1
|
Cui H, Yu Q, Xu Q, Lin C, Zhang L, Ye W, Yang Y, Tian S, Zhou Y, Sun R, Meng Y, Yao N, Wang H, Cao F, Liu M, Ma J, Liao C, Sun R. EGFR and MUC1 as dual-TAA drug targets for lung cancer and colorectal cancer. Front Oncol 2024; 14:1433033. [PMID: 39664199 PMCID: PMC11631732 DOI: 10.3389/fonc.2024.1433033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/23/2024] [Indexed: 12/13/2024] Open
Abstract
Background Epidermal growth factor receptor (EGFR) is a key protein in cellular signaling that is overexpressed in many human cancers, making it a compelling therapeutic target. On-target severe skin toxicity has limited its clinical application. Dual-targeting therapy represents a novel approach to overcome the challenges of EGFR-targeted therapies. Methods A single-cell tumor-normal RNA transcriptomic meta-atlas of lung adenocarcinoma (LUAD) and normal lung tissues was constructed from published data. Tumor associated antigens (TAAs) were screened from the genes which were expressed on cell surface and could distinguish cancer cells from normal cells. Expression of MUC1 and EGFR in tumors and normal tissues was detected by immunohistochemistry (IHC), bulk transcriptomic and single-cell transcriptomic analyses. RNA cut-off values were calculated using paired analysis of RNA sequencing and IHC in patient-derived tumor xenograft samples. They were used to estimate the abundance of EGFR- and MUC-positive subjects in The Cancer Genome Atlas Program (TCGA) database. Survival analysis of EGFR and MUC1 expression was carried out using the transcription and clinical data from TCGA. Results A candidate TAA target, transmembrane glycoprotein mucin 1 (MUC1), showed strong expression in cancer cells and low expression in normal cells. Single-cell analysis suggested EGFR and MUC1 together had better tumor specificity than the combination of EGFR with other drug targets. IHC data confirmed that EGFR and MUC1 were highly expressed on LUAD and colorectal cancer (CRC) clinical samples but not on various normal tissues. Notably, co-expression of EGFR and MUC1 was observed in 98.4% (n=64) of patients with LUAD and in 91.6% (n=83) of patients with CRC. It was estimated that EGFR and MUC1 were expressed in 97.5% of LUAD samples in the TCGA dataset. Besides, high expression of EGFR and MUC1 was significantly associated with poor prognosis of LUAD and CRC patients. Conclusions Single-cell RNA, bulk RNA and IHC data demonstrated the high expression levels and co-expression patterns of EGFR and MUC1 in tumors but not normal tissues. Therefore, it is a promising TAA combination for therapeutic targeting which could enhance on-tumor efficacy while reducing off-tumor toxicity.
Collapse
Affiliation(s)
- Huilin Cui
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qianqian Yu
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qumiao Xu
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Chen Lin
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Long Zhang
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Wei Ye
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Yifei Yang
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Sijia Tian
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Yilu Zhou
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Runzhe Sun
- School of Basic Medicine, Shanxi Medical University, Jinzhong, China
| | - Yongsheng Meng
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ningning Yao
- Department of Radiobiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Haizhen Wang
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Feilin Cao
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Meilin Liu
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jinfeng Ma
- Department of Hepatobiliary and Pancreatogastric Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Cheng Liao
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Ruifang Sun
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Long AW, Xu H, Santich BH, Guo H, Hoseini SS, de Stanchina E, Cheung NKV. Heterodimerization of T cell engaging bispecific antibodies to enhance specificity against pancreatic ductal adenocarcinoma. J Hematol Oncol 2024; 17:20. [PMID: 38650005 PMCID: PMC11036555 DOI: 10.1186/s13045-024-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND EGFR and/or HER2 expression in pancreatic cancers is correlated with poor prognoses. We generated homodimeric (EGFRxEGFR or HER2xHER2) and heterodimeric (EGFRxHER2) T cell-engaging bispecific antibodies (T-BsAbs) to direct polyclonal T cells to these antigens on pancreatic tumors. METHODS EGFR and HER2 T-BsAbs were constructed using the 2 + 2 IgG-[L]-scFv T-BsAbs format bearing two anti-CD3 scFvs attached to the light chains of an IgG to engage T cells while retaining bivalent binding to tumor antigens with both Fab arms. A Fab arm exchange strategy was used to generate EGFRxHER2 heterodimeric T-BsAb carrying one Fab specific for EGFR and one for HER2. EGFR and HER2 T-BsAbs were also heterodimerized with a CD33 control T-BsAb to generate 'tumor-monovalent' EGFRxCD33 and HER2xCD33 T-BsAbs. T-BsAb avidity for tumor cells was studied by flow cytometry, cytotoxicity by T-cell mediated 51Chromium release, and in vivo efficacy against cell line-derived xenografts (CDX) or patient-derived xenografts (PDX). Tumor infiltration by T cells transduced with luciferase reporter was quantified by bioluminescence. RESULTS The EGFRxEGFR, HER2xHER2, and EGFRxHER2 T-BsAbs demonstrated high avidity and T cell-mediated cytotoxicity against human pancreatic ductal adenocarcinoma (PDAC) cell lines in vitro with EC50s in the picomolar range (0.17pM to 18pM). They were highly efficient in driving human polyclonal T cells into subcutaneous PDAC xenografts and mediated potent T cell-mediated anti-tumor effects. Both EGFRxCD33 and HER2xCD33 tumor-monovalent T-BsAbs displayed substantially reduced avidity by SPR when compared to homodimeric EGFRxEGFR or HER2xHER2 T-BsAbs (∼150-fold and ∼6000-fold respectively), tumor binding by FACS (8.0-fold and 63.6-fold), and T-cell mediated cytotoxicity (7.7-fold and 47.2-fold), while showing no efficacy against CDX or PDX. However, if either EGFR or HER2 was removed from SW1990 by CRISPR-mediated knockout, the in vivo efficacy of heterodimeric EGFRxHER2 T-BsAb was lost. CONCLUSION EGFR and HER2 were useful targets for driving T cell infiltration and tumor ablation. Two arm Fab binding to either one or both targets was critical for robust anti-tumor effect in vivo. By engaging both targets, EGFRxHER2 heterodimeric T-BsAb exhibited potent anti-tumor effects if CDX or PDX were EGFR+HER2+ double-positive with the potential to spare single-positive normal tissue.
