1
|
De Arrigunaga S, Wall S, Theotoka D, Friehmann A, Camacho M, Dubovy S, Galor A, Karp CL. Chronic inflammation as a proposed risk factor for ocular surface squamous neoplasia. Ocul Surf 2024; 33:1-10. [PMID: 38432640 DOI: 10.1016/j.jtos.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE Chronic inflammation is a predisposing factor for metaplastic changes and ultimately dysplasia. We describe cases of OSSN occurring in the setting of chronic ocular surface inflammation. METHODS Sixteen eyes from 14 individuals were included from one ocular oncology clinic between 2010 and 2023. Patients presented with ocular surface squamous neoplasia (OSSN) in the setting of chronic inflammation. The diagnosis of OSSN was made using anterior segment high-resolution optical coherence tomography (HR-OCT) and confirmed by histopathological analysis in all cases. RESULTS Median age on presentation was 61 [IQR 47.5-69.2] years. Eleven (86%) individuals were male and five (36%) identified as White Hispanic. Ten eyes were referred with ocular surface diagnoses including pannus (n = 4), scarring (n = 3), pterygium (n = 2), and herpetic keratitis (n = 1). Only six eyes were referred as possible neoplasia. All individuals had a history of ocular surface inflammation. The most common inflammatory conditions were ocular rosacea (seven individuals) and atopic keratoconjunctivitis (AKC) (five individuals). Two individuals were found to have bilateral OSSN, one in the setting of ocular rosacea and the other in the setting of AKC. All 16 eyes from 14 individuals were suspected to have OSSN based on HR-OCT findings which guided the location of the incisional biopsies that subsequently confirmed histopathological diagnosis in all cases. CONCLUSION OSSN may arise in the setting of chronic inflammation on the ocular surface. Identification of the tumor can be challenging in these cases, and HR-OCT can be a key diagnostic tool in detecting OSSN.
Collapse
Affiliation(s)
- Sofia De Arrigunaga
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA
| | - Sarah Wall
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology and Visual Science, Yale School of Medicine, 40 Temple Street, New Haven, CT, 06510, USA
| | - Despoina Theotoka
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology and Visual Science, Yale School of Medicine, 40 Temple Street, New Haven, CT, 06510, USA
| | - Asaf Friehmann
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology, Meir Medical Center, Kfar Saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Israel
| | - Matthew Camacho
- Florida Lions Ocular Pathology Laboratory, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sander Dubovy
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Florida Lions Ocular Pathology Laboratory, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology, Miami Veterans Administration Medical Center, Miami, FL, USA
| | - Carol L Karp
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA.
| |
Collapse
|
2
|
Karvela A, Veloudiou OZ, Karachaliou A, Kloukina T, Gomatou G, Kotteas E. Lung microbiome: an emerging player in lung cancer pathogenesis and progression. Clin Transl Oncol 2023:10.1007/s12094-023-03139-z. [PMID: 36995519 DOI: 10.1007/s12094-023-03139-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/26/2023] [Indexed: 03/31/2023]
Abstract
The microbiome of the lungs, although until recently neglected, is now emerging as a potential contributor to chronic lung diseases, including cancer. Preclinical evidence suggests that the microbial burden of the lungs shapes the host immunity mechanisms and affects local antitumor immune responses. Studies of cohorts of patients with lung cancer reveal that different microbiome profiles are detected in patients with lung cancer compared to controls. In addition, an association between differential lung microbiome composition and distinct responses to immunotherapy has been suggested, yet, with limited data. Scarce evidence exists on the role of the lung microbiome in the development of metastases in the lungs. Interestingly, the lung microbiome is not isolated and interacts with the gut microbiome through a dynamic axis. Future research on the involvement of the lung microbiome in lung cancer pathogenesis and potential therapeutic implications is greatly anticipated.
Collapse
Affiliation(s)
- Alexandra Karvela
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece
| | - Orsalia-Zoi Veloudiou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece
| | - Anastasia Karachaliou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece
| | - Theoni Kloukina
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece
| | - Georgia Gomatou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece.
| | - Elias Kotteas
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Messogion Ave 152, 11527, Athens, Greece
| |
Collapse
|
3
|
Haque S, Raina R, Afroze N, Hussain A, Alsulimani A, Singh V, Mishra BN, Kaul S, Kharwar RN. Microbial dysbiosis and epigenetics modulation in cancer development - A chemopreventive approach. Semin Cancer Biol 2022; 86:666-681. [PMID: 34216789 DOI: 10.1016/j.semcancer.2021.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/27/2023]
Abstract
An overwhelming number of research articles have reported a strong relationship of the microbiome with cancer. Microbes have been observed more commonly in the body fluids like urine, stool, mucus of people with cancer compared to the healthy controls. The microbiota is responsible for both progression and suppression activities of various diseases. Thus, to maintain healthy human physiology, host and microbiota relationship should be in a balanced state. Any disturbance in this equilibrium, referred as microbiome dysbiosis becomes a prime cause for the human body to become more prone to immunodeficiency and cancer. It is well established that some of these microbes are the causative agents, whereas others may encourage the formation of tumours, but very little is known about how these microbial communications causing change at gene and epigenome level and trigger as well as encourage the tumour growth. Various studies have reported that microbes in the gut influence DNA methylation, DNA repair and DNA damage. The genes and pathways that are altered by gut microbes are also associated with cancer advancement, predominantly those implicated in cell growth and cell signalling pathways. This study exhaustively reviews the current research advancements in understanding of dysbiosis linked with colon, lung, ovarian, breast cancers and insights into the potential molecular targets of the microbiome promoting carcinogenesis, the epigenetic alterations of various potential targets by altered microbiota, as well as the role of various chemopreventive agents for timely prevention and customized treatment against various types of cancers.
Collapse
Affiliation(s)
- Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia; Bursa Uludağ University Faculty of Medicine, Görükle Campus, 16059, Nilüfer, Bursa, Turkey
| | - Ritu Raina
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Nazia Afroze
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates.
| | - Ahmad Alsulimani
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Vineeta Singh
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow, 226021, Uttar Pradesh, India
| | - Bhartendu Nath Mishra
- Department of Biotechnology, Institute of Engineering and Technology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow, 226021, Uttar Pradesh, India
| | - Sanjana Kaul
- School of Biotechnology, University of Jammu, Jammu, 180006, J&K, India
| | - Ravindra Nath Kharwar
- Centre of Advanced Study in Botany, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
4
|
Ganduri V, Rajasekaran K, Duraiyarasan S, Adefuye MA, Manjunatha N. Colorectal Carcinoma, Cyclooxygenases, and COX Inhibitors. Cureus 2022; 14:e28579. [PMID: 36185863 PMCID: PMC9521169 DOI: 10.7759/cureus.28579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal carcinoma (CRC) is the most common of gastrointestinal cancers, the majority presenting with sporadic occurrence compared to the less frequently inherited syndromes. The increasing incidence, decreasing gender and age disparities, and the prevalent risk factors are concerning. The malignancy arising from benign precursor polyps transforms slowly over time. The adenoma variant polyps reported a marked upregulation of cyclooxygenases (COX), significantly COX-2 isoform, influenced by various determinants such as genetics, pathology, histology, and site of the carcinoma. These COX enzymes are responsible for prostaglandin synthesis and the consequent cascade of cell inflammation and proliferation. Therefore, COX inhibition by non-steroidal anti-inflammatory drugs (NSAIDs) targeted against both the isoforms COX-1 and COX-2 have been studied for decades in anticipation of preventing the occurrence of colorectal carcinoma in high-risk populations. This article has collated and highlighted the overexpression of COX enzymes by the adenomatous polyps and provides corroborating evidence from multiple studies in favor of COX inhibition by NSAIDs. Aspirin and Sulindac were two drugs to be initially proven to halt the progression and cause regression of the polyps. Celecoxib, a selective COX-2 inhibitor besides NSAIDs, was also used in experimental studies.
