1
|
Garrett C, Steffens D, Ackland S, Solomon M, Koh C. Risk factors, histopathological landscape, biomarkers, treatment patterns and survival of early-onset colorectal cancer: A narrative review. Asia Pac J Clin Oncol 2024; 20:444-449. [PMID: 38776256 DOI: 10.1111/ajco.14081] [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: 11/14/2023] [Accepted: 05/07/2024] [Indexed: 05/24/2024]
Abstract
Early-onset colorectal cancer (EOCRC) incidence has increased in most Western countries over the last decade, with Australia at the forefront. Recent literature has thus focused on characterizing EOCRC from later-onset colorectal cancer (LOCRC). Earlier exposure to modifiable risk factors resulting in gut dysbiosis has been linked with EOCRC development. EOCRCs have more aggressive histopathological features with somatic mutations resulting in pro-inflammatory tumor microenvironments. There is a tendency to treat EOCRCs with multimodal chemotherapeutic regimens and more extensive surgery than LOCRCs with conflicting postoperative outcomes and survival data. Current research is limited by a lack of Australasian studies, retrospective study designs, and heterogeneous definitions of EOCRC. Future research should address these and focus on investigating the role of immunotherapies, establishing minimally invasive diagnostic biomarkers and nomograms, and evaluating the survival and functional outcomes of EOCRC.
Collapse
Affiliation(s)
- Celine Garrett
- Surgical Outcomes Research Centre, Royal Prince Alfred Hospital, Camperdown, Australia
- Faculty of Medicine & Health, Central Clinical School, The University of Sydney, Camperdown, Australia
- Faculty of Medicine & Health, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Daniel Steffens
- Surgical Outcomes Research Centre, Royal Prince Alfred Hospital, Camperdown, Australia
- Faculty of Medicine & Health, Central Clinical School, The University of Sydney, Camperdown, Australia
| | - Stephen Ackland
- Faculty of Health, University of Newcastle, Callaghan, Australia
| | - Michael Solomon
- Surgical Outcomes Research Centre, Royal Prince Alfred Hospital, Camperdown, Australia
- Faculty of Medicine & Health, Central Clinical School, The University of Sydney, Camperdown, Australia
| | - Cherry Koh
- Surgical Outcomes Research Centre, Royal Prince Alfred Hospital, Camperdown, Australia
- Faculty of Medicine & Health, Central Clinical School, The University of Sydney, Camperdown, Australia
| |
Collapse
|
2
|
Cui G, Li G, Pang Z, Florholmen J, Goll R. The presentation and regulation of the IL-8 network in the epithelial cancer stem-like cell niche in patients with colorectal cancer. Biomed Pharmacother 2022; 152:113252. [PMID: 35687912 DOI: 10.1016/j.biopha.2022.113252] [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: 04/12/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Accumulative evidence suggests that the biological behavior of cancer stem-like cells (CSCs) is regulated by their surrounding niche, in which cytokines function as one of the main mediators for the interaction between CSCs and their microenvironment in the colorectal cancer (CRC). METHODS We characterized the presentation of CSCs and the interleukin (IL)- 8 network in the adenoma/CRC epithelium using quantitative real-time PCR (q-PCR), immunohistochemistry (IHC) and double immunofluorescence. In addition, the capacity of IL-1β to stimulate epithelial IL-8 production in colon cancer Caco-2 cells was examined in vitro and the IL-8 product was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS IHC observation showed increased expression of both CSCs and IL-8 in the adenoma and CRC epithelium, and q-PCR results revealed that increased expression of IL-1β transcript was strongly correlated with increased IL-8 transcript levels in both adenoma and CRC tissues. Double immunofluorescence images demonstrated the coexpression of the IL-8 receptors IL-8RA and IL-8RB with LGR5 labeled CSCs in CRC tissue sections. Consistently, in vitro experiments showed that coculture of Caco-2 cells with IL-1β at concentrations of 1, 5, 10 and 20 ng/ml resulted in a dose-dependent release of IL-8, which could be specifically inhibited by cotreatment with the IL-1β receptor antagonist. CONCLUSIONS These results demonstrate activation of the IL-8 network in the niche of CSCs from the precancerous adenoma stage to the CRC stage, which is potentially stimulated by IL-1β in CRC cells.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Faculty of Health Science, Nord University, Campus Levanger, Levanger, Norway.