Collapse
Affiliation(s)
- Alan W Long
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Brian H Santich
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Hongfen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | | | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
3
|
Yaghjyan L, Heng YJ, Baker GM, Bret-Mounet VC, Murthy D, Mahoney MB, Rosner B, Tamimi RM. Associations of reproductive breast cancer risk factors with expression of stem cell markers in benign breast tissue. Front Oncol 2024; 14:1354094. [PMID: 38577336 PMCID: PMC10991780 DOI: 10.3389/fonc.2024.1354094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Background We investigated the associations of reproductive factors known to influence breast cancer risk with the expression of breast stem cell markers CD44, CD24, and ALDH1A1 in benign breast biopsy samples. Methods We included 439 cancer-free women with biopsy-confirmed benign breast disease within the Nurses' Health Study (NHS) and NHSII. The data on reproductive and other breast cancer risk factors were obtained from biennial questionnaires. Immunohistochemistry (IHC) was performed on tissue microarrays. For each core, the IHC expression was assessed using a semi-automated platform and expressed as % of cells that stained positive for a specific marker out of the total cell count. Generalized linear regression was used to examine the associations of reproductive factors with a log-transformed expression of each marker (in epithelium and stroma), adjusted for other breast cancer risk factors. Results In multivariate analysis, the time between menarche and age at first birth was inversely associated with CD44 in epithelium (β per 5 years = -0.38, 95% CI -0.69; -0.06). Age at first birth and the time between menarche and age at first birth were inversely associated with ALDH1A1 (stroma: β per 5 years = -0.43, 95% CI -0.76; -0.10 and β = -0.47, 95% CI -0.79; -0.15, respectively; epithelium: β = -0.15, 95% CI -0.30; -0.01 and β = -0.17, 95% CI -0.30; -0.03, respectively). Time since last pregnancy was inversely associated with stromal ALDH1A1 (β per 5 years = -0.55, 95% CI -0.98; -0.11). No associations were found for CD24. The observed associations were similar in premenopausal women. In postmenopausal women, lifetime duration of breastfeeding was inversely associated with stromal ALDH1A1 expression (β for ≥24 vs. 0 to <1 months = -2.24, 95% CI 3.96; -0.51, p-trend = 0.01). Conclusion Early-life reproductive factors may influence CD44 and ALDH1A1 expression in benign breast tissue.
Collapse
Affiliation(s)
- Lusine Yaghjyan
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, United States
| | - Yujing J Heng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Gabrielle M Baker
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Vanessa C Bret-Mounet
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Divya Murthy
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Matt B Mahoney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Bernard Rosner
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
4
|
HPV Prevalence and Predictive Biomarkers for Oropharyngeal Squamous Cell Carcinoma in Mexican Patients. Pathogens 2022; 11:pathogens11121527. [PMID: 36558861 PMCID: PMC9783288 DOI: 10.3390/pathogens11121527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Worldwide prevalence of Oropharyngeal Squamous Cell Carcinoma (OPSCC) has increased, affecting mostly young males. OPSCC associated with Human Papillomavirus (HPV) infection exhibits particular characteristics in terms of response to treatment, hence HPV has been proposed as a prognostic factor. The impact of HPV positivity and associated biomarkers on OPSCC in the Mexican population has not been addressed. Therefore, the analysis of OPSCC prognostic markers in the Mexican population is necessary. METHODS Retrolective study in Mexican OPSCC patients, where HPV prevalence, p16 and EGFR levels were assessed using INNO-LiPA and immunohistochemistry. RESULTS We found an HPV prevalence of 57.6% in OPSCC cases treated at a reference center in Mexico. HPV and p16 positivity, as well as EGFR, associate with better outcomes in OPSCC patients, and they also promote reduced death risk. Notably, HPV presence and p16 positivity showed a significant association with disease-free survival (DFS), with a HR of 0.15 (p = 0.006) and a HR of 0.17 (p = 0.012), respectively, indicating a possible role as predictive biomarkers in Mexican OPSCC patients. CONCLUSIONS Our results reflect the clinical utility of p16 analysis to improve overall survival (OS) and to predict recurrence in oropharyngeal cancer. These results position p16 and HPV as predictive biomarkers for OPSCC.
Collapse
|
5
|
Conde E, Rojo F, Gómez J, Enguita AB, Abdulkader I, González A, Lozano D, Mancheño N, Salas C, Salido M, Salido-Ruiz E, de Álava E. Molecular diagnosis in non-small-cell lung cancer: expert opinion on ALK and ROS1 testing. J Clin Pathol 2022; 75:145-153. [PMID: 33875457 PMCID: PMC8862096 DOI: 10.1136/jclinpath-2021-207490] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/09/2023]
Abstract
The effectiveness of targeted therapies with tyrosine kinase inhibitors in non-small-cell lung cancer (NSCLC) depends on the accurate determination of the genomic status of the tumour. For this reason, molecular analyses to detect genetic rearrangements in some genes (ie, ALK, ROS1, RET and NTRK) have become standard in patients with advanced disease. Since immunohistochemistry is easier to implement and interpret, it is normally used as the screening procedure, while fluorescence in situ hybridisation (FISH) is used to confirm the rearrangement and decide on ambiguous immunostainings. Although FISH is considered the most sensitive method for the detection of ALK and ROS1 rearrangements, the interpretation of results requires detailed guidelines. In this review, we discuss the various technologies available to evaluate ALK and ROS1 genomic rearrangements using these techniques. Other techniques such as real-time PCR and next-generation sequencing have been developed recently to evaluate ALK and ROS1 gene rearrangements, but some limitations prevent their full implementation in the clinical setting. Similarly, liquid biopsies have the potential to change the treatment of patients with advanced lung cancer, but further research is required before this technology can be applied in routine clinical practice. We discuss the technical requirements of laboratories in the light of quality assurance programmes. Finally, we review the recent updates made to the guidelines for the determination of molecular biomarkers in patients with NSCLC.