Collapse
|
5
|
Human microbiota: a crucial gatekeeper in lung cancer initiation, progression, and treatment. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
6
|
Influence of gut and intratumoral microbiota on the immune microenvironment and anti-cancer therapy. Pharmacol Res 2021; 174:105966. [PMID: 34728366 DOI: 10.1016/j.phrs.2021.105966] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 12/31/2022]
Abstract
Microbiota has been implicated in the regulation of tumor progression and therapeutic efficacy. However, the effect of microbiota on disease progression is context dependent, differing according to tumor types, therapeutic regimens, and composition of the microbiota, calling for a deeper understanding of host-microbiome interactions. Previous studies have demonstrated that gut microbiota influences disease progression by regulating local and systemic immunity. Notably, with the advent of next-generation sequencing technology, intratumoral microbiota has also been found and constitutes an important component of the tumor microenvironment. In this review, we summarize recent knowledge about the identification of intra-tumor microbiota and discuss the role of gut and intratumoral microbiota in solid tumors in the angle of immune microenvironment interaction. Furthermore, we discuss how these findings may benefit current anti-cancer approaches. Key problems to be solved in ongoing and future research are highlighted.
Collapse
|
7
|
Xu N, Wang L, Li C, Ding C, Li C, Fan W, Cheng C, Gu B. Microbiota dysbiosis in lung cancer: evidence of association and potential mechanisms. Transl Lung Cancer Res 2020; 9:1554-1568. [PMID: 32953527 PMCID: PMC7481604 DOI: 10.21037/tlcr-20-156] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Over the past decade, revolution in microbial research has provided valuable insights into the function of microbes that inhabit human body. This complex community of microbes, collectively named as microbiota, displays tremendous interaction with a host to maintain homeostasis of the local environment. Lungs were even previously regarded as sterile for a long time. With the development of high-throughput next-generation sequencing technology, a low-density, diversified microbial ecosystem is found in bronchoalveolar lavage fluid, sputum, and lung tissues. Current research confirms that, compared with healthy people, patients with lung cancer show changes in the relative abundance of multiple genera. Emerging evidence has suggested that dysbiosis of the lung microbiota may play a critical role in lung carcinogenesis by affecting metabolic, inflammatory pathways and immune response. We briefly summarize the relationship between lung microbiome and lung cancer and discuss the potential mechanisms mediating lung microbiota and lung cancer. Thus, we provide innovative strategies for early prevention and personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Nana Xu
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, China
| | - Lei Wang
- Department of Histology and Embryology, Xuzhou Medical University, Xuzhou, China
| | - Chenxi Li
- Medical Technology Institute of Xuzhou Medical University, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Cong Li
- Emergency Intensive Care Unit, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenting Fan
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chen Cheng
- Medical Technology Institute of Xuzhou Medical University, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Bing Gu
- Medical Technology Institute of Xuzhou Medical University, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
8
|
Moshi JM, Hoogduin KJ, Ummelen M, Henfling MER, van Engeland M, Wouters KAD, Stoop H, Demers I, Looijenga LHJ, Ramaekers FCS, Hopman ANH. Switches of SOX17 and SOX2 expression in the development of squamous metaplasia and squamous intraepithelial lesions of the uterine cervix. Cancer Med 2020; 9:6330-6343. [PMID: 32644288 PMCID: PMC7476841 DOI: 10.1002/cam4.3201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS The dynamics and topographical distribution of SOX17 and SOX2 expression was studied in the transformation zone (TZ) of the uterine cervix. This TZ is a dynamic area where switches from glandular into squamous epithelium can be recognized, new squamocolumnar junctions are formed, and premalignant lesions originate. SOX17 and SOX2 show mutually exclusive expression patterns in the normal uterine cervix, with SOX2 being exclusively found in squamous epithelium, while SOX17 is detected in endocervical columnar cells and reserve cells. METHODS AND RESULTS Normal cervices and squamous intraepithelial lesions (SIL) were studied with immunohistochemistry, methylation of SOX17, human papilloma virus (HPV) genotyping, and in situ hybridization. In the TZ squamous metaplasia originating from these reserve cells can still show SOX17 expression, while also remnants of SOX17-positive immature metaplasia can be recognized in the normal squamous epithelium. SOX17 expression is gradually lost during maturation, resulting in the exclusive expression of SOX2 in the majority of (SIL). This loss of SOX17 expression is independent of methylation of the CpG island in its promotor region. HPV can be detected in SOX17-positive immature metaplastic regions in the immediate vicinity of SOX2-positive SIL, suggesting that switches in SOX17 and 2 expression can occur upon HPV infection. CONCLUSIONS This switch in expression, and the strong association between the distribution of reserve cells and squamous areas within the columnar epithelium in the TZ, suggests that reserve cell proliferations, next to basal cells in the squamous epithelium, are potential targets for the formation of squamous lesions upon viral infection.
Collapse
Affiliation(s)
- Jobran M Moshi
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Klaas J Hoogduin
- Laboratory of Pathology, Pathan B.V., Rotterdam, The Netherlands
| | - Monique Ummelen
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mieke E R Henfling
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Manon van Engeland
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kim A D Wouters
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Hans Stoop
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Imke Demers
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leendert H J Looijenga
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Frans C S Ramaekers
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Anton N H Hopman
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
9
|
Increased Retinoic Acid Catabolism in Olfactory Sensory Neurons Activates Dormant Tissue-Specific Stem Cells and Accelerates Age-Related Metaplasia. J Neurosci 2020; 40:4116-4129. [PMID: 32385093 DOI: 10.1523/jneurosci.2468-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/03/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
The cellular and molecular basis of metaplasia and declining neurogenesis in the aging olfactory epithelium (OE) remains unknown. The horizontal basal cell (HBC) is a dormant tissue-specific stem cell presumed to only be forced into self-renewal and differentiation by injury. Here we analyze male and female mice and show that HBCs also are activated with increasing age as well as non-cell-autonomously by increased expression of the retinoic acid-degrading enzyme CYP26B1. Activating stimuli induce HBCs throughout OE to acquire a rounded morphology and express IP3R3, which is an inositol-1,4,5-trisphosphate receptor constitutively expressed in stem cells of the adjacent respiratory epithelium. Odor/air stimulates CYP26B1 expression in olfactory sensory neurons mainly located in the dorsomedial OE, which is spatially inverse to ventrolateral constitutive expression of the retinoic acid-synthesizing enzyme (RALDH1) in supporting cells. In ventrolateral OE, HBCs express low p63 levels and preferentially differentiate instead of self-renewing when activated. When activated by chronic CYP26B1 expression, repeated injury, or old age, ventrolateral HBCs diminish in number and generate a novel type of metaplastic respiratory cell that is RALDH- and secretes a mucin-like mucus barrier protein (FcγBP). Conversely, in the dorsomedial OE, CYP26B1 inhibits injury-induced and age-related replacement of RALDH- supporting cells with RALDH1+ ciliated respiratory cells. Collectively, these results support the concept that inositol-1,4,5-trisphosphate type 3 receptor signaling in HBCs, together with altered retinoic acid metabolism within the niche, promote HBC lineage commitment toward two types of respiratory cells that will maintain epithelial barrier function once the capacity to regenerate OE cells ceases.SIGNIFICANCE STATEMENT Little is known about signals that activate dormant stem cells to self-renew and regenerate odor-detecting neurons and other olfactory cell types after loss due to injury, infection, or toxin exposure in the nose. It is also unknown why the stem cells do not prevent age-dependent decline of odor-detecting neurons. We show that (1) stem cells are kept inactive by the vitamin A derivative retinoic acid, which is synthesized and degraded locally by olfactory cells; (2) old age as well as repeated injuries activate the stem cells and exhaust their potential to produce olfactory cells; and (3) exhausted stem cells alter the local retinoic acid metabolism and maintain the epithelial tissue barrier by generating airway cells instead of olfactory cells.
Collapse
|
10
|
Hopman ANH, Moshi JM, Hoogduin KJ, Ummelen M, Henfling MER, van Engeland M, Wouters KAD, Stoop H, Looijenga LHJ, Ramaekers FCS. SOX17 expression and its down-regulation by promoter methylation in cervical adenocarcinoma in situ and adenocarcinoma. Histopathology 2019; 76:383-393. [PMID: 31444787 PMCID: PMC7027543 DOI: 10.1111/his.13980] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022]
Abstract
AIMS SOX17 expression has not been studied in glandular lesions of the uterine cervix like adenocarcinoma in situ (AIS) and invasive adenocarcinomas (AdC), whereas SOX17 promoter CpG island methylation has been reported. Therefore, the aim of this study was to relate the topographical distribution of SOX17 expression and SOX17 methylation status to each other, and to SOX2 expression, human papillomavirus (HPV) type, and physical status of the virus. METHODS AND RESULTS Immunohistochemistry was used in 45 cases to assess expression of SOX17 and SOX2. SOX17 promoter methylation was determined in 25 cases by means of bisulphite conversion and methylation-specific polymerase chain reaction. SOX17 and SOX2 showed a mutually exclusive expression pattern in normal epithelium, with a sharp delineation in the squamocolumnar junction. SOX17 was found in endocervical columnar and reserve cells, whereas SOX2 was exclusively found in squamous epithelium. In both glandular lesions and cases with coexisting glandular and squamous intraepithelial components, a complex combination of SOX17 and SOX2 expression patterns was seen and mutually exclusive expression was lost. Frequently, gain of expression of SOX2 was found and expression of SOX17 was lost. Methylation of the CpG island in the SOX17 promoter was shown to be strongly associated with loss of expression of SOX17 (P = 0.0016). CONCLUSIONS In this study, we show for the first time a direct correlation between the topographical distribution of SOX17 expression and the methylation status of its gene promoter. This explains the heterogeneity of SOX17 expression in the glandular lesions of the cervix. No correlation was found between HPV type and physical status of the virus on the one hand and methylation status on the other.