| | - Gui Li
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Pang
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jon Florholmen
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| | - Rasmus Goll
- Department of Gastroenterology, University Hospital of North Norway, University of Tromsø, Tromsø, Norway
| |
Collapse
|
3
|
Canizalez-Roman A, Reina-Reyes JE, Angulo-Zamudio UA, Geminiano-Martínez EE, Flores-Carrillo AF, García-Matus RR, Valencia-Mijares NM, Leon-Sicairos N, Velazquez-Roman J, Martínez-Villa FA, Tapia-Pastrana G. Prevalence of Cyclomodulin-Positive E. coli and Klebsiella spp. Strains in Mexican Patients with Colon Diseases and Antimicrobial Resistance. Pathogens 2021; 11:pathogens11010014. [PMID: 35055962 PMCID: PMC8779131 DOI: 10.3390/pathogens11010014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Colon diseases, such as colorectal cancer (CRC), are multifactor diseases that affect more than one million people per year; recently, the microbiota has been associated with an etiologic factor, specifically bacterial cyclomodulin positivity (CM+). Unfortunately, there are no studies from Mexico that detail the presence of bacterial CM+ in patients with colon diseases. We therefore performed a comprehensive study to investigate the associations and prevalence of cyclomodulin-positive Diarrheagenic E. coli (DEC), non-DEC, and Klebsiella spp. strains isolated from Mexican subjects with colon diseases. In this work, we analyzed 43 biopsies, 87 different bacteria were isolated, and E. coli was the most frequently noted, followed by Klebsiella spp., and Enterococcus spp. E. coli, non-DEC, and EPEC belonging to phylogroup B2 were the most prevalent. More than 80% of E. coli and Klebsiella were CM+. pks, cdt, cnf, and cif were identified. cdt was associated with non-DEC, cif and its combinations with EPEC, as well as cdt and psk with Klebsiella. Lastly, all the CM+ bacteria were resistant to at least one antibiotic (34% were MDR, and 48% XDR). In conclusion, the high prevalence of bacterial CM+ in colon disease patients suggests that these bacteria play an important role in the genesis of these diseases.
Collapse
Affiliation(s)
- Adrian Canizalez-Roman
- Centro de Investigación Aplicada a la Salud Pública (CIASaP), School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80246, Mexico; (A.C.-R.); (U.A.A.-Z.); (N.L.-S.); (J.V.-R.)
- The Women’s Hospital, Secretariat of Health, Culiacan Sinaloa 80127, Mexico
| | - Juan E. Reina-Reyes
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca City 68120, Mexico
| | - Uriel A. Angulo-Zamudio
- Centro de Investigación Aplicada a la Salud Pública (CIASaP), School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80246, Mexico; (A.C.-R.); (U.A.A.-Z.); (N.L.-S.); (J.V.-R.)
| | - Eloy E. Geminiano-Martínez
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
| | - Antonio F. Flores-Carrillo
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
| | - Rolando R. García-Matus
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
| | - Norma M. Valencia-Mijares
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
| | - Nidia Leon-Sicairos
- Centro de Investigación Aplicada a la Salud Pública (CIASaP), School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80246, Mexico; (A.C.-R.); (U.A.A.-Z.); (N.L.-S.); (J.V.-R.)