Collapse
Affiliation(s)
- Esther Conde
- Department of Pathology and Laboratory of Therapeutic Targets & CIBERONC, HM Hospitales, Madrid, Spain
| | - Federico Rojo
- Department of Pathology, Hospital Universitario Fundacion Jiménez Díaz, Madrid, Spain
| | - Javier Gómez
- Department of Pathology, Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
- Instituto de Investigación Sanitaria Valdecilla IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Ana Belén Enguita
- Department of Pathology, Clínica Dermatológica Internacional, Madrid, Spain
| | - Ihab Abdulkader
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Ana González
- Department of Pathology, Hospital Álvaro Cunqueiro, Vigo, Spain
| | - Dolores Lozano
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Nuria Mancheño
- Department of Pathology, La Fe University and Polytechnic Hospital, Valencia, Comunidad Valenciana, Spain
| | - Clara Salas
- Department of Pathology, Hospital Universitario Puerta del Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Marta Salido
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Eduardo Salido-Ruiz
- Department of Pathology, Hospital Universitario de Canarias, La Laguna, Canarias, Spain
| | - Enrique de Álava
- Department of Pathology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
6
|
Lapoirie M, Dijoud F, Lejeune H, Plotton I. Effect of androgens on Sertoli cell maturation in human testis from birth to puberty. Basic Clin Androl 2021; 31:31. [PMID: 34906089 PMCID: PMC8670046 DOI: 10.1186/s12610-021-00150-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/21/2021] [Indexed: 11/11/2022] Open
Abstract
Background Androgens are well known to be necessary for spermatogenesis. The purpose of this study was to determine Sertoli cell responsiveness to androgens according to age from birth to puberty. Results Testicular tissue samples were studied in a population of 84 control boys classified into seven groups according to age: group 1 (1–30 days), group 2 (1–3 months), group 3 (3–6 months), group 4 (0.5–3 years), group 5 (3–6 years), group 6 (6–12 years), and group 7 (12–16 years). We compared these data with those of 2 situations of pathology linked to androgens: 1/premature secretion of testosterone: 4 cases of Leydig cell tumor (LCT) in childhood; and 2 /defect of androgen receptors (AR): 4 cases of complete form of insensitivity to androgen syndrome (CAIS). In control boys, AR immunoreactivity (ir) in Sertoli cells appeared between 4.6 and 10.8 years of age, Anti-Mullerian Hormone (AMH) ir in Sertoli cells disappeared between 9.2 and 10.2 years of age. Connexin 43 (Cx43) ir in Sertoli cells and histological features of the onset of spermatogenesis appeared between 10.8 and 13,8 years of age. Cx43 ir was significantly higher in 12–16 year-olds than in younger boys. In case of CAIS, no spermatogenesis was observed, both AR and Cx43 ir were undetectable and AMH ir was elevated in Sertoli cells even at pubertal age. In the vicinity of LCTs, spermatogenesis occurred and both AR and Cx43 ir were strongly positive and AMH ir in Sertoli cells was low for age. Conclusions Androgen action on Sertoli cells is required for onset of spermatogenesis and premature androgen secretion by LCT can induce spermatogenesis in the vicinity of the tumor. AR ir appeared earlier than onset of spermatogenesis, with large interindividual variability. The timing and mechanisms of Sertoli cell responsiveness to androgens are important issues for understanding the induction of spermatogenesis at puberty.
Collapse
Affiliation(s)
- Marion Lapoirie
- Université Claude Bernard Lyon 1, Lyon, France.,Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Femme Mère Enfant, Bron, France
| | - Frederique Dijoud
- Université Claude Bernard Lyon 1, Lyon, France.,Institut de pathologie Multisite des Hospices Civils de Lyon, Site Est, Boulevard Pinel, Bron, France.,Inserm, U1208, Bron, France
| | - Hervé Lejeune
- Université Claude Bernard Lyon 1, Lyon, France.,Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Femme Mère Enfant, Bron, France.,Inserm, U1208, Bron, France
| | - Ingrid Plotton
- Université Claude Bernard Lyon 1, Lyon, France. .,Inserm, U1208, Bron, France. .,Service de Biochimie et Biologie Moléculaire, Université Claude Bernard Lyon1, INSERM 1208, Groupement Hospitalier Est, Centre de Biologie et Pathologie Est, 59, Boulevard Pinel, 69677, Bron, Cedex, France.
| |
Collapse
|
7
|
Automated scoring of CerbB2/HER2 receptors using histogram based analysis of immunohistochemistry breast cancer tissue images. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Wharton KA, Wood D, Manesse M, Maclean KH, Leiss F, Zuraw A. Tissue Multiplex Analyte Detection in Anatomic Pathology - Pathways to Clinical Implementation. Front Mol Biosci 2021; 8:672531. [PMID: 34386519 PMCID: PMC8353449 DOI: 10.3389/fmolb.2021.672531] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Multiplex tissue analysis has revolutionized our understanding of the tumor microenvironment (TME) with implications for biomarker development and diagnostic testing. Multiplex labeling is used for specific clinical situations, but there remain barriers to expanded use in anatomic pathology practice. Methods: We review immunohistochemistry (IHC) and related assays used to localize molecules in tissues, with reference to United States regulatory and practice landscapes. We review multiplex methods and strategies used in clinical diagnosis and in research, particularly in immuno-oncology. Within the framework of assay design and testing phases, we examine the suitability of multiplex immunofluorescence (mIF) for clinical diagnostic workflows, considering its advantages and challenges to implementation. Results: Multiplex labeling is poised to radically transform pathologic diagnosis because it can answer questions about tissue-level biology and single-cell phenotypes that cannot be addressed with traditional IHC biomarker panels. Widespread implementation will require improved detection chemistry, illustrated by InSituPlex technology (Ultivue, Inc., Cambridge, MA) that allows coregistration of hematoxylin and eosin (H&E) and mIF images, greater standardization and interoperability of workflow and data pipelines to facilitate consistent interpretation by pathologists, and integration of multichannel images into digital pathology whole slide imaging (WSI) systems, including interpretation aided by artificial intelligence (AI). Adoption will also be facilitated by evidence that justifies incorporation into clinical practice, an ability to navigate regulatory pathways, and adequate health care budgets and reimbursement. We expand the brightfield WSI system “pixel pathway” concept to multiplex workflows, suggesting that adoption might be accelerated by data standardization centered on cell phenotypes defined by coexpression of multiple molecules. Conclusion: Multiplex labeling has the potential to complement next generation sequencing in cancer diagnosis by allowing pathologists to visualize and understand every cell in a tissue biopsy slide. Until mIF reagents, digital pathology systems including fluorescence scanners, and data pipelines are standardized, we propose that diagnostic labs will play a crucial role in driving adoption of multiplex tissue diagnostics by using retrospective data from tissue collections as a foundation for laboratory-developed test (LDT) implementation and use in prospective trials as companion diagnostics (CDx).