Collapse
Affiliation(s)
- Anton N H Hopman
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Jobran M Moshi
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Klaas J Hoogduin
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Monique Ummelen
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Mieke E R Henfling
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Manon van Engeland
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Kim A D Wouters
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Hans Stoop
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Leendert H J Looijenga
- Laboratory for Experimental Patho-Oncology, Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands.,Princess Maxima Centre for Paediatric Oncology, Utrecht, the Netherlands
| | - Frans C S Ramaekers
- Department of Molecular Cell Biology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
11
|
Yang ZH, Dang YQ, Ji G. Role of epigenetics in transformation of inflammation into colorectal cancer. World J Gastroenterol 2019; 25:2863-2877. [PMID: 31249445 PMCID: PMC6589733 DOI: 10.3748/wjg.v25.i23.2863] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/24/2019] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
Molecular mechanisms associated with inflammation-promoted tumorigenesis have become an important topic in cancer research. Various abnormal epigenetic changes, including DNA methylation, histone modification, chromatin remodeling, and noncoding RNA regulation, occur during the transformation of chronic inflammation into colorectal cancer (CRC). These changes not only accelerate transformation but also lead to cancer progression and metastasis by activating carcinogenic signaling pathways. The NF-κB and STAT3 signaling pathways play a particularly important role in the transformation of inflammation into CRC, and both are critical to cellular signal transduction and constantly activated in cancer by various abnormal changes including epigenetics. The NF-κB and STAT3 signals contribute to the microenvironment for tumorigenesis through secretion of a large number of pro-inflammatory cytokines and their crosstalk in the nucleus makes it even more difficult to treat CRC. Compared with gene mutation that is irreversible, epigenetic inheritance is reversible or can be altered by the intervention. Therefore, understanding the role of epigenetic inheritance in the inflammation-cancer transformation may elucidate the pathogenesis of CRC and promote the development of innovative drugs targeting transformation to prevent and treat this malignancy. This review summarizes the literature on the roles of epigenetic mechanisms in the occurrence and development of inflammation-induced CRC. Exploring the role of epigenetics in the transformation of inflammation into CRC may help stimulate futures studies on the role of molecular therapy in CRC.
Collapse
Affiliation(s)
- Zhen-Hua Yang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Digestive Endoscopy Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Qi Dang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
12
|
Koleva M, Dikov D, Belovejdov V, Sarafian V. Expression of MUC1 in eosinophilic metaplasia of the prostate. Prostate 2019; 79:622-627. [PMID: 30652335 DOI: 10.1002/pros.23769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/27/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Eosinophilic metaplasia (EM) in the prostate is characterized by the presence of eosinophilic cytoplasmic granules in benign prostatic epithelium. These granules show exocrine-type morphology and positive expression for prostate specific antigen (PSA) and some lysosomal markers. The nature and the full immunohistochemical profile of the granules of EM have not been studied in detail yet. AIM The aim of the current study is to investigate the expression of epithelial mucins (MUCs) in prostatic epithelium with EM. METHODS Twenty specimens from transurethral resection of the prostate (TURP) were reviewed for the presence of EM and were stained with Periodic acid-Schiff's procedure with diastase digestion (PAS.D) and immunostained with PSA and MUCs: MUC1, MUC2, MUC5AC, and MUC6. RESULTS The EM-foci of all prostate glands are PAS.D, PSA positive and show constant immunoreactivity for MUC1. The expression of MUC1 is with membranous and cytoplasmic localization: predominantly apical with membranous accentuation in the cases of EM with large eosinophilic granules, and perinuclear in EM with small eosinophilic granules. There is no expression of other MUCs (MUC2, MUC5AC, and MUC6) in prostatic EM. CONCLUSION We report for the first time that eosinophilic cytoplasmic granules in prostatic EM are MUC1 positive and can vary in size. Based on our immunohistochemical study we suggest that EM of the prostate is not a form of mucinous metaplasia. The present results enrich the available information about the immunophenotype of EM. We assume that MUC1 might serve as a reliable and constant, although nonspecific, immunohistochemical marker of benign EM-phenotype.
Collapse
Affiliation(s)
- Maria Koleva
- Department of General and Clinical Pathology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Dorian Dikov
- Department of General and Clinical Pathology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Service d'Anatomie et Cytologie Pathologiques, Grand Hôpital de l'Est Francilien, Jossigny, France
| | - Veselin Belovejdov
- Department of General and Clinical Pathology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Technological Center for Emergency Medicine, Plovdiv, Bulgaria
| |
Collapse
|
13
|
Lin YS, Lu SY, Wu HP, Chang CF, Chiu YT, Yang HT, Chao PM. Is frying oil a dietary source of an endocrine disruptor? Anti-estrogenic effects of polar compounds from frying oil in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:18-27. [PMID: 30412894 DOI: 10.1016/j.ecoenv.2018.10.111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 06/08/2023]
Abstract
The objective was to investigate endocrine-disrupting effects of polar compounds from oxidized frying oil. Estrogenicity of polar compounds was tested with a rat uterotrophic bioassay. Dietary oxidized frying oil (containing 51% polar compounds) or polar compounds isolated from it were incorporated into feed (in lieu of fresh soybean oil) and fed to ovariectomized rats, with or without treatment with exogenous ethynyl estradiol. Exogenous estrogen restored uterine weight, and caused histological abnormalities (stratified epithelia and conglomerate glands) as well as proliferation of uterine epithelial cells. However, tamoxifen or polar compounds reduced these effects. Furthermore, tamoxifen or polar compounds down-regulated uterine mRNA expression of estrogen receptor (ER)-target genes, implicating reduced ER activity in this hypo-uterotrophic effect. Inhibition of ER signaling and mitosis by polar compounds were attributed to reduced MAPK and AKT activation, as well as a reduced ligand binding domain-transactivity of ERα/β. We concluded polar compounds from frying oil are potential endocrine-disrupting chemicals, with implications for food and environmental safety.
Collapse
Affiliation(s)
- Yu-Shun Lin
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Shui-Yuan Lu
- Department of Applied Toxicology, Taiwan Agricultural Chemicals and Toxic Substances Research Institute, Taichung 413, Taiwan
| | - Hai-Ping Wu
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404, Taiwan
| | - Yung-Tsung Chiu
- Department of Medical Education and Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Hui-Ting Yang
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Pei-Min Chao
- Department of Nutrition, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
14
|
A dualistic model of primary anal canal adenocarcinoma with distinct cellular origins, etiologies, inflammatory microenvironments and mutational signatures: implications for personalised medicine. Br J Cancer 2018; 118:1302-1312. [PMID: 29700411 PMCID: PMC5959925 DOI: 10.1038/s41416-018-0049-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/19/2022] Open
Abstract
Background Primary adenocarcinoma of the anal canal is a rare and aggressive gastrointestinal disease with unclear pathogenesis. Because of its rarity, no clear clinical practice guideline has been defined and a targeted therapeutic armamentarium has yet to be developed. The present article aimed at addressing this information gap by in-depth characterising the anal glandular neoplasms at the histologic, immunologic, genomic and epidemiologic levels. Methods In this multi-institutional study, we first examined the histological features displayed by each collected tumour (n = 74) and analysed their etiological relationship with human papillomavirus (HPV) infection. The intratumoural immune cell subsets (CD4, CD8, Foxp3), the expression of immune checkpoints (PD-1, PD-L1), the defect in mismatch repair proteins and the mutation analysis of multiple clinically relevant genes in the gastrointestinal cancer setting were also determined. Finally, the prognostic significance of each clinicopathological variable was assessed. Results Phenotypic analysis revealed two region-specific subtypes of anal canal adenocarcinoma. The significant differences in the HPV status, density of tumour-infiltrating lymphocytes, expression of immune checkpoints and mutational profile of several targetable genes further supported the separation of these latter neoplasms into two distinct entities. Importantly, anal gland/transitional-type cancers, which poorly respond to standard treatments, displayed less mutations in downstream effectors of the EGFR signalling pathway (i.e., KRAS and NRAS) and demonstrated a significantly higher expression of the immune inhibitory ligand-receptor pair PD-1/PD-L1 compared to their counterparts arising from the colorectal mucosa. Conclusions Taken together, the findings reported in the present article reveal, for the first time, that glandular neoplasms of the anal canal arise by HPV-dependent or independent pathways. These etiological differences leads to both individual immune profiles and mutational landscapes that can be targeted for therapeutic benefits.