- Pediatric Hospital of Sinaloa, Constitución 530, Jorge Almada, Culiacan Sinaloa 80200, Mexico
| | - Jorge Velazquez-Roman
- Centro de Investigación Aplicada a la Salud Pública (CIASaP), School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80246, Mexico; (A.C.-R.); (U.A.A.-Z.); (N.L.-S.); (J.V.-R.)
| | - Francisco A. Martínez-Villa
- Programa de Maestría en Ciencias en Biomedicina Molecular, Facultad de Medicina, UAS, Culiacan Sinaloa 80246, Mexico;
| | - Gabriela Tapia-Pastrana
- Laboratorio de Investigación Biomédica, Hospital Regional de Alta Especialidad de Oaxaca, San Bartolo Coyotepec, Oaxaca City 71256, Mexico; (J.E.R.-R.); (E.E.G.-M.); (A.F.F.-C.); (R.R.G.-M.); (N.M.V.-M.)
- Correspondence: ; Tel.: +52-0195150180 (ext. 1230); Fax: +52-0195150152
| |
Collapse
|
4
|
Bert M, Devilliers H, Orry D, Rat P, Facy O, Ortega-Deballon P. Preoperative inflammation is an independent factor of worse prognosis after colorectal cancer surgery. J Visc Surg 2021; 158:305-311. [PMID: 33446466 DOI: 10.1016/j.jviscsurg.2020.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND We know that inflammation is related to colorectal cancer prognosis and to the onset of postoperative infections. OBJECTIVE This study aimed to understand the relationship between preoperative inflammation and the prognosis of colorectal cancer and to elucidate whether the impact of inflammation on cancer prognosis was related to an increased risk of surgical infection or was independent of it. METHODS Patients who underwent elective colorectal cancer surgery between November 2011 and April 2014 were included in a prospective database (IMACORS). Preoperative c reactive protein was collected for each patient. Patients were followed up according to the French national guidelines. A cut-off of preoperative CRP of 5mg/L was chosen. Clinical characteristics were compared according to CRP using Chi2 and Mann-Whitney tests. The Overall Survival (OS) and Disease-Free-Survival (DFS) were compared by Kaplan-Meier curves. A Cox proportional hazards regression model was applied to perform a multivariate analysis of OS and DFS's predictors. RESULTS A total of 254 patients were included. The median age was 68 years old. The median follow up was 41.8 months. The overall median preoperative CRP was 5mg/L. Preoperative CRP was significantly associated with N status; CRP being significantly higher among patients with colonic cancer and with patients who didn't receive a neoadjuvant treatment. Multivariate analyse revealed that preoperative CRP is an independent prognostic factor of OS and DFS respectively (HR=2.34 (1.26-4.31), P=0.006 and HR=1.83 (1.15-2.90), P=0.01). CONCLUSION Preoperative inflammation measured by CRP is independently related with overall and disease-free survival of colorectal cancer.
Collapse
Affiliation(s)
- M Bert
- Department of Digestive Surgical Oncology, University Hospital and School of Medicine, CHU Dijon Bourgogne, 14, rue Paul-Gaffarel, 21079 Dijon cedex, France
| | - H Devilliers
- INSERM CIC-EC 1432 Clinical Investigation, Clinical Epidemiology Unit, Dijon University Hospital, Dijon, France; Department of Internal medicine and systemic disease, Dijon University Hospital, Dijon, France
| | - D Orry
- Department of Surgical Oncology, Georges-François Leclerc Anticancer Center, Dijon, France
| | - P Rat
- Department of Digestive Surgical Oncology, University Hospital and School of Medicine, CHU Dijon Bourgogne, 14, rue Paul-Gaffarel, 21079 Dijon cedex, France; INSERM Unit 1231, Locoregional therapy in surgical oncology, Dijon, France
| | - O Facy
- Department of Digestive Surgical Oncology, University Hospital and School of Medicine, CHU Dijon Bourgogne, 14, rue Paul-Gaffarel, 21079 Dijon cedex, France; INSERM Unit 1231, Locoregional therapy in surgical oncology, Dijon, France
| | - P Ortega-Deballon
- Department of Digestive Surgical Oncology, University Hospital and School of Medicine, CHU Dijon Bourgogne, 14, rue Paul-Gaffarel, 21079 Dijon cedex, France; INSERM Unit 1231, Locoregional therapy in surgical oncology, Dijon, France.