Collapse
|
9
|
Wijesinghe HD, Fernando J, Senarath U, Wijesinghe GK, S Lokuhetty MD. A clinicopathological study of triple-negative breast carcinoma in a patient cohort from a tertiary care center in Sri Lanka. INDIAN J PATHOL MICR 2021; 63:388-396. [PMID: 32769327 DOI: 10.4103/ijpm.ijpm_657_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background Triple negative breast carcinoma (TNBC) and basal-like breast carcinoma (BLBC) are subtypes of breast carcinoma (BCa) that are associated with poor survival. Aims To study the prevalence, clinicopathological profile and survival of TNBC among a Sri Lankan patient cohort and to determine the proportion and predictive histological features of BLBC among TNBCs. Study Setting and Design A cohort of 221 women undergoing primary surgery for BCa at a tertiary-care center in Sri Lanka was studied. Materials and Methods Clinicopathological and follow-up information were collected by patient interviews and review of slides and clinical records. Estrogen, progesterone, HER2 receptors, and basal markers (CK5/6, CK14, EGFR, 34βE12) were evaluated immunohistochemically. Statistical Analysis Data was analyzed with Chi-square test, multinomial logistic regression, and Cox regression using SPSS20.0. Results Fifty-three (24%) tumors were triple-negative (95%CI = 18.37%-29.63%). On multivariate analysis, young age (P = 0.002), high Nottingham grade (P = 0.005), moderate to severe tumor necrosis (P = 0.004), absent ductal carcinoma in situ (DCIS) (P = 0.04), reduced vascular density at tumor edge (P = 0.016) and distinct cell margins (P = 0.047) predicted TNBC over luminal subgroups, whereas reduced vascular density (P = 0.004) and low TNM stage (P = 0.011) distinguished TNBC and HER2. BLBC accounted for 45.28% (95%CI 32.66%-58.55%-24/53) of TNBC. The presence of extensive necrosis in TNBC correlated significantly with BLBC (P = 0.03). The survival among the TNBC subgroup did not differ significantly from other subgroups. Conclusion Twenty four percent were TNBCs by immunohistochemical analysis, comparable to studies in the Indian subcontinent, however higher than the West. TNBC status correlated with younger age, high tumor grade, necrosis, absent DCIS, reduced vascular density at tumor edge, and distinct cell margins. The presence of moderate to extensive necrosis in TNBC was predictive of BLBC.
Collapse
Affiliation(s)
| | - Janakie Fernando
- Department of Pathology, National Hospital of Sri Lanka, Sri Lanka
| | - Upul Senarath
- Department of Community Medicine, Faculty of Medicine, University of Colombo, Sri Lanka
| | - Gayani K Wijesinghe
- Department of Pathology, Faculty of Medicine, University of Colombo, Sri Lanka
| | | |
Collapse
|
10
|
Wang H, Xu Y, Jin M, Li H, Li S. miR-383 reduces keratinocyte proliferation and induces the apoptosis in psoriasis via disruption of LCN2-dependent JAK/STAT pathway activation. Int Immunopharmacol 2021; 96:107587. [PMID: 33819732 DOI: 10.1016/j.intimp.2021.107587] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/25/2022]
Abstract
Psoriasis is a chronic and relapsing disorder with considerable negative effects on patients' quality of life. The finer details associated with the molecular mechanism of psoriasis and its pathogenesis remain somewhat elusive. Extensive studies have highlighted the crucial role of microRNAs (miRNAs) in the development of psoriasis. Hence, the current study aimed to investigate the effect of miR-383 on a psoriasis rat model and elucidate the underlying molecular mechanism. The rat psoriasis model was established via imiquimod (IMQ) induction followed by verification of miR-383 and LCN2 expression in the skin tissues of the models. ELISA was conducted to determine the secretion of inflammatory factors. Keratinocyte proliferation and apoptosis was evaluated by MTT assay and flow cytometric analysis. Down-regulation of miR-383 and up-regulation of LCN2 were detected in the psoriasis rat model. Our data indicated that miR-383 targeted LCN2 by binding to its 3'UTR and inhibited JAK/STAT pathway activation. Notably, miR-383 overexpression or LCN2 knockdown attenuated psoriasis-like symptoms, suppressed inflammatory response, reduced the expression of JAK3 and STAT3, ceased keratinocyte proliferation, and promoted the apoptosis. The findings of our study suggest that miR-383 may inhibit LCN2 and inactivate the JAK/STAT pathway, suppressing the progression of psoriasis in a rat model. This study provided novel insights into the pathogenesis of psoriasis and offered potential targets for psoriasis treatment.
Collapse
Affiliation(s)
- Hong Wang
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China.
| | - Yangchun Xu
- Department of Dermatology, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, PR China
| | - Meishan Jin
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China
| | - Hongxia Li
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China
| | - Shanshan Li
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China.
| |
Collapse
|
11
|
Rosenkranz AA, Slastnikova TA. Epidermal Growth Factor Receptor: Key to Selective Intracellular Delivery. BIOCHEMISTRY (MOSCOW) 2021; 85:967-1092. [PMID: 33050847 DOI: 10.1134/s0006297920090011] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidermal growth factor receptor (EGFR) is an integral surface protein mediating cellular response to a number of growth factors. Its overexpression and increased activation due to mutations is one of the most common traits of many types of cancer. Development and clinical use of the agents, which block EGFR activation, became a prime example of the personalized targeted medicine. However, despite the obvious success in this area, cancer cure remains unattainable in most cases. Because of that, as well as the result of the search for possible ways to overcome the difficulties of treatment, a huge number of new treatment methods relying on the use of EGFR overexpression and its changes to destroy cancer cells. Modern data on the structure, functioning, and intracellular transport of EGFR, its natural ligands, as well as signaling cascades triggered by the EGFR activation, peculiarities of the EGFR expression and activation in oncological disorders, as well as applied therapeutic approaches aimed at blocking EGFR signaling pathway are summarized and analyzed in this review. Approaches to the targeted delivery of various chemotherapeutic agents, radionuclides, immunotoxins, photosensitizers, as well as the prospects for gene therapy aimed at cancer cells with EGFR overexpression are reviewed in detail. It should be noted that increasing attention is being paid nowadays to the development of multifunctional systems, either carrying several different active agents, or possessing several environment-dependent transport functions. Potentials of the systems based on receptor-mediated endocytosis of EGFR and their possible advantages and limitations are discussed.