Collapse
|
15
|
Mattiucci D, Maurizi G, Leoni P, Poloni A. Aging- and Senescence-associated Changes of Mesenchymal Stromal Cells in Myelodysplastic Syndromes. Cell Transplant 2018; 27:754-764. [PMID: 29682980 PMCID: PMC6047275 DOI: 10.1177/0963689717745890] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hematopoietic stem and progenitor cells reside within the bone marrow (BM) microenvironment. By a well-balanced interplay between self-renewal and differentiation, they ensure a lifelong supply of mature blood cells. Physiologically, multiple different cell types contribute to the regulation of stem and progenitor cells in the BM microenvironment by cell-extrinsic and cell-intrinsic mechanisms. During the last decades, mesenchymal stromal cells (MSCs) have been identified as one of the main cellular components of the BM microenvironment holding an indispensable role for normal hematopoiesis. During aging, MSCs diminish their functional and regenerative capacities and in some cases encounter replicative senescence, promoting inflammation and cancer progression. It is now evident that alterations in specific stromal cells that comprise the BM microenvironment can contribute to hematologic malignancies, and there is growing interest regarding the contribution of MSCs to the pathogenesis of myelodysplastic syndromes (MDSs), a clonal hematological disorder, occurring mostly in the elderly, characterized by ineffective hematopoiesis and increased tendency to acute myeloid leukemia evolution. The pathogenesis of MDS has been associated with specific genetic and epigenetic events occurring both in hematopoietic stem cells (HSCs) and in the whole BM microenvironment with an aberrant cross talk between hematopoietic elements and stromal compartment. This review highlights the role of MSCs in MDS showing functional and molecular alterations such as altered cell-cycle regulation with impaired proliferative potential, dysregulated cytokine secretion, and an abnormal gene expression profile. Here, the current knowledge of impaired functional properties of both aged MSCs and MSCs in MDS have been described with a special focus on inflammation and senescence induced changes in the BM microenvironment. Furthermore, a better understanding of aberrant BM microenvironment could improve future potential therapies.
Collapse
Affiliation(s)
- Domenico Mattiucci
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Maurizi
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Pietro Leoni
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Antonella Poloni
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
16
|
Dong W, Ding T, Wu L, Ren X, Epling-Burnette PK, Yang L. Effect of IL-7 and IL-15 on T cell phenotype in myelodysplastic syndromes. Oncotarget 2018; 7:27479-88. [PMID: 27036031 PMCID: PMC5053665 DOI: 10.18632/oncotarget.8459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
Aberrant T cell phenotype is one of the characteristics of myelodysplastic syndromes (MDS). In this study, we detected an increased concentration of IL-15 in the plasma of MDS patients (n = 20) compared with that in the plasma of healthy controls (n = 20). In MDS patients, reduced naïve CD4+ and CD8+ T cells [16.11 ± 6.56 vs. 24.11 ± 7.18 for CD4+ T cells (p < 0.001) and 13.15 ± 5.67 vs. 23.51 ± 6.25 for CD8+ T cells (p < 0.001)] were observed. The reduced naïve and increased effector memory T cells were significantly correlated with IL-15 plasma level. Then, the effect of IL-15 and IL-7 was tested in vitro. Peripheral blood mononuclear cells from MDS were treated for 15 days with IL-15. This treatment significantly decreased naïve CD4+ (p < 0.001) and CD8+ (p < 0.001) T cells and correspondingly increased terminal memory CD4+ and CD8+ T cells (p < 0.001). Treatment with IL-7 increased naïve CD4+ (p < 0.05) and CD8+ (p < 0.001) T cells. Our results indicated that exposure to high levels of IL-15 may be involved in the T cell phenotype conversion observed in MDS. IL-7 may be one of the promising therapeutic candidates for recovering the effector immune compartment in MDS patients.
Collapse
Affiliation(s)
- Wen Dong
- Department of Orthopaedic Surgery, Tianjin Hongqiao Hospital, Tianjin, P.R. China
| | - Tingting Ding
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Lei Wu
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | | | - Lili Yang
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| |
Collapse
|
17
|
PARTICIPATION OF INTESTINAL DIFFERENTIATION TRANSCRIPTION FACTOR CDX2 IN THE HISTOGENESIS OF THE GASTROINTESTINAL TRACT. WORLD OF MEDICINE AND BIOLOGY 2018. [DOI: 10.26724/2079-8334-2018-4-66-198-202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
18
|
Mao Q, Jiang F, Yin R, Wang J, Xia W, Dong G, Ma W, Yang Y, Xu L, Hu J. Interplay between the lung microbiome and lung cancer. Cancer Lett 2017; 415:40-48. [PMID: 29197615 DOI: 10.1016/j.canlet.2017.11.036] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
The human microbiome confers benefits or disease susceptibility to the human body through multiple pathways. Disruption of the symbiotic balance of the human microbiome is commonly found in systematic diseases such as diabetes, obesity, and chronic gastric diseases. Emerging evidence has suggested that dysbiosis of the microbiota may also play vital roles in carcinogenesis at multiple levels, e.g., by affecting metabolic, inflammatory, or immune pathways. Although the impact of the gut microbiome on the digestive cancer has been widely explored, few studies have investigated the interplay between the microbiome and lung cancer. Some recent studies have shown that certain microbes and microbiota dysbiosis are correlated with development of lung cancer. In this mini-review, we briefly summarize current research findings describing the relationship between the lung microbiome and lung cancer. We further discuss the potential mechanisms through which the lung microbiome may play a role in lung carcinogenesis and impact lung cancer treatment. A better knowledge of the interplay between the lung microbiome and lung cancer may promote the development of innovative strategies for early prevention and personalized treatment in lung cancer.
Collapse
Affiliation(s)
- Qixing Mao
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Jie Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Wenjie Xia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Gaochao Dong
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Weidong Ma
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Yao Yang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China.
| | - Jianzhong Hu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA.