| |
Collapse
|
5
|
Dun Y, Chen J, Liu J, Guo Y, Zhang C, Yuan D. Changes of Wnt/β-catenin signalling, BMP2, and BMP4 in the jejunum during ageing in rats. Arab J Gastroenterol 2020; 21:43-48. [PMID: 32241700 DOI: 10.1016/j.ajg.2019.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/19/2019] [Accepted: 12/22/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND STUDY AIMS The renewal of intestinal epithelium is maintained by intestinal stem cells (ISCs). Studies have found an age-dependent increase of Esg+/Dl+ progenitor cells in the midgut of Drosophila. However, changes of ISCs and the molecular regulation in mammalian animals with age are yet unknown. The aim of this study was to find out the changes of ISCs and molecular regulation in mammalian animals during the process of ageing. MATERIAL AND METHODS Thirty Sprague-Dawley rats were divided into three groups: young (3 months old), adult (6 months old), and ageing (24 months old). Levels of PCNA, Bmi1, β-catenin and BMP4 were examined by Immunohistochemistry staining. Levels of Bmi1, GSK-3β, Dkk1 and BMP2 were determined by Western Blot. RESULTS Our results showed that the proliferation of ISCs was decreased and the number of intestinal stem cells declined in ageing rats. The niches of ISCs, including Wnt signalling pathway and some proteins of Bone morphogenetic protein (BMP) signalling pathway, were downregulated in the jejunum of ageing rats. CONCLUSION Our study indicated that age-related decreased proliferation of intestinal stem cells in the jejunum could be associated with the alleviation of niches, including Wnt signalling pathway and some proteins of BMP signalling pathway.
Collapse
Affiliation(s)
- Yaoyan Dun
- Medical College of China Three Gorges University, Yichang 443002, China
| | - Jing Chen
- Medical College of China Three Gorges University, Yichang 443002, China
| | - Jie Liu
- Medical College of China Three Gorges University, Yichang 443002, China
| | - Yuhui Guo
- Medical College of China Three Gorges University, Yichang 443002, China
| | - Changcheng Zhang
- Medical College of China Three Gorges University, Yichang 443002, China.
| | - Ding Yuan
- Medical College of China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
6
|
Switching to a Healthy Diet Prevents the Detrimental Effects of Western Diet in a Colitis-Associated Colorectal Cancer Model. Nutrients 2019; 12:nu12010045. [PMID: 31877961 PMCID: PMC7019913 DOI: 10.3390/nu12010045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 01/19/2023] Open
Abstract
Inflammatory bowel disease increases the odds of developing colitis-associated cancer. We hypothesized that Western-style diet (WD) aggravates azoxymethane (AOM)/dextran sulfate sodium salt (DSS)-induced colitis-associated tumorigenesis and that switching to the standard AIN93G diet will ameliorate disease symptoms even after cancer initiation. Female BALB/c mice received either WD (WD group) or standard AIN93G diet (AIN group) for the whole experimental period. After five weeks, the mice received 12.5 mg/kg AOM intraperitoneally, followed by three DSS cycles. In one group of mice, the WD was switched to AIN93G the day before starting the first DSS cycle (WD/AIN group). Feeding the WD during the whole experimental period aggravated colitis symptoms, shortened the colon (p < 0.05), changed microbiota composition and increased tumor promotion. On molecular level, the WD reduced proliferation (p < 0.05) and increased expression of the vitamin D catabolizing enzyme Cyp24a1 (p < 0.001). The switch to the AIN93G diet ameliorated this effect, reflected by longer colons, fewer (p < 0.05) and smaller (p < 0.01) aberrant colonic crypt foci, comparable with the AIN group. Our results show that switching to a healthy diet, even after cancer initiation is able to revert the deleterious effect of the WD and could be an effective preventive strategy to reduce colitis symptoms and prevent tumorigenesis.