Collapse
Affiliation(s)
- A A Rosenkranz
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia. .,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - T A Slastnikova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
12
|
Wan SY, Li GS, Tu C, Chen WL, Wang XW, Wang YN, Peng LB, Tan F. MicroNAR-194-5p hinders the activation of NLRP3 inflammasomes and alleviates neuroinflammation during intracerebral hemorrhage by blocking the interaction between TRAF6 and NLRP3. Brain Res 2021; 1752:147228. [PMID: 33385377 DOI: 10.1016/j.brainres.2020.147228] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
The possible role of miR-194-5p in brain and neurodegenerative diseases has been reported, but its role in intracerebral hemorrhage (ICH) has not been studied. This study estimated the mechanism of miR-194-5p in ICH. ICH rat model was established by injecting collagenase type VII. miR-194-5p expression in brain tissue of ICH rats was overexpressed by injection of miR-194-5p agomir. Then neurological function score and brain water content were measured. The morphological changes of brain tissue and neuronal apoptosis were evaluated by histological staining. Levels of NLRP3 inflammasomes, IL-1β and IL-18 were measured. The target relation between miR-194-5p and TRAF6 was verified and the binding of TRAF6 to NLRP3 was explored. miR-194-5p was decreased in ICH rats. After overexpression of miR-194-5p, the neuropathological injury in ICH rats was significantly reduced, and NLRP3-mediated inflammatory injury was inhibited. miR-194-5p targeted TRAF6. TRAF6 interacted with NLRP3 to promote the activation of NLRP3 inflammasomes. Overexpression of miR-194-5p reduced the interaction between TRAF6 and NLRP3, thereby alleviating the neuroinflammation. Collectively, overexpression of miR-194-5p reduced the TRAF6/NLRP3 interaction, thus inhibiting the activation of NLRP3 inflammasomes and reducing neuroinflammation during ICH. This study may shed new light on ICH treatment.
Collapse
Affiliation(s)
- Sai-Ying Wan
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Gui-Su Li
- Department of Neurology, Shenzhen Longhua District People's Hospital, China
| | - Chen Tu
- Department of Bone, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Wen-Lin Chen
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Xue-Wen Wang
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Yun-Nan Wang
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Lie-Biao Peng
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China
| | - Feng Tan
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine of Guangdong Province, China.
| |
Collapse
|
13
|
Meng Q, Zhang B, Zhang Y, Wang S, Zhu X. Human bone marrow mesenchymal stem cell-derived extracellular vesicles impede the progression of cervical cancer via the miR-144-3p/CEP55 pathway. J Cell Mol Med 2021; 25:1867-1883. [PMID: 33417281 PMCID: PMC7882924 DOI: 10.1111/jcmm.15573] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/04/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is the most common gynaecological malignancy, with a high incidence rate and mortality rate in middle‐aged women. Human bone marrow mesenchymal stem cells (hBMSCs) have been implicated in the initiation and subsequent development of cancer, along with the involvement of extracellular vesicles (EVs) mediating intracellular communication by delivering microRNAs (miRNAs or miRs). This study is aimed at investigating the physiological mechanisms by which EVs‐encapsulated miR‐144‐3p derived from hBMSCs might mediate the progression of cervical cancer. The expression profiles of centrosomal protein, 55 Kd (CEP55) and miR‐144‐3p in cervical cancer cell lines and tissues, were quantified by RT‐qPCR and Western blot analysis. The binding affinity between miR‐144‐3p and CEP55 was identified using in silico analysis and luciferase activity determination. Cervical cancer cells were co‐cultured with EVs derived from hBMSCs that were treated with either miR‐144‐3p mimic or miR‐144‐3p inhibitor. Cervical cancer cell proliferation, invasion, migration and apoptosis were detected in vitro. The effects of hBMSCs‐miR‐144‐3p on tumour growth were also investigated in vivo. miR‐144‐3p was down‐regulated, whereas CEP55 was up‐regulated in cervical cancer cell lines and tissues. CEP55 was targeted by miR‐144‐3p, which suppressed cervical cancer cell proliferation, invasion and migration and promoted apoptosis via CEP55. Furthermore, similar results were obtained by hBMSCs‐derived EVs carrying miR‐144‐3p. In vivo assays confirmed the tumour‐suppressive effects of miR‐144‐3p in hBMSCs‐derived EVs on cervical cancer. Collectively, hBMSCs‐derived EVs‐loaded miR‐144‐3p impedes the development and progression of cervical cancer through target inhibition of CEP55, therefore providing us with a potential therapeutic target for treating cervical cancer.
Collapse
Affiliation(s)
- Qin Meng
- Department of Obstetrics and Gynecology, Shandong Medical College, Linyi, P. R. China
| | - Baofang Zhang
- Department of Physical Examination, The Third People's Hospital of Linyi, Linyi, P. R. China
| | - Yingming Zhang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, P. R. China
| | - Shuyan Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, P. R. China
| | - Xiaohui Zhu
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, P. R. China
| |
Collapse
|
14
|
Elsharawy KA, Althobiti M, Mohammed OJ, Aljohani AI, Toss MS, Green AR, Rakha EA. Nucleolar protein 10 (NOP10) predicts poor prognosis in invasive breast cancer. Breast Cancer Res Treat 2020; 185:615-627. [PMID: 33161513 PMCID: PMC7920889 DOI: 10.1007/s10549-020-05999-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/26/2020] [Indexed: 01/16/2023]
Abstract