| |
Collapse
|
19
|
Cochrane DR, Tessier-Cloutier B, Lawrence KM, Nazeran T, Karnezis AN, Salamanca C, Cheng AS, McAlpine JN, Hoang LN, Gilks CB, Huntsman DG. Clear cell and endometrioid carcinomas: are their differences attributable to distinct cells of origin? J Pathol 2017; 243:26-36. [PMID: 28678427 DOI: 10.1002/path.4934] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/09/2017] [Accepted: 06/29/2017] [Indexed: 01/18/2023]
Abstract
Endometrial epithelium is the presumed tissue of origin for both eutopic and endometriosis-derived clear cell and endometrioid carcinomas. We had previously hypothesized that the morphological, biological and clinical differences between these carcinomas are due to histotype-specific mutations. Although some mutations and genomic landscape features are more likely to be found in one of these histotypes, we were not able to identify a single class of mutations that was exclusively present in one histotype and not the other. This lack of genomic differences led us to an alternative hypothesis that these cancers could arise from distinct cells of origin within endometrial tissue, and that it is the cellular context that accounts for their differences. In a proteomic screen, we identified cystathionine γ-lyase (CTH) as a marker for clear cell carcinoma, as it is expressed at high levels in clear cell carcinomas of the ovary and endometrium. In the current study, we analysed normal Müllerian tissues, and found that CTH is expressed in ciliated cells of endometrium (both eutopic endometrium and endometriosis) and fallopian tubes. We then demonstrated that other ciliated cell markers are expressed in clear cell carcinomas, whereas endometrial secretory cell markers are expressed in endometrioid carcinomas. The same differential staining of secretory and ciliated cells was demonstrable in a three-dimensional organoid culture system, in which stem cells were stimulated to differentiate into an admixture of secretory and ciliated cells. These data suggest that endometrioid carcinomas are derived from cells of the secretory cell lineage, whereas clear cell carcinomas are derived from, or have similarities to, cells of the ciliated cell lineage. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dawn R Cochrane
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Basile Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Tayyebeh Nazeran
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anthony N Karnezis
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Clara Salamanca
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Angela S Cheng
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
| | - Jessica N McAlpine
- Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, BC, Canada
| | - Lien N Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, BC, Canada.,Department of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
20
|
Minacapelli CD, Bajpai M, Geng X, Cheng CL, Chouthai AA, Souza R, Spechler SJ, Das KM. Barrett's metaplasia develops from cellular reprograming of esophageal squamous epithelium due to gastroesophageal reflux. Am J Physiol Gastrointest Liver Physiol 2017; 312:G615-G622. [PMID: 28336546 DOI: 10.1152/ajpgi.00268.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 02/06/2023]
Abstract
Gastroesophageal reflux disease (GERD) clinically predisposes to columnar Barrett's metaplasia (BM) in the distal esophagus. We demonstrate evidence supporting the cellular origin of BM from reprograming or transcommitment of resident normal esophageal squamous (NES) epithelial cells in response to acid and bile (A + B) exposure using an in vitro cell culture model. The hTERT-immortalized NES cell line NES-B10T was exposed 5 min/day to an A + B mixture for 30 wk. Morphological changes, mRNA, and protein expression levels for the inflammatory marker cyclooxygenase-2; the lineage-determining transcription factors TAp63 (squamous), CDX2, and SOX9 (both columnar); and the columnar lineage markers Villin, Muc-2, CK8, and mAb Das-1 (incomplete phenotype of intestinal metaplasia) were assessed every 10 wk. Markers of columnar lineage and inflammation increased progressively, while squamous lineage-determining transcriptional factors were significantly decreased both at the mRNA and/or protein level in the NES-B10T cells at/after A + B treatment for 30 wk. Distinct modifications in morphological features were only observed at/after 30 wk of A + B exposure. These changes acquired by the NES-B10T 30-wk cells were retained even after cessation of A + B exposure for at least 3 wk. This study provides evidence that chronic exposure to the physiological components of gastric refluxate leads to repression of the discernable squamous transcriptional factors and activation of latent columnar transcriptional factors. This reflects the alteration in lineage commitment of the precursor-like biphenotypic, NES-B10T cells in response to A + B exposure as the possible origin of BM from the resident NES cells.NEW & NOTEWORTHY This study provides evidence of the origins of Barrett's metaplasia from lineage transcommitment of resident esophageal cells after chronic exposure to gastroesophageal refluxate. The preterminal progenitor-like squamous cells alter their differentiation and develop biphenotypic characteristics, expressing markers of incomplete-type columnar metaplasia. Development of these biphenotypic precursors in vitro is a unique model to study pathogenesis of Barrett's metaplasia and esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Carlos D Minacapelli
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| | - Manisha Bajpai
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| | - Xin Geng
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| | - Christina L Cheng
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| | - Abhishek A Chouthai
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| | - Rhonda Souza
- Veterans Affairs North Texas Health Care System-Dallas and the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stuart J Spechler
- Veterans Affairs North Texas Health Care System-Dallas and the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kiron M Das
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey; and
| |
Collapse
|
21
|
Inflammatory networks underlying colorectal cancer. Nat Immunol 2016; 17:230-40. [PMID: 26882261 DOI: 10.1038/ni.3384] [Citation(s) in RCA: 367] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/17/2015] [Indexed: 02/07/2023]
Abstract
Inflammation is emerging as one of the hallmarks of cancer, yet its role in most tumors remains unclear. Whereas a minority of solid tumors are associated with overt inflammation, long-term treatment with non-steroidal anti-inflammatory drugs is remarkably effective in reducing cancer rate and death. This indicates that inflammation might have many as-yet-unrecognized facets, among which an indolent course might be far more prevalent than previously appreciated. In this Review, we explore the various inflammatory processes underlying the development and progression of colorectal cancer and discuss anti-inflammatory means for its prevention and treatment.
Collapse
|
22
|
Nowell CS, Odermatt PD, Azzolin L, Hohnel S, Wagner EF, Fantner GE, Lutolf MP, Barrandon Y, Piccolo S, Radtke F. Chronic inflammation imposes aberrant cell fate in regenerating epithelia through mechanotransduction. Nat Cell Biol 2015; 18:168-80. [PMID: 26689676 DOI: 10.1038/ncb3290] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/18/2015] [Indexed: 12/13/2022]
Abstract
Chronic inflammation is associated with a variety of pathological conditions in epithelial tissues, including cancer, metaplasia and aberrant wound healing. In relation to this, a significant body of evidence suggests that aberration of epithelial stem and progenitor cell function is a contributing factor in inflammation-related disease, although the underlying cellular and molecular mechanisms remain to be fully elucidated. In this study, we have delineated the effect of chronic inflammation on epithelial stem/progenitor cells using the corneal epithelium as a model tissue. Using a combination of mouse genetics, pharmacological approaches and in vitro assays, we demonstrate that chronic inflammation elicits aberrant mechanotransduction in the regenerating corneal epithelium. As a consequence, a YAP-TAZ/β-catenin cascade is triggered, resulting in the induction of epidermal differentiation on the ocular surface. Collectively, the results of this study demonstrate that chronic inflammation and mechanotransduction are linked and act to elicit pathological responses in regenerating epithelia.
Collapse
Affiliation(s)
- Craig S Nowell
- Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland 1015, Switzerland
| | - Pascal D Odermatt
- Laboratory for Bio- and Nano-Instrumentation (LBNI), Institute of Bioengineering (IBI), EPFL, Lausanne, Switzerland 1015, Switzerland
| | - Luca Azzolin
- University of Padua, Department of Molecular Medicine, via G. Colombo 3, 35131 Padova, Italy
| | - Sylke Hohnel
- Laboratory of Stem Cell Bioengineering (LSCB), IBI, EPFL, Lausanne, Switzerland 1015, Switzerland
| | - Erwin F Wagner
- Genes, Development, and Disease Group, F-BBVA Cancer Cell Biology Programme, National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Georg E Fantner
- Laboratory for Bio- and Nano-Instrumentation (LBNI), Institute of Bioengineering (IBI), EPFL, Lausanne, Switzerland 1015, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering (LSCB), IBI, EPFL, Lausanne, Switzerland 1015, Switzerland
| | - Yann Barrandon
- Stem Cell Dynamics Laboratory (LDSC), IBI, EPFL, Lausanne, Switzerland 1015, Switzerland
| | - Stefano Piccolo
- University of Padua, Department of Molecular Medicine, via G. Colombo 3, 35131 Padova, Italy
| | - Freddy Radtke
- Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland 1015, Switzerland
| |
Collapse
|
23
|
Senescence-associated inflammatory responses: aging and cancer perspectives. Trends Immunol 2015; 36:217-28. [PMID: 25801910 DOI: 10.1016/j.it.2015.02.009] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023]
Abstract
Senescent cells, albeit not proliferating, are metabolically and transcriptionally active, thereby capable of affecting their microenvironment, notably via the production of inflammatory mediators. These mediators maintain and propagate the senescence process to neighboring cells, and then recruit immune cells for clearing senescent cells. Among the inflammatory cues are molecules with pronounced tumor-controlling properties, both growth and invasion factors and inhibitory factors, working directly or via recruited immune cells. These senescence-inflammatory effects also prevail within tumors, mediated by the senescent tumor cells and the senescent tumor stroma. Here, we review the course and impact of senescence-associated inflammatory responses in aging and cancer. We propose that controlling senescence-associated inflammation by targeting specific inflammatory mediators may have a beneficial therapeutic effect in treatment of cancer and aging-related diseases.