Collapse
|
7
|
Yu H, Feng C, Wang Z, Li J, Wang Y, Hu X, Li Z, Shen Y, Hu D. Potential of diffusion-weighted imaging in magnetic resonance enterography to identify neoplasms in the ileocecal region: Use of ultra-high b-value diffusion-weighted imaging. Oncol Lett 2019; 18:1451-1457. [PMID: 31423210 DOI: 10.3892/ol.2019.10441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 05/13/2019] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic inflammatory bowel disease have an increased risk of colorectal cancer, and the differentiation between neoplastic and inflammatory lesions often poses a clinical dilemma. The aim of the present study was to investigate whether diffusion-weighted (DW) magnetic resonance (MR) enterography with ultra-high b-value facilitates the identification of neoplastic lesions in the ileocecal region. A total of 76 patients (22 patients with neoplasms, 26 inflammatory lesions and 28 normal subjects) from 292 cases of suspected bowel disorders were included in the present study. All patients were examined with conventional MR enterography and DW imaging (DWI) with seven different b-values (400, 600, 800, 1,000, 1,200, 1,500 and 3,000 sec/mm2) in a 3T MR scanner. DWI scans with different b-values were analyzed independently by two radiologists for the presence of ileocecal lesions. The signal intensity of the majority of inflammatory lesions and normal bowel segments gradually decreased to the background intensity with increasing b-values; however, neoplastic lesions demonstrated relative hyperintensity compared with the background. In addition, ~76% of the positive findings from b=3,000 sec/mm2 DWI were neoplasms. In conclusion, a lesion with consistently high signal intensity from DWI images with b-values increasing to 3,000 sec/mm2 indicated the presence of neoplasms. The results suggested that ultra-high b-value (3,000 sec/mm2) imaging may aid the clinical differentiation of neoplasms from benign conditions.
Collapse
Affiliation(s)
- Hao Yu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Cui Feng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zi Wang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jianjun Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yanchun Wang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xuemei Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yaqi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Daoyu Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
8
|
Bonfini A, Liu X, Buchon N. From pathogens to microbiota: How Drosophila intestinal stem cells react to gut microbes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:22-38. [PMID: 26855015 DOI: 10.1016/j.dci.2016.02.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 06/05/2023]
Abstract
The intestine acts as one of the interfaces between an organism and its external environment. As the primary digestive organ, it is constantly exposed to a multitude of stresses as it processes and absorbs nutrients. Among these is the recurring damage induced by ingested pathogenic and commensal microorganisms. Both the bacterial activity and immune response itself can result in the loss of epithelial cells, which subsequently requires replacement. In the Drosophila midgut, this regenerative role is fulfilled by intestinal stem cells (ISCs). Microbes not only trigger cell loss and replacement, but also modify intestinal and whole organism physiology, thus modulating ISC activity. Regulation of ISCs is integrated through a complex network of signaling pathways initiated by other gut cell populations, including enterocytes, enteroblasts, enteroendocrine and visceral muscles cells. The gut also receives signals from circulating immune cells, the hemocytes, to properly respond against infection. This review summarizes the types of gut microbes found in Drosophila, mechanisms for their elimination, and provides an integrated view of the signaling pathways that regulate tissue renewal in the midgut.