Purpose Nucleolar protein 10 (NOP10) is required for ribosome biogenesis and telomere maintenance and plays a key role in carcinogenesis. This study aims to evaluate the clinical and prognostic significance of NOP10 in breast cancer (BC). Methods NOP10 expression was assessed at mRNA level employing the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) (n = 1980) and Cancer Genome Atlas (TCGA) BC cohorts (n = 854). Protein expression was evaluated on tissue microarray of a large BC cohort (n = 1081) using immunohistochemistry. The correlation between NOP10 expression, clinicopathological parameters and patient outcome was assessed. Results NOP10 expression was detected in the nucleus and nucleolus of the tumour cells. At the transcriptomic and proteomic levels, NOP10 was significantly associated with aggressive BC features including high tumour grade, high nucleolar score and poor Nottingham Prognostic Index. High NOP10 protein expression was an independent predictor of poor outcome in the whole cohort and in triple-negative BC (TNBC) class (p = 0.002 & p = 0.014, respectively). In chemotherapy- treated patients, high NOP10 protein expression was significantly associated with shorter survival (p = 0.03) and was predictive of higher risk of death (p = 0.028) and development of distant metastasis (p = 0.02) independent of tumour size, nodal stage and tumour grade. Conclusion High NOP10 expression is a poor prognostic biomarker in BC and its expression can help in predicting chemotherapy resistance. Functional assessments are necessary to decipher the underlying mechanisms and to reveal its potential therapeutic values in various BC subtypes especially in the aggressive TNBC class. Electronic supplementary material The online version of this article (doi:10.1007/s10549-020-05999-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Khloud A Elsharawy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK.,Faculty of Science, Damietta University, Damietta, Egypt
| | - Maryam Althobiti
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK.,Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra University, Shaqra, Saudi Arabia
| | - Omar J Mohammed
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Abrar I Aljohani
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK. .,Division of Cancer and Stem Cell, University of Nottingham, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
15
|
Silencing of SPARC represses heterotopic ossification via inhibition of the MAPK signaling pathway. Biosci Rep 2020; 39:BSR20191805. [PMID: 31548362 PMCID: PMC6851515 DOI: 10.1042/bsr20191805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/08/2019] [Accepted: 09/18/2019] [Indexed: 11/17/2022] Open
Abstract
Heterotopic ossification (HO), the pathologic formation of extraskeletal bone, can be disabling and lethal. However, the underlying molecular mechanisms were largely unknown. The present study aimed to clarify the involvement of secreted protein acidic and rich in cysteine (SPARC) and the underlying mechanism in rat model of HO. The mechanistic investigation on roles of SPARC in HO was examined through gain- and loss-of-function approaches of SPARC, with alkaline-phosphatase (ALP) activity, mineralized nodules, and osteocalcin (OCN) content measured. To further confirm the regulatory role of SPARC, levels of mitogen-activated protein kinase (MAPK) signaling pathways-related proteins (extracellular signal-regulated kinase (ERK), c-jun N-terminal kinase (JNK), p38, nuclear factor κ-B (NF-κB), and IkB kinase β (IKKβ)) were determined. Bone marrow mesenchymal stem cells were treated with pathway inhibitor to investigate the relationship among SPARC, MAPK signaling pathway, and HO. The results suggested that SPARC expression was up-regulated in Achilles tendon tissues of HO rats. Silencing of SPARC could decrease phosphorylation of ERK, JNK, p38, NF-κB, and IKKβ. Additionally, silencing of SPARC or inhibition of MAPK signaling pathway could reduce the ALP activity, the number of mineralized nodules, and OCN content, thus impeding HO. To sum up, our study identifies the inhibitory role of SPARC gene silencing in HO via the MAPK signaling pathway, suggesting SPARC presents a potential target for HO therapy.
Collapse
|
16
|
Wijesinghe HD, Wijesinghe GK, Mansoor Z, Vigneshwara S, Fernando J, Gunasekera D, Lokuhetty MDS. Androgen receptor expression in a Sri Lankan patient cohort with early breast carcinoma. BMC Womens Health 2020; 20:206. [PMID: 32928183 PMCID: PMC7490868 DOI: 10.1186/s12905-020-01068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 09/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Androgen receptor (AR) expression is emerging as a prognostic biomarker in breast carcinoma (BCa). The study aimed to determine the prevalence of AR expression by immunohistochemical analysis among a cohort of Sri Lankan women with early BCa and to evaluate its association with clinicopathological features including immunohistochemical molecular subtype and early survival. METHOD We studied the clinical and pathological features and immunohistochemical profile of 141 women undergoing primary surgery for early BCa, followed by standard adjuvant therapy. AR status was assessed by immunohistochemistry in all cases. Overall survival (OS) and disease-free survival (DFS) was determined. The relationship between AR expression and clinical and pathological parameters and immunohistochemical molecular subtype was analyzed using Student T test and chi-square tests. Cox regression analysis was used to analyze the prognostic impact of AR expression. RESULTS AR expression was seen in 40.8%(95%CI 33.10-49.07%) of the BCa study cohort. None of the clinical data studied showed a significant association with the AR status(p > 0.05). Ductal carcinoma in situ(p = 0.003), oestrogen receptor (ER) (p = 0.001) and progesterone receptor (PR) (p = 0.001) positivity and luminal IHC molecular subtype(p = 0.016) were significantly associated with AR-positive status. AR-negative status was significantly associated with tumour necrosis > 50%(p = 0.031), moderate to extensive lymphocytic infiltrate at the tumour margin(p = 0.025) and basal triple negative breast carcinoma(p = 0.016). The mean duration of patient follow-up was 46.70(95% CI 46.495-46.905) months (3.89 years). On univariate analysis, AR-positivity was associated with better OS among ER-positive tumours(p = 0.047), specifically in postmenopausal women (p = 0.030). In ER-negative tumours, AR positivity was associated with worse DFS (p = 0.036). On multivariate analysis, TNM stage and ER/AR status were predictive of survival. ER-positive/AR-positive (ER+/AR+) tumours demonstrated better OS than ER-positive/AR-negative (ER+/AR-) tumours(p = 0.015). ER-negative/AR-positive (ER-/AR+) tumours (p = 0.014) had a worse DFS than ER-negative/AR-negative (ER-/AR-) tumours. CONCLUSIONS AR prevalence obtained was low. AR positivity was associated with positivity for ER and PR. On multivariate analysis, apart from TNM stage only ER/AR status were predictive of OS and DFS, with concordant expression of ER/AR demonstrating a better, early survival.