Collapse
|
24
|
Demoulin SA, Somja J, Duray A, Guénin S, Roncarati P, Delvenne PO, Herfs MF, Hubert PM. Cervical (pre)neoplastic microenvironment promotes the emergence of tolerogenic dendritic cells via RANKL secretion. Oncoimmunology 2015; 4:e1008334. [PMID: 26155412 PMCID: PMC4485731 DOI: 10.1080/2162402x.2015.1008334] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/16/2014] [Accepted: 01/08/2015] [Indexed: 12/20/2022] Open
Abstract
The progression of genital human papillomavirus (HPV) infections into preneoplastic lesions suggests that infected/malignant cells are not adequately recognized by the immune system. In this study, we demonstrated that cervical/vulvar cancer cells secrete factor(s) that affect both the maturation and function of dendritic cells (DC) leading to a tolerogenic profile. Indeed, DC cocultured with cancer cell lines display both a partially mature phenotype after lipopolysaccharide (LPS) maturation and an altered secretory profile (IL-10high and IL-12p70low). In addition, tumor-converted DC acquire the ability to alter T-cell proliferation and to induce FoxP3+ suppressive T cells from naive CD4+ T cells. Among the immunosuppressive factors implicated in DC alterations in genital (pre)neoplastic microenvironment, we identified receptor activator of nuclear factor kappa-B ligand (RANKL), a TNF family member, as a potential candidate. For the first time, we showed that RANKL expression strongly increases during cervical progression. We also confirmed that RANKL is directly secreted by cancer cells and this expression is not related to HPV viral oncoprotein induction. Interestingly, the addition of osteoprotegerin (OPG) in coculture experiments reduces significantly the inhibition of DC maturation, the release of a tolerogenic cytokine profile (IL-12low IL-10high) and the induction of regulatory T (Treg) cells. Our findings suggest that the use of inhibitory molecules directed against RANKL in cervical/vulvar (pre)neoplastic lesions might prevent alterations of DC functionality and represent an attractive strategy to overcome immune tolerance in such cancers.
Collapse
Key Words
- LC, Langerhans cells; LPS, lipopolysaccharide
- APC, antigen presenting cells; DC, dendritic cells
- GILZ, glucocorticoid-induced leucine zipper; HPV, human papillomavirus
- HSIL, high grade intraepithelial lesions
- IHC, immunohistochemistry
- ILT3, Immunoglobulin-like transcript 3
- KN, normal keratinocytes
- LSIL, low grade intraepithelial lesion
- MFI, mean fluorescence intensity
- OPG, osteoprotegerin
- PBMC, peripheral blood mononuclear cells; pDC, plasmacytoid dendritic cells
- RANKL
- RANKL, Receptor activator of nuclear factor kappa-B ligand
- SCC, squamous cell carcinoma
- SIL, squamous intraepithelial neoplasia
- Treg cells
- Treg cells, regulatory T cells
- VIN, vulvar intraepithelial neoplasia
- cervical cancers
- dendritic cells
- tolerogenicity
Collapse
Affiliation(s)
- Stéphanie A Demoulin
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| | - Joan Somja
- Department of Pathology; University Hospital of Liège ; Liège, Belgium
| | - Anaëlle Duray
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| | - Samuel Guénin
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| | - Patrick Roncarati
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| | | | - Michael F Herfs
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| | - Pascale M Hubert
- Laboratory of Experimental Pathology; GIGA-Cancer; University of Liège ; Liège, Belgium
| |
Collapse
|
25
|
Demoulin S, Herfs M, Somja J, Roncarati P, Delvenne P, Hubert P. HMGB1 secretion during cervical carcinogenesis promotes the acquisition of a tolerogenic functionality by plasmacytoid dendritic cells. Int J Cancer 2014; 137:345-58. [PMID: 25492101 DOI: 10.1002/ijc.29389] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 11/26/2014] [Indexed: 12/18/2022]
Abstract
Acquisition of an impaired functionality by plasmacytoid dendritic cells (pDCs) contributing to cancer progression has been documented in different types of cancers. In the present study, we postulate that molecules secreted by (pre)neoplastic epithelial cells of the genital tract (cervix/vulva) might attract pDCs but also modify their proper functionality, allowing these cells to initiate a tolerogenic response interfering with antitumor immunity. We demonstrated that pDCs are recruited during the cervical metaplasia-dysplasia-cancer sequence, through the action of their chemoattractant, chemerin. We showed that stimulated-pDCs exposed to cervical/vulvar tumor microenvironment display an altered phenotype. We also demonstrated that cervical/vulvar neoplastic keratinocytes inhibit the proper function of pDCs by decreasing their IFNα secretion in response to CpG oligonucleotides. In parallel, we observed that (pre)neoplastic areas of the cervix are infiltrated by FoxP3(+) Treg cells which colocalize with pDCs. Accordingly, pDCs cocultured with cervical/vulvar neoplastic keratinocytes have the capacity to induce a Treg cell differentiation from naïve CD4(+) T cells, which is in agreement with the development of a tolerogenic response. We identified HMGB1 as a soluble factor produced by neoplastic keratinocytes from the genital tract involved in pDCs functional alteration. Indeed, this molecule inhibited pDC maturation, decreased IFNα secretion following TLR9 stimulation and forced these cells to become tolerogenic. In contrast, inhibition of HMGB1 restored pDC phenotype. Our findings indicate that the use of inhibitory molecules notably directed against HMGB1 in cervical/vulvar (pre)neoplastic lesions might prevent alterations of pDCs functionality and represent an attractive therapeutic strategy to overcome immune tolerance in cancers.
Collapse
Affiliation(s)
- Stéphanie Demoulin
- Department of Pathology, Laboratory of Experimental Pathology, University of Liège, GIGA-Cancer, 4000, Liège, Belgium
| | - Michael Herfs
- Department of Pathology, Laboratory of Experimental Pathology, University of Liège, GIGA-Cancer, 4000, Liège, Belgium
| | - Joan Somja
- Department of Pathology, University Hospital of Liège, 4000, Liège, Belgium
| | - Patrick Roncarati
- Department of Pathology, Laboratory of Experimental Pathology, University of Liège, GIGA-Cancer, 4000, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital of Liège, 4000, Liège, Belgium
| | - Pascale Hubert
- Department of Pathology, Laboratory of Experimental Pathology, University of Liège, GIGA-Cancer, 4000, Liège, Belgium
| |
Collapse
|
26
|
Ng K, Meyerhardt JA, Chan AT, Sato K, Chan JA, Niedzwiecki D, Saltz LB, Mayer RJ, Benson AB, Schaefer PL, Whittom R, Hantel A, Goldberg RM, Venook AP, Ogino S, Giovannucci EL, Fuchs CS. Aspirin and COX-2 inhibitor use in patients with stage III colon cancer. J Natl Cancer Inst 2014; 107:345. [PMID: 25432409 DOI: 10.1093/jnci/dju345] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We conducted a prospective, observational study of aspirin and COX-2 inhibitor use and survival in stage III colon cancer patients enrolled in an adjuvant chemotherapy trial. Among 799 eligible patients, aspirin use was associated with improved recurrence-free survival (RFS) (multivariable hazard ratio [HR] = 0.51, 95% confidence interval [CI] = 0.28 to 0.95), disease-free survival (DFS) (HR = 0.68, 95% CI = 0.42 to 1.11), and overall survival (OS) (HR = 0.63, 95% CI = 0.35 to 1.12). Adjusted HRs for DFS and OS censored at five years (in an attempt to minimize misclassification from noncancer death) were 0.61 (95% CI = 0.36 to 1.04) and 0.48 (95% CI = 0.23 to 0.99). Among 843 eligible patients, those who used COX-2 inhibitors had multivariable HRs for RFS, DFS, and OS of 0.53 (95% CI = 0.27 to 1.04), 0.60 (95% CI = 0.33 to 1.08), and 0.50 (95% CI = 0.23 to 1.07), and HRs of 0.47 (95% CI = 0.24 to 0.91) and 0.26 (95% CI = 0.08 to 0.81) for DFS and OS censored at five years. Aspirin and COX-2 inhibitor use may be associated with improved outcomes in stage III colon cancer patients.
Collapse
Affiliation(s)
- Kimmie Ng
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA.
| | - Jeffrey A Meyerhardt
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Andrew T Chan
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Kaori Sato
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Jennifer A Chan
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Donna Niedzwiecki
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Leonard B Saltz
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Robert J Mayer
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Al B Benson
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Paul L Schaefer
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Renaud Whittom
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Alexander Hantel
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Richard M Goldberg
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Alan P Venook
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Shuji Ogino
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Edward L Giovannucci
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| | - Charles S Fuchs
- : Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (KN, JAM, KS, JAC, RJM, SO, CSF); Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (ATC, ELG, CSF); Alliance Statistics and Data Center, Duke University Medical Center, Durham, NC (DN); Memorial Sloan-Kettering Cancer Center, New York, NY (LBS); Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL (ABB); Toledo Community Hospital Oncology Program, Toledo, OH (PLS); Hopital du Sacre-Coeur de Montreal, Universite de Montreal, Quebec, Canada (RW); Edward Cancer Center, Naperville, IL (AH); Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH (RMG); Division of Medical Oncology, University of California at San Francisco, San Francisco, CA (APV); Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SO); Department of Epidemiology (SO, ELG) and Department of Nutrition (ELG), Harvard School of Public Health, Boston, MA
| |
Collapse
|
27
|
Subchronic inhalation of coal dust particulate matter 10 induces bronchoalveolar hyperplasia and decreases MUC5AC expression in male Wistar rats. ACTA ACUST UNITED AC 2014; 66:383-9. [DOI: 10.1016/j.etp.2014.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/09/2014] [Accepted: 06/02/2014] [Indexed: 11/23/2022]
|
28
|
Sauder CAM, Koziel JE, Choi M, Fox MJ, Grimes BR, Badve S, Blosser RJ, Radovich M, Lam CC, Vaughan MB, Herbert BS, Clare SE. Phenotypic plasticity in normal breast derived epithelial cells. BMC Cell Biol 2014; 15:20. [PMID: 24915897 PMCID: PMC4066279 DOI: 10.1186/1471-2121-15-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/22/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Normal, healthy human breast tissue from a variety of volunteer donors has become available for research thanks to the establishment of the Susan G. Komen for the Cure® Tissue Bank at the IU Simon Cancer Center (KTB). Multiple epithelial (K-HME) and stromal cells (K-HMS) were established from the donated tissue. Explant culture was utilized to isolate the cells from pieces of breast tissue. Selective media and trypsinization were employed to select either epithelial cells or stromal cells. The primary, non-transformed epithelial cells, the focus of this study, were characterized by immunohistochemistry, flow cytometry, and in vitro cell culture. RESULTS All of the primary, non-transformed epithelial cells tested have the ability to differentiate in vitro into a variety of cell types when plated in or on biologic matrices. Cells identified include stratified squamous epithelial, osteoclasts, chondrocytes, adipocytes, neural progenitors/neurons, immature muscle and melanocytes. The cells also express markers of embryonic stem cells. CONCLUSIONS The cell culture conditions employed select an epithelial cell that is pluri/multipotent. The plasticity of the epithelial cells developed mimics that seen in metaplastic carcinoma of the breast (MCB), a subtype of triple negative breast cancer; and may provide clues to the origin of this particularly aggressive type of breast cancer. The KTB is a unique biorepository, and the normal breast epithelial cells isolated from donated tissue have significant potential as new research tools.
Collapse
Affiliation(s)
- Candice AM Sauder
- Department of Surgery, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Jillian E Koziel
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, Indianapolis, IN 46202, USA
| | - MiRan Choi
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Melanie J Fox
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Brenda R Grimes
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Sunil Badve
- Department of Pathology, Indiana University School of Medicine, 350 West 11th Street, Indianapolis, IN 46202, USA
| | - Rachel J Blosser
- Department of Surgery, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Milan Radovich
- Department of Surgery, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Christina C Lam
- Department of Biology, University of Central Oklahoma, 100 North University Drive, Edmond, OK 73034, USA
| | - Melville B Vaughan
- Department of Biology, University of Central Oklahoma, 100 North University Drive, Edmond, OK 73034, USA
| | - Brittney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 975 W. Walnut Street, Indianapolis, IN 46202, USA
| | - Susan E Clare
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Improved computer-assisted analysis of the global lymphatic network in human cervical tissues. Mod Pathol 2014; 27:887-98. [PMID: 24309324 DOI: 10.1038/modpathol.2013.195] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 09/20/2013] [Accepted: 09/23/2013] [Indexed: 11/08/2022]
Abstract
Lymphatic dissemination is a key event in cervical cancer progression and related tumor lymphatic markers are viewed as promising prognostic factor of nodal extension. However, validating such parameters requires an objective characterization of the lymphatic vasculature. Here, we performed a global analysis of the lymphatic network using a new computerized method applied on whole uterine cervical digital images. Sixty-eight cases of cervical neoplasia (12 CIN3, 10 FIGO stage 1A and 46 stage IB1) and 10 cases of normal cervical tissue were reacted with antibodies raised against D2-40, D2-40/p16 and D2-40/Ki67. Immunostained structures were automatically detected on whole slides. The lymphatic vessel density (D2-40), proliferating lymphatic vessel density (D2-40/ki67) and spatial lymphatic distribution in respect to the adjacent epithelium were assessed from normal cervix to early cervical cancer and correlated with lymphovascular space invasion and lymph node status. Prominent lymphatic vessel density and proliferating lymphatic vessel density are detected under the transformation zone of benign cervix and no further increase is noted during cancer progression. Notably, a shift of lymphatic vessel distribution toward the neoplastic edges is detected. In IB1 cervical cancer, although intra- and peritumoral lymphatic vessel density are neither correlated with lymphovascular space invasion nor with lymph node metastasis, a specific spatial distribution with more lymphatic vessels in the vicinity of tumor edges is predictive of lymphatic dissemination. Herein, we provide a new computerized method suitable for an innovative detailed analysis of the lymphatic network. We show that the transformation zone of the benign cervix acts as a baseline lymphangiogenic niche before the initiation of neoplastic process. During cancer progression, this specific microenvironment is maintained with lymphatic vessels even in closer vicinity to tumor cells.
Collapse
|
30
|
Bile acid at low pH reduces squamous differentiation and activates EGFR signaling in esophageal squamous cells in 3-D culture. J Gastrointest Surg 2013; 17:1723-31. [PMID: 23921815 DOI: 10.1007/s11605-013-2287-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 07/05/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND Barrett's esophagus is a preneoplastic metaplasia in which the normal squamous epithelium of the esophagus changes to an intestinal, columnar phenotype due to long-term gastro-esophageal reflux. The major components of this reflux are bile and stomach acid. Previous in vitro studies on the effect of bile and acid on esophageal cells have predominantly relied on transformed esophageal squamous cells or cancer cells grown in monolayer culture. DISCUSSION In this study, we expanded our previous work using an immortalized primary esophageal squamous cell line (EPC1). We demonstrate that EPC1 cells form a multi-layer, stratified epithelium when grown on polyester transwell filters in media supplemented with calcium. When exposed to short pulses of bile and pH 5, but not either condition alone, EPC1 cells demonstrate a reduction in stratification layers and reduced expression of squamous epithelium-specific genes. Bile at pH 5 also causes activation of epidermal growth factor receptor and down-stream pathways. Blocking epidermal growth factor receptor activation partially attenuates the effects of bile acid and pH 5. These results suggest that bile at low pH, but not bile or low pH alone, promotes loss of differentiation status of stratified squamous esophageal epithelium in vitro, possibly by initiating a mucosal repair response through epidermal growth factor activation.
Collapse
|
31
|
Herfs M, Vargas SO, Yamamoto Y, Howitt BE, Nucci MR, Hornick JL, McKeon FD, Xian W, Crum CP. A novel blueprint for 'top down' differentiation defines the cervical squamocolumnar junction during development, reproductive life, and neoplasia. J Pathol 2013; 229:460-8. [PMID: 23007879 DOI: 10.1002/path.4110] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 08/17/2012] [Accepted: 09/13/2012] [Indexed: 02/05/2023]
Abstract
The cervical squamocolumnar (SC) junction is the site of a recently discovered 'embryonic' cell population that was proposed as the cell of origin for cervical cancer and its precursors. How this population participates in cervical remodelling and neoplasia is unclear. In the present study, we analysed the SC junction immunophenotype during pre- and post-natal human and mouse development and in the adult, processes of metaplastic evolution of the SC junction, microglandular change, and early cervical neoplasia. Early in life, embryonic cervical epithelial cells were seen throughout the cervix and subsequently diminished in number to become concentrated at the SC junction in the adult. In all settings, there was a repetitive scenario in which cuboidal embryonic/SC junction cells gave rise to subjacent metaplastic basal/reserve cells with a switch from the SC junction positive to negative immunophenotype. This downward or basal (rather than upward or apical) evolution from progenitor cell to metaplastic progeny was termed reverse or 'top down' differentiation. A similar pattern was noted in high-grade squamous intraepithelial lesions (HSILs), suggesting that HPV infection of the cuboidal SC junction cells initiated outgrowth of basally-oriented neoplastic progeny. The progressive loss of the embryonic/SC junction markers occurred with 'top down' differentiation during development, remodelling, and early neoplasia. Interestingly, most low-grade SILs were SC junction-negative, implying infection of metaplastic progeny rather than the original SC junction cells. This proposed model of 'top down' differentiation resolves the mystery of how SC junction cells both remodel the cervix and participate in neoplasia and provides for a second population of metaplastic progeny (including basal and reserve cells), the infection of which is paradoxically less likely to produce a biologically aggressive precursor. It also provides new targets in animal models to determine why the SC junction is uniquely susceptible to carcinogenic HPV infection.
Collapse
Affiliation(s)
- Michael Herfs
- Division of Women's and Perinatal Pathology, Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
A high-quality body of evidence supports the use of aspirin in reducing sporadic and hereditary adenomatous polyps, and numerous observational studies point to a reduction in colorectal cancer (CRC) risk. However, using aspirin as an adjuvant therapy in established CRC was until recently inconceivable. Now, evidence from both observational and clinical trials of aspirin for other indications suggests that aspirin initiation after (or before) the diagnosis of CRC improves CRC-specific mortality. These exciting findings need to be confirmed in prospective randomized trials that are underway. The recent failure of adjuvant irinotecan, bevacizumab, and cetuximab clinical trials compels us to reconsider our assumptions and paradigms for treating CRC. In this Review, we summarize clinical and preclinical evidence supporting aspirin use in established CRC and outline a framework for better understanding aspirin activity in the pathogenesis of CRC. We describe the data supporting adjuvant aspirin in resected CRC, including the issues of dose, duration and toxicity, and discuss potential biomarkers that may help better select patients for aspirin therapy.
Collapse
|
33
|
Herfs M, Hubert P, Poirrier AL, Vandevenne P, Renoux V, Habraken Y, Cataldo D, Boniver J, Delvenne P. Proinflammatory Cytokines Induce Bronchial Hyperplasia and Squamous Metaplasia in Smokers. Am J Respir Cell Mol Biol 2012; 47:67-79. [DOI: 10.1165/rcmb.2011-0353oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
34
|
Abstract
Although development leads unidirectionally toward more restricted cell fates, recent work in cellular reprogramming has proven that one cellular identity can strikingly convert into another, promising countless applications in biomedical research and paving the way for modeling diseases with patient-derived stem cells. To date, there has been little discussion of which disease models are likely to be most informative. Here, we review evidence demonstrating that, because environmental influences and epigenetic signatures are largely erased during reprogramming, patient-specific models of diseases with strong genetic bases and high penetrance are likely to prove most informative in the near term. We also discuss the implications of the new reprogramming paradigm in biomedicine and outline how reprogramming of cell identities is enhancing our understanding of cell differentiation and prospects for cellular therapies and in vivo regeneration.
Collapse
|
35
|
Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol 2012; 95:17-25. [DOI: 10.1007/s12185-011-1001-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/12/2011] [Indexed: 12/11/2022]
|
36
|
|
37
|
Abstract
Barrett's esophagus is an epithelial metaplasia associated with an increased risk for cancer, but its underlying mechanisms have been debated. Now Wang et al. (2011) suggest an intriguing explanation for this puzzle: a population of residual embryonic cells, lacking the transcription factor p63, migrates and repopulates a normal tissue damaged by inflammation or gastroesophageal reflux.
Collapse
Affiliation(s)
- Karine Lefort
- Department of Biochemistry, University of Lausanne, Epalinges CH-1066, Switzerland
| | | |
Collapse
|
38
|
Mucosal junctions: open doors to HPV and HIV infections? Trends Microbiol 2011; 19:114-20. [DOI: 10.1016/j.tim.2010.12.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 12/09/2010] [Accepted: 12/14/2010] [Indexed: 12/29/2022]
|
39
|
Tafani M, Russo A, Di Vito M, Sale P, Pellegrini L, Schito L, Gentileschi S, Bracaglia R, Marandino F, Garaci E, Russo MA. Up-regulation of pro-inflammatory genes as adaptation to hypoxia in MCF-7 cells and in human mammary invasive carcinoma microenvironment. Cancer Sci 2010; 101:1014-23. [PMID: 20151982 PMCID: PMC11159242 DOI: 10.1111/j.1349-7006.2010.01493.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The role of tumor cells in synthesizing pro-inflammatory molecules is still controversial. Here we report that hypoxic treatment of the MCF-7 human mammary adenocarcinoma cell line induced activation of hypoxia-inducible factor 1alpha (HIF-1alpha) and nuclear factor-kappa B (NF-kappaB). Importantly, hypoxia regulated expression of alarmin receptors such as the receptor for advanced glycation end products (RAGE) and the purinoreceptor (P2X7R), and up-regulated inflammatory response (IR) genes such as the inducible enzymes nitric oxide synthase (NOS2), cycloxygenase (COX2), and the acute-phase protein pentraxin-3 (PTX3). Hypoxia also stimulated chemokine (C-X-C motif) receptor 4 (CXCR4) mRNA synthesis. In fact, the CXCR4 ligand stromal-derived factor-1alpha (SDF-1alpha) increased invasion and migration of hypoxic MCF-7 cells. Inhibition of HIF-1alpha by chetomin and NF-kappaB by parthenolide reduced mRNA and protein expression of the studied molecules and prevented invasion of hypoxic MCF-7 cells. Moreover, solid invasive mammary tumor microenvironment was analyzed after laser-capture microdissection (LCMD) comparing tumor versus host normal tissue. Nuclear translocation of HIF-1alpha and NF-kappaB and up-regulation of IR, CXCR4, estrogen receptor alpha (ERalpha), and epithelial growth factor receptor (EGFR) was observed in tumor but not in host normal tissue in the absence of a local inflammatory leukocyte infiltrate. We conclude that under hypoxic conditions MCF-7 cells acquire a pro-inflammatory phenotype, and that solid human mammary carcinoma evidenced a similar activation of HIF-1alpha, NF-kappaB, and IR genes in malignant tumor cells as compared to the normal host tissues. We suggest a role for IR activation in the malignant progression of transformed cells.
Collapse
Affiliation(s)
- Marco Tafani
- Department of Cellular and Molecular Pathology, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kyrgidis A, Tzellos TG, Kechagias N, Patrikidou A, Xirou P, Kitikidou K, Bourlidou E, Vahtsevanos K, Antoniades K. Cutaneous squamous cell carcinoma (SCC) of the head and neck: risk factors of overall and recurrence-free survival. Eur J Cancer 2010; 46:1563-72. [PMID: 20338745 DOI: 10.1016/j.ejca.2010.02.046] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 02/22/2010] [Accepted: 02/24/2010] [Indexed: 11/30/2022]
Abstract
BACKGROUND Head and neck cutaneous squamous cell carcinoma (HNCSCC) although rarely fatal has significant adverse public health effects due to high medical costs, compromised quality of life, functional impairment and other serious consequences. The present longitudinal cohort study of HNCSCC was designed to determine whether certain clinical-pathologic features of HNCSCC are associated with reduced overall and recurrence-free survival, as suggested by previous data. PATIENTS The cohort sample consisted of 315 consecutive patients presenting with primary HNCSCC of the head and neck. Life-table analysis and Kaplan-Meier survival analysis were performed. Multivariate Cox's proportional hazards regression models were used to assess the effects of covariates on the length of the interval. RESULTS There were 145 male and 170 female Caucasian patients. At the time of analysis, 222 patients were alive. The mean follow-up time of a patient after enrolment has been 46.7 months (range, 12-124 months). Broder's differentiation grade, perineural involvement, the presence of inflammation and T-stage were independent adjusted predictors for overall survival. pT and N-stage, inflammation and perineural involvement were significant predictors for recurrence-free survival while adjuvant irradiation was associated with a 92% reduced risk for recurrence. Life-table analysis showed that 87% and 69% study patients were free from recurrence at years 3 and 5, respectively. CONCLUSIONS Certain clinico-pathological predictors can be used to discriminate subsets of high-risk patients that could benefit from long-term follow-up. After excision in negative margins, patients with HNCSCC should be referred to specialised multidisciplinary oncology clinics for counselling on adjuvant radiotherapy and follow-up.
Collapse
Affiliation(s)
- Athanassios Kyrgidis
- Department of Maxillofacial Surgery, Theagenio Cancer Hospital, Thessaloniki, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|