Collapse
Affiliation(s)
| | - Xi Liu
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
9
|
Chen JX, Wang H, Liu A, Zhang L, Reuhl K, Yang CS. From the Cover: PhIP/DSS-Induced Colon Carcinogenesis in CYP1A-Humanized Mice and the Possible Role of Lgr5+ Stem Cells. Toxicol Sci 2016; 155:224-233. [PMID: 27664423 DOI: 10.1093/toxsci/kfw190] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In the past decades, experimental rodent models developed to study the pathogenesis of human colorectal cancer (CRC) generally employed synthetic chemical carcinogens or genetic manipulation. Our lab, in order to establish a more physiologically relevant CRC model, recently developed a colon carcinogenesis model induced by the meat-derived dietary carcinogen, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), and promoted by dextran sodium sulfate (DSS)-induced colitis in the cytochrome P450 1A-humanized (hCYP1A) mice. The resulting colon tumors shared many histologic and molecular features of human colon cancer. In this study, we characterized the early stages of PhIP/DSS-induced colon carcinogenesis. We found that PhIP/DSS treatments caused rapid destruction of the colon mucosa with severe inflammation, followed by the presence of reactive changes and low-grade dysplastic lesions, and then manifestation of high-grade dysplastic lesions and finally adenocarcinomas. Molecular analysis of the early time-points (ie, days 1, 3, 7, 11, 14, and 21 after DSS exposure) indicates Ctnnb1/β-catenin mutations and β-catenin nuclear accumulation in the high-grade dysplastic lesions, but not low-grade dysplastic lesions or adjacent normal tissues. In addition, we investigated the role of Lgr5+ colon stem cells in the PhIP/DSS-induced colon carcinogenesis and found the presence of Lgr5-enhance green fluorescent protein-expressing cells amidst some ulcerated mucosa, high-grade dysplastic lesions and adenocarcinomas, suggesting a possible role of Lgr5+ stem cells in this dietary carcinogen-induced, inflammation-promoted colon carcinogenesis model. Overall, the findings suggest that PhIP/DSS-induced colon carcinogenesis is likely initiated by dominant active Ctnnb1/β-catenin mutation in residual epithelial cells, which when promoted by colitis, developed into high-grade dysplasia and adenocarcinoma.
Collapse
Affiliation(s)
- Jayson X Chen
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey.,Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Hong Wang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Anna Liu
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, New Jersey
| | - Kenneth Reuhl
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey;
| |
Collapse
|
10
|
Wang D, Lin Y, Gao B, Yan S, Wu H, Li Y, Wu Q, Wei Y. Reduced Expression of FADS1 Predicts Worse Prognosis in Non-Small-Cell Lung Cancer. J Cancer 2016; 7:1226-32. [PMID: 27390597 PMCID: PMC4934030 DOI: 10.7150/jca.15403] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/26/2016] [Indexed: 12/19/2022] Open
Abstract
Objective: Fatty acid desaturase 1 is a member of the fatty acid desaturase, which is related to a number of diseases. However, its role in cancers remains unclear. This study was to explore the clinical importance of FADS1 expression in non-small-cell lung cancer (NSCLC). Materials and Methods: Immunochemistry was used to evaluate FADS1 expressions in 216 paraffin-embedded specimens. The expression of FADS1 was divided into high and low groups. The clinical and prognostic significance of FADS1 expression was analyzed statistically by Kaplan-Meier estimate and Cox regression model. Results: FADS1 overexpressed in normal bronchial mucosa compared with non-small-cell lung cancer. Reduced FADS1 expression was associated with tumor size (P=0.023) and histological grade (P<0.0001). Patients with lower expression of FADS1 had shorter overall survival and disease free survival (P=0.001 and P=0.002). Multivariate analysis showed FADS1 expression was an independent prognostic factor in NSCLC (P=0.011). Conclusion: Reduced expression of FADS1 suggests pessimistic prognosis for NSCLC patients. Further studies are warranted.
Collapse
Affiliation(s)
- Dong Wang
- 1. Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266000, P.R. China
| | - Yan Lin
- 2. Department of First Chemotherapy, Affiliated Cancer Hospital of Guangxi Medical University, 71 Hedi Road, Nanning 530021, P. R. China
| | - Bei Gao
- 3. State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Shumei Yan
- 4. State Key Laboratory of Oncology in South China; Department of Pathology, Sun Yat-Sen University Cancer Center; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R. China
| | - Huini Wu
- 5. University of Illinois at Chicago, Biological Science, Chicago, Illinois 60607, USA
| | - Yong Li
- 4. State Key Laboratory of Oncology in South China; Department of Pathology, Sun Yat-Sen University Cancer Center; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R. China
| | - Qiuliang Wu
- 4. State Key Laboratory of Oncology in South China; Department of Pathology, Sun Yat-Sen University Cancer Center; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R. China
| | - Yucheng Wei
- 1. Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266000, P.R. China
| |
Collapse
|
11
|
De Lerma Barbaro A. The complex liaison between cachexia and tumor burden (Review). Oncol Rep 2015; 34:1635-49. [PMID: 26239384 DOI: 10.3892/or.2015.4164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
Cachexia is a wasting syndrome that afflicts end-stage cancer patients. Whereas a consensus statement for a definition of cachexia recently has been accomplished, a useful measurement for this condition at present is lacking. The aim of the present review is to discuss the advantage of introducing the measurement of tumor burden for a better overall evaluation of cachexia. Our suggestion ensues from a somewhat novel perspective in the field of infectious disease research where a careful measurement of the pathogen load, between i.e. different host genotypes, leads to the definition of the concept of tolerance to the infectious insult. Indeed tolerance concurs, together the more classical resistance, in maintaining the host reproductive fitness or health state. Noticeably a similar reasoning may apply to tumor biology as well. Whereas the extent of cachexia increases with tumor burden, the relationship between these two correlates of tumor progression fluctuates in a broad range. We have selected from the literature studies in the rodent model where significant variation in the course of the wasting illness during cancer was observed and quantitatively assessed comparing experimental groups marked by different genotype, drug treatment, diet or gender. These studies may be further classified in two categories: the former where the experimental condition associated to milder cachexia is accompanied to a lesser tumor burden, the latter where the inhibition of cachexia results disentangled from the tumor burden, that is the whole number of cancer cells results unchanged or even, paradoxically, is increased. In addition we survey, even in the context of human malignancy, the significance and feasibility of plotting quantitative estimates of cachexia against the whole tumor burden. Ultimately, the principal endeavor of introducing the measurement of tumor burden, in both experimental and clinical oncology, may be to achieve a better assessment of the inter-individual variation in the host vulnerability to cancer cachexia.
Collapse
Affiliation(s)
- Andrea De Lerma Barbaro
- Biomedical Research Division, Department of Theoretical and Applied Sciences, University of Insubria, Busto Arsizio, Varese, Italy
| |
Collapse
|
12
|
Agarwal S, Lakoma A, Chen Z, Hicks J, Metelitsa LS, Kim ES, Shohet JM. G-CSF Promotes Neuroblastoma Tumorigenicity and Metastasis via STAT3-Dependent Cancer Stem Cell Activation. Cancer Res 2015; 75:2566-79. [PMID: 25908586 DOI: 10.1158/0008-5472.can-14-2946] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 04/11/2015] [Indexed: 12/16/2022]
Abstract
Increasing evidence suggests that inflammatory cytokines play a critical role in tumor initiation and progression. A cancer stem cell (CSC)-like subpopulation in neuroblastoma is known to be marked by expression of the G-CSF receptor (G-CSFR). Here, we report on the mechanistic contributions of the G-CSFR in neuroblastoma CSCs. Specifically, we demonstrate that the receptor ligand G-CSF selectively activates STAT3 within neuroblastoma CSC subpopulations, promoting their expansion in vitro and in vivo. Exogenous G-CSF enhances tumor growth and metastasis in human xenograft and murine neuroblastoma tumor models. In response to G-CSF, STAT3 acts in a feed-forward loop to transcriptionally activate the G-CSFR and sustain neuroblastoma CSCs. Blockade of this G-CSF-STAT3 signaling loop with either anti-G-CSF antibody or STAT3 inhibitor depleted the CSC subpopulation within tumors, driving correlated tumor growth inhibition, decreased metastasis, and increased chemosensitivity. Taken together, our results define G-CSF as a CSC-activating factor in neuroblastoma, suggest a comprehensive reevaluation of the clinical use of G-CSF in these patients to support white blood cell counts, and suggest that direct targeting of the G-CSF-STAT3 signaling represents a novel therapeutic approach for neuroblastoma.
Collapse
Affiliation(s)
- Saurabh Agarwal
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Anna Lakoma
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Zaowen Chen
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - John Hicks
- Department of Pathology, Section of Pediatric Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Leonid S Metelitsa
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Eugene S Kim
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas. Division of Pediatric Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, Houston, Texas. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
13
|
Ostan R, Lanzarini C, Pini E, Scurti M, Vianello D, Bertarelli C, Fabbri C, Izzi M, Palmas G, Biondi F, Martucci M, Bellavista E, Salvioli S, Capri M, Franceschi C, Santoro A. Inflammaging and cancer: a challenge for the Mediterranean diet. Nutrients 2015; 7:2589-621. [PMID: 25859884 PMCID: PMC4425163 DOI: 10.3390/nu7042589] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/19/2015] [Accepted: 03/24/2015] [Indexed: 12/19/2022] Open
Abstract
Aging is considered the major risk factor for cancer, one of the most important mortality causes in the western world. Inflammaging, a state of chronic, low-level systemic inflammation, is a pervasive feature of human aging. Chronic inflammation increases cancer risk and affects all cancer stages, triggering the initial genetic mutation or epigenetic mechanism, promoting cancer initiation, progression and metastatic diffusion. Thus, inflammaging is a strong candidate to connect age and cancer. A corollary of this hypothesis is that interventions aiming to decrease inflammaging should protect against cancer, as well as most/all age-related diseases. Epidemiological data are concordant in suggesting that the Mediterranean Diet (MD) decreases the risk of a variety of cancers but the underpinning mechanism(s) is (are) still unclear. Here we review data indicating that the MD (as a whole diet or single bioactive nutrients typical of the MD) modulates multiple interconnected processes involved in carcinogenesis and inflammatory response such as free radical production, NF-κB activation and expression of inflammatory mediators, and the eicosanoids pathway. Particular attention is devoted to the capability of MD to affect the balance between pro- and anti-inflammaging as well as to emerging topics such as maintenance of gut microbiota (GM) homeostasis and epigenetic modulation of oncogenesis through specific microRNAs.
Collapse
Affiliation(s)
- Rita Ostan
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Catia Lanzarini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Elisa Pini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Maria Scurti
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Dario Vianello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Claudia Bertarelli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Cristina Fabbri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Massimo Izzi
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Giustina Palmas
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Fiammetta Biondi
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Morena Martucci
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Elena Bellavista
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
- IRCCS, Institute of Neurological Sciences, Via Altura 3, 40139 Bologna, Italy.
- National Research Council of Italy, CNR, Institute for Organic Synthesis and Photoreactivity (ISOF), Via P. Gobetti 101, 40129 Bologna, Italy.
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| |
Collapse
|
14
|
Karim BO, Rhee KJ, Liu G, Yun K, Brant SR. Prom1 function in development, intestinal inflammation, and intestinal tumorigenesis. Front Oncol 2014; 4:323. [PMID: 25452936 PMCID: PMC4231842 DOI: 10.3389/fonc.2014.00323] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/28/2014] [Indexed: 12/17/2022] Open
Abstract
Prom1/CD133 has been identified in colorectal, hepatocellular, and pancreatic cancer as a cancer stem cell marker and has been used as such to predict colon cancer recurrence in humans. Its potential molecular function as well as its role as a marker of intestinal regeneration is still not fully known. We evaluated the role of Prom1 in intestinal regeneration in inflammatory bowel disease (IBD), determined the function of Prom1, and characterized the effect of a lack of Prom1 on intestinal tumor formation in animal models. Our results suggest that Apc mutations lead to an increase in Prom1 expressing cells in the intestinal crypt stem cell compartment and in early intestinal adenomas. Also, Prom1 knockout mice are more susceptible to intestinal tumor formation. We conclude that Prom1 likely plays a role in regulating intestinal homeostasis and that these results clearly illustrate the role of Prom1 in intestinal regeneration. We further conclude that Prom1 may provide a novel therapeutic target for patients with gastrointestinal conditions such as IBD, short bowel syndrome, and colorectal cancer.
Collapse
Affiliation(s)
- Baktiar O Karim
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University , Baltimore, MD , USA
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, Yonsei University , Gangwon-do, Wonju , South Korea
| | - Guosheng Liu
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University , Baltimore, MD , USA
| | - Kyuson Yun
- The Jackson Laboratory , Bar Harbor, ME , USA
| | - Steven R Brant
- Department of Medicine, The Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|