Collapse
Affiliation(s)
| | | | - Zahara Mansoor
- Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Sanjeev Vigneshwara
- Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Janakie Fernando
- Department of Pathology, National of Hospital of Sri Lanka, Colombo, Sri Lanka
| | | | | |
Collapse
|
17
|
He Z, Li W, Zheng T, Liu D, Zhao S. Human umbilical cord mesenchymal stem cells-derived exosomes deliver microRNA-375 to downregulate ENAH and thus retard esophageal squamous cell carcinoma progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:140. [PMID: 32698859 PMCID: PMC7374920 DOI: 10.1186/s13046-020-01631-w] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/29/2020] [Indexed: 01/07/2023]
Abstract
Background Exosomal microRNAs (miRNAs or miRs) from bone marrow-derived mesenchymal stem cells (UCMSCs) have emerged as promising therapeutic strategies for cancer treatment. The current study aimed to elucidate the underlying mechanism of human umbilical cord mesenchymal stem cells (hUCMSCs)-derived exosomal miR-375 in esophageal squamous cell carcinoma (ESCC). Methods After determining the expression of miR-375 and its putative target enabled homolog (ENAH) in ESCC tissues and cells, we tested effects of their altered expression on ESCC proliferation, invasion, migration, and tumorsphere formation was subsequently measured. Transfected hUCMSCs-derived exosomes (hUCMSCs-exo) were isolated and co-cultured with ESCC cells to measure the effects of miR-375 delivered by hUCMSCs-exo on ESCC development. Finally, we investigated the effect of miR-375 on tumor growth in vivo. Results The expression of miR-375 was reduced, while the expression of ENAH was elevated in ESCC. ENAH was identified as a target gene of miR-375. Elevated miR-375 or depleted ENAH expression inhibited ESCC cell proliferation, invasion, migration, tumorsphere formation, and promoted apoptosis. Moreover, miR-375 delivered by hUCMSCs-exo could suppress ESCC cell proliferation, invasion, migration, tumorsphere formation, but promoted apoptosis in vitro, as well as inhibiting tumor growth in vivo. Conclusions Taken together, hUCMSCs-exo can deliver miR-375 to suppress ENAH expression and subsequently inhibit the initiation and progression of ESCC.
Collapse
Affiliation(s)
- Zhanfeng He
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Weihao Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Tianliang Zheng
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Donglei Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
18
|
Wang C, Yin W, Liu H. MicroRNA-10a promotes epithelial-to-mesenchymal transition and stemness maintenance of pancreatic cancer stem cells via upregulating the Hippo signaling pathway through WWC2 inhibition. J Cell Biochem 2020; 121:4505-4521. [PMID: 32542845 DOI: 10.1002/jcb.29716] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/14/2020] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs)-mediated cancer stem cells (CSCs) have drawn wide attention. This study aimed to probe the role of miR-10a in epithelial-mesenchymal transition (EMT) and stemness maintenance of pancreatic CSCs (PCSCs). Differentially expressed miRs and genes in pancreatic cancer (PC) were predicted via an online database, and the miR-10a and WW and C2 domain containing 2 (WWC2) expression were identified via a comparative study in PC and pancreatitis tissues. PCNCs were isolated and identified, and then the functional roles of miR-10a and WWC2 in proliferation, invasion, migration, self-renewal, colony formation abilities, EMT, and stemness maintenance of PCNCs were determined. The effects of miR-10a on tumor growth in vivo were studied by performing a xenograft tumor in nude mice. Consequently, miR-10a was highly expressed while WWC2 was lowly expressed in PC tissues. miR-10a could target WWC2 expression. miR-10a inhibition reduced EMT and stemness maintenance of PCSCs via enhancing WWC2 expression. The in vitro results were reproduced in in vivo studies. miR-10a promoted EMT and stemness maintenance of PCSCs via activating the Hippo signaling pathway. Our study provided evidence that miR-10a inhibition reduced EMT and stemness maintenance of PCSCs via upregulating WWC2 expression and inhibiting the Hippo signaling pathway.
Collapse
Affiliation(s)
- Caiyan Wang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Wen Yin
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Hui Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
19
|
Yuan YF, Zhu WX, Liu T, He JQ, Zhou Q, Zhou X, Zhang X, Yang J. Cyclopamine functions as a suppressor of benign prostatic hyperplasia by inhibiting epithelial and stromal cell proliferation via suppression of the Hedgehog signaling pathway. Int J Mol Med 2020; 46:311-319. [PMID: 32319534 PMCID: PMC7255449 DOI: 10.3892/ijmm.2020.4569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 03/12/2019] [Indexed: 12/17/2022] Open
Abstract
Stromal-epithelial interaction serves a pivotal role in normal prostate growth, as well as the onset of benign prostatic hyperplasia (BPH). The present study aimed to explore the role of cyclopamine in the proliferation and apoptosis of epithelial and stromal cells in rats with BPH by blocking the Hedgehog signaling pathway. Cyclopamine (an inhibitor of the Hedgehog signaling pathway) was administered in a rat model of BPH, and the expression of Ki67 (proliferation factor) was determined by immunohistochemistry. In addition, epithelial and stromal cells were separated and cultured in order to investigate the role of cyclopamine in the progression of BPH. The expression of Hedgehog signaling pathway- and apoptosis-related genes, including basic fibroblastic growth factor (b-FGF) and transforming growth factor β (TGF-β), was evaluated using reverse transcription-quantitative polymerase chain reaction and western blot analysis. Cell proliferation, cell cycle and apoptosis were analyzed using an MTT assay and flow cytometry. We identified upregulated Ki67 expression and activated Hedgehog signaling pathway in rats with BPH. Cyclopamine inhibited the activation of the Hedgehog signaling pathway. In response to cyclopamine treatment, epithelial and stromal cell proliferation was inhibited; this was concomitant with decreased Ki67, TGF-β, and b-FGF expression. On the other hand, epithelial cell apoptosis was enhanced, which was associated with increased Bax and reduced Bcl-2 expression. Based on these findings, we proposed that cyclopamine may serve as a potential therapeutic agent in the treatment of BPH. Cyclopamine could inhibit epithelial and stromal cell proliferation, and induce epithelial cell apoptosis by suppressing the Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Yi-Feng Yuan
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Wen-Xiong Zhu
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Tao Liu
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Ju-Qiao He
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Qing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xi Zhang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jing Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
20
|
Fei L, Wang Y. microRNA‐495 reduces visceral sensitivity in mice with diarrhea‐predominant irritable bowel syndrome through suppression of the PI3K/AKT signaling pathway via PKIB. IUBMB Life 2020; 72:1468-1480. [PMID: 32187820 DOI: 10.1002/iub.2270] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/16/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Lifeng Fei
- Department of PhysiotherapyLinyi People's Hospital Linyi China
| | - Yanjing Wang
- Department of Children's RehabilitationLinyi People's Hospital Linyi China
| |
Collapse
|
21
|
Wang H, Wei H, Wang J, Li L, Chen A, Li Z. MicroRNA-181d-5p-Containing Exosomes Derived from CAFs Promote EMT by Regulating CDX2/HOXA5 in Breast Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:654-667. [PMID: 31955007 PMCID: PMC6970169 DOI: 10.1016/j.omtn.2019.11.024] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/22/2019] [Indexed: 12/24/2022]
Abstract
Recently, novel mechanisms underlying the pro-tumorigenic effects of cancer-associated fibroblasts (CAFs) have been identified in several cancers, including breast cancer. CAFs can secrete exosomes that are loaded with proteins, lipids, and RNAs to affect tumor microenvironment. Herein, we identify CAF-derived exosomes that can transfer miR-181d-5p to enhance the aggressiveness of breast cancer. Cancerous tissues and matched paracancerous tissues were surgically resected from 122 patients with breast cancer. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter assays were employed to identify interaction between homeobox A5 (HOXA5) and caudal-related homeobox 2 (CDX2), as well as between CDX2 and miR-181d-5p, respectively. Human breast cancer Michigan Cancer Foundation-7 (MCF-7) cells were cocultured with CAF-derived exosomes. 5-Ethynyl-2'-deoxyuridine (EdU) assay, TUNEL staining, Transwell invasion assays, and scratch tests were carried out to evaluate MCF-7 cell functions. Nude mice bearing xenografted MCF-7 cells were injected with CAF-derived exosomes, and the tumor formation was evaluated. HOXA5 expressed at a poor level in breast cancer tissues, and its overexpression retarded MCF-7 cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) and facilitated its apoptosis in vitro. miR-181d-5p targets CDX2, a transcription factor binding to HOXA5 promoter. Coculture of CAFs and MCF-7 cells showed that CAFs prolonged proliferation and antagonized apoptosis of MCF-7 cells via release of exosomes. Coculture of MCF-7 cells and exosomes derived from CAFs identified miR-181d-5p as a mediator of the exosomal effects on MCF-7 cells, in part, via downregulation of CDX2 and HOXA5. CAF-derived exosomes containing miR-181d-5p promoted the tumor growth of nude mice bearing xenografted MCF-7 cells. In conclusion, exosomal miR-181d-5p plays a key role in CAF-mediated effects on tumor environment in breast cancer, likely via CDX2 and HOXA5.
Collapse
Affiliation(s)
- Hongbin Wang
- The Second Ward, Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Hong Wei
- In-Patient Department of Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Jingsong Wang
- The Second Ward, Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Lin Li
- The Second Ward, Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Anyue Chen
- The Second Ward, Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Zhigao Li
- The Second Ward, Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China.
| |
Collapse
|
22
|
Pang X, Lin X, Du J, Zeng D. LTBP2 knockdown by siRNA reverses myocardial oxidative stress injury, fibrosis and remodelling during dilated cardiomyopathy. Acta Physiol (Oxf) 2020; 228:e13377. [PMID: 31512380 DOI: 10.1111/apha.13377] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/19/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022]
Abstract
AIM Dilated cardiomyopathy (DCM) is characterised by left ventricular dilation and associated with systolic dysfunction. Recent evidence has reported the high expression of latent transforming growth factor beta binding protein 2 (LTBP2) in heart diseases, which may play a role in regulating multiple biological functions of myocardial cells. Thus, this study set out to investigate the molecular mechanism and effects of LTBP2 in myocardial oxidative stress injury, fibrosis and remodelling in a rat model of DCM, with the involvement of NF-κB signalling pathway. METHODS The rat model of DCM was treated with si-LTBP2 and/or activator of NF-κB signalling pathway to examine the haemodynamic indexes, cardiac functions, oxidative stress injury, fibrosis and remodelling. Moreover, in vitro experiments were conducted to verify the regulatory role of LTBP2 and NF-κB signalling pathway in DCM. RESULTS LTBP2 was up-regulated in DCM rats. After LTBP2 was knocked down, haemodynamic indexes, HW/BW ratio, collagen volume fraction (CVF) level, positive expression of LTBP2, levels of reactive oxygen species (ROS), malondialdehyde (MDA), interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α), tumour necrosis factor beta 1 (TGF-β1) and brain natriuretic peptide (BNP) were all decreased. Meanwhile, levels of LTBP2, Col-I, Col-III, p65 and p52 were also reduced, while HW, BW and levels of SOD and TAOC were increased. In contrast, activation of NF-κB signalling pathway reversed effects of LTBP2 gene silencing. These findings were confirmed by in vivo experiments. CONCLUSIONS LTBP2 silencing can attenuate myocardial oxidative stress injury, myocardial fibrosis and myocardial remodelling in DCM rats by down-regulating the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Xue‐Feng Pang
- Department of Cardiovascular The First Hospital of China Medical University Shenyang China
| | - Xue Lin
- Department of Cardiovascular Peking Union Medical College Hospital Beijing China
| | - Jian‐Jun Du
- Department of Cardiovascular The First Hospital of China Medical University Shenyang China
| | - Ding‐Yin Zeng
- Department of Cardiovascular The First Hospital of China Medical University Shenyang China
| |
Collapse
|
23
|
Ghosh C, Bhowmik J, Ghosh R, Das MC, Sandhu P, Kumari M, Acharjee S, Daware AV, Akhter Y, Banerjee B, De UC, Bhattacharjee S. The anti-biofilm potential of triterpenoids isolated from Sarcochlamys pulcherrima (Roxb.) Gaud. Microb Pathog 2020; 139:103901. [PMID: 31790796 DOI: 10.1016/j.micpath.2019.103901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/23/2019] [Accepted: 11/28/2019] [Indexed: 10/25/2022]
Abstract
Formation of biofilm is the major cause of Pseudomonas aeruginosa associated pathological manifestations in the urinary tract, respiratory system, gastrointestinal tract, skin, soft tissues etc. Triterpenoid group of compounds have shown their potential in reducing planktonic and biofilm form of bacteria. Sarcochlamys pulcherrima (Roxb.) Gaud. is an ethnomedicinal plant traditionally used for its anti-microbial and anti-inflammatory property. In the present study two triterpenoids, have been isolated from this plant, characterised and evaluated for their antibacterial and antibiofilm potential against P. aeruginosa. Compounds were characterised as 2α, 3β, 19α-trihydroxy-urs-12-ene-28-oic acid (Tormentic acid) and 2α, 3β, 23-trihydroxyurs-12-ene-28-oic acid (23-hydroxycorosolic acid) through spectroscopic studies viz. infrared (IR), nuclear magnetic resonance (NMR) and mass spectroscopy (MS). Depolarization of bacterial membrane and zone of inhibition studies revealed that both the compounds inhibited the growth of planktonic bacteria. Compounds were also found to inhibit the formation of P. aeruginosa biofilm. Inhibition of biofilm found to be mediated through suppressed secretion of pyoverdin, protease and swarming motility of P. aeruginