMCID: PMC4610058 DOI: 10.1111/j.1463-1318.2009.02159.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM In patients with familial adenomatous polyposis (FAP), ileoanal pouch cancer is rare whereas rectal cancer is common, despite polyp initiation at the two sites being similar at the molecular level. This study investigated whether the disparity in adenoma aggressiveness reflects underlying differences in histogenesis. METHOD Normal mucosal biopsies and 2-3 mm adenomas from patients with FAP were dissected into individual crypts. Crypt area, morphology, fission and mitoses were analysed for crypts from pouch, rectum and supra-anastomotic ileum. Immunohistochemistry of similar archival samples was performed for lysozyme, β-catenin and TP53 expression. RESULTS The morphology of normal crypts was similar at each site, although crypt area differed. The area of normal pouch crypts was intermediate between rectum and ileum. The area of adenomatous crypts of rectum and pouch was similar, but the latter had increased asymmetrical fission. Crypt mitoses were proportional to area in all tissues, but crypt fission was reduced in adenomatous crypts from the rectum compared with the pouch. Pouch adenomas retained lysozyme expression as seen in normal ileum. Nuclear β-catenin accumulation was similar, but TP53 expression was increased in rectal adenomas. CONCLUSION Diminutive polyps from rectum and pouch differ in morphology and proliferation. Aggressiveness in rectal polyps is not conferred by increased crypt proliferation, fission, or activation of the Wnt signalling pathway. Increased TP53 expression suggests other molecular mechanisms may be responsible. While crypt mitoses are proportional to crypt area, the threshold for fission may be site specific, indicating that tissue origin may influence histogenesis and thus malignant potential.
Collapse
Affiliation(s)
- O. C. C. Will
- The Polyposis Registry, St Mark’s Hospital, Harrow, UK
,London Research Institute, Cancer Research UK, London, UK
| | - M. Deheragoda
- London Research Institute, Cancer Research UK, London, UK
,Department of Histopathology, University College Hospital, London, UK
| | | | - S. K. Clark
- The Polyposis Registry, St Mark’s Hospital, Harrow, UK
| | | |
Collapse
|
12
|
Kress E, Skah S, Sirakov M, Nadjar J, Gadot N, Scoazec JY, Samarut J, Plateroti M. Cooperation between the thyroid hormone receptor TRalpha1 and the WNT pathway in the induction of intestinal tumorigenesis. Gastroenterology 2010; 138:1863-74. [PMID: 20114049 DOI: 10.1053/j.gastro.2010.01.041] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 12/30/2009] [Accepted: 01/21/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Colorectal tumorigenesis is a multistep process involving the alteration of oncogenes and tumor suppressor genes, leading to the deregulation of molecular pathways that govern intestinal homeostasis. We have previously shown that the thyroid hormone receptor alpha1 (TRalpha1) controls intestinal development and homeostasis through the WNT pathway. More precisely, TRalpha1 directly enhances the transcription of several components of this pathway, allowing increased expression of beta-catenin/Tcf4 target genes and stimulation of cell proliferation. Because the WNT pathway is a major player in controlling intestinal homeostasis, we addressed whether the TRalpha1 receptor has tumor-inducing potential. METHODS We generated mice overexpressing TRalpha1 specifically in the intestinal epithelium in a wild-type (vil-TRalpha1) or a WNT-activated (vil-TRalpha1/Apc(+/1638N)) genetic background. RESULTS The intestine of vil-TRalpha1 mice presents aberrant intestinal mucosal architecture and increased cell proliferation and develops adenoma at a low rate. However, TRalpha1 overexpression is unable to induce cancer development. On the contrary, we observed accelerated tumorigenesis in vil-TRalpha1/Apc(+/1638N) mice compared with the Apc(+/1638N) mutants. CONCLUSION Our results suggest that this phenotype is due to cooperation between the activated TRalpha1 and WNT pathways. This is the first report describing the tumor-inducing function of TRalpha1 in the intestine.
Collapse
Affiliation(s)
- Elsa Kress
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Upper gastrointestinal carcinogenesis: H. pylori and stem cell cross-talk. J Surg Res 2010; 166:255-64. [PMID: 20452613 DOI: 10.1016/j.jss.2010.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/29/2010] [Accepted: 02/09/2010] [Indexed: 02/06/2023]
Abstract
Chronic inflammation of the gastric epithelium has been associated with the pathogenesis of gastric cancer, as it was postulated by Corea's model of gastric carcinogenesis. Helicobacter pylori (Hp) regulates this inflammatory process and promotes gastric carcinogenesis through induction of gene mutations and protein modulation. Recent data raise the cancer stem cell hypothesis, which implies a central role of multipotent cancer cells in oncogenesis of various solid tumors. This review provides a synopsis of gastric cancer initiation and promotion through Hp and stem cell signaling pathways. The expanding research field of Hp-related cancer stem cell biology may offer novel implications for future treatment of upper gastrointestinal cancer.
Collapse
|
14
|
Vermeulen L, Sprick MR, Kemper K, Stassi G, Medema JP. Cancer stem cells--old concepts, new insights. Cell Death Differ 2008; 15:947-58. [PMID: 18259194 DOI: 10.1038/cdd.2008.20] [Citation(s) in RCA: 251] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cancer has long been viewed as an exclusively genetic disorder. The model of carcinogenesis, postulated by Nowell and Vogelstein, describes the formation of a tumor by the sequential accumulation of mutations in oncogenes and tumor suppressor genes. In this model, tumors are thought to consist of a heterogeneous population of cells that continue to acquire new mutations, resulting in a highly dynamic process, with clones that out compete others due to increased proliferative or survival capacity. However, novel insights in cancer stem cell research suggest another layer of complexity in the process of malignant transformation and preservation. It has been reported that only a small fraction of the cancer cells in a malignancy have the capacity to propagate the tumor upon transplantation into immuno-compromised mice. Those cells are termed 'cancer stem cells' (CSC) and can be selected based on the expression of cell surface markers associated with immature cell types. In this review, we will critically discuss these novel insights in CSC-related research. Where possible we integrate these results within the genetic model of cancer and illustrate that the CSC model can be considered an extension of the classic genetic model rather than a contradictory theory. Finally, we discuss some of the most controversial issues in this field.
Collapse
Affiliation(s)
- L Vermeulen
- LEXOR (Laboratory for Experimental Oncology and Radiobiology), Center for Experimental Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
15
|
Abstract
The obesity is the second most frequent cause of death which can be prevented. It elevates the risk of cardiovascular diseases, diabetes mellitus type 2, cancers and premature mortality. Overweight and obesity responsible for 14% of cancer caused death in males, and 20% in females, respectively. Authors review the connection between obesity, metabolic syndrome and related metabolic alterations with colorectal cancers. They summarize the role of inflammation, hyperinsulinemia, insulin-like growth factor-I and adipokines in the colorectal carcinogenesis.
Collapse
Affiliation(s)
- Krisztina Hagymási
- Semmelweis Egyetem, Altalános Orvostudományi Kar II. Belgyógyászati Klinika, Magyar Tudományos Akadémia Molekuláris Medicina Kutatócsoport Budapest Szentkirályi u. 46. 1088.
| | | |
Collapse
|
16
|
Hagymási K, Molnár B, Sipos F, Galamb O, Tulassay Z. [Stem cell theory of colorectal cancer and its connection with molecular-biological data]. Orv Hetil 2007; 148:779-785. [PMID: 17452307 DOI: 10.1556/oh.2007.28071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Colorectal cancer is the second leading cause of death in Hungary as well as in the developed countries. The high cancer prevalence of the gastrointestinal tract is the result of the rapid turnover of epithelial cells and exposure to dietary toxins. Adult stem cells are in the lime-light of the medicine. The adult stem cells and tumor cells resemble to each other on the basis of their properties, like self-renewal and proliferation. Cancer is believed to be a disease of stem cells. Recent years have seen major advances in our understanding of location (niche), life cycle, regulation (Wnt/beta-catenin signaling pathway) and markers (mathusashi-1, beta-catenin) of gastrointestinal stem cells. The exact role of adult stem cells in intestinal carcinogenesis is open for debate. New works suggest the role for inflammation-induced engraftment of circulating marrow-derived stem cells in colorectal carcinogenesis. The causes of malignant transformation of local or engrafting bone marrow-derived stem cells are mutations (APC, MMR genes) or methylation (CDKN2A, p16/INK4a, MGMT, MLH1). The spread of dysplastic cells (bottom-up, top-down hypothesis) is also ambiguous.
Collapse
Affiliation(s)
- Krisztina Hagymási
- Semmelweis Egyetem, Altalános Orvostudományi Kar II. Belgyógyászati Klinika, Budapest.
| | | | | | | | | |
Collapse
|
17
|
Ward EJ, Shcherbata HR, Reynolds SH, Fischer KA, Hatfield SD, Ruohola-Baker H. Stem cells signal to the niche through the Notch pathway in the Drosophila ovary. Curr Biol 2006; 16:2352-8. [PMID: 17070683 DOI: 10.1016/j.cub.2006.10.022] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 10/05/2006] [Accepted: 10/06/2006] [Indexed: 12/28/2022]
Abstract
Stem cells are maintained and retain their capacity to continue dividing because of the influence of a niche. Although niches are important to maintain "stemness" in a wide variety of tissues, control of these niches is poorly understood. The Drosophila germline stem cells (GSCs) reside in a somatic cell niche. We show that Notch activation can induce the expression of niche-cell markers even in an adult fly; overexpression of Delta in the germline, or activated Notch in the somatic cells, results in extra niche cells, up to 10-fold over the normal number. In turn, these ectopic niche cells induce ectopic GSCs. Conversely, when GCSs do not produce functional Notch ligands, Delta and Serrate, the TGF-beta pathway is not activated in the GSCs, and they differentiate and subsequently leave the niche. Importantly, clonal analysis reveals that the receiving end of the Notch pathway is required in the somatic cells. These data show that a feedback loop exists between the stem cells and niche cells. Demonstration that stem cells can contribute to niche function has far-reaching consequences for stem cell therapies and may provide insight into how cancer can spread throughout an organism via populations of cancer stem cells.
Collapse
Affiliation(s)
- Ellen J Ward
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, School of Medicine, Box 357350, 1959 NE Pacific Street, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Tumour-wide 'omics' approaches have long held sway as the approach to identifying useful therapeutic targets. This view is changing with the realization that many, if not all, cancers contain a minority population of self-renewing stem cells, the cancer stem cells, which are entirely responsible for sustaining the tumour as well as giving rise to proliferating but progressively differentiating cells that are responsible for much of the cellular heterogeneity that is so familiar to histopathologists. Moreover, although many tumours probably have their origins in normal stem cells, persuasive evidence from the haematopoietic system suggests that genetic alterations in more committed progenitor cells can reactivate the self-renewal machinery, resulting in a further source of cancer stem cells. Thus, the bulk of the tumour is not the problem, and so the identification of cancer stem cells and the factors that regulate their behaviour are likely to have an enormous bearing on the way that we treat neoplastic disease in the future.
Collapse
Affiliation(s)
- J Burkert
- Histopathology Unit, Cancer Research UK, and ICMS, Queen Mary's School of Medicine and Dentistry, London WC2A 3PX, UK.
| | | | | |
Collapse
|
19
|
Bapat SA. Evolution of cancer stem cells. Semin Cancer Biol 2006; 17:204-13. [PMID: 16787749 DOI: 10.1016/j.semcancer.2006.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 05/08/2006] [Accepted: 05/09/2006] [Indexed: 12/31/2022]
Abstract
Cancer as a disease driven by cancer stem cells is a concept that has emerged over the last few years. However, several issues relating to this phenomenon as yet remain unaddressed. A fundamental question is one relating to the identification of events leading to transformation of a normal tissue stem cell to a cancer stem cell. Complete knowledge of this evolutionary process may be crucial for the development of novel effective therapies that influence patient prognosis. The scope of this review is to discuss reports that have begun to elucidate stem cell transformation either as an isolated event or as a progression as an attempt towards understanding some of the critical events involved in the process.
Collapse
Affiliation(s)
- S A Bapat
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| |
Collapse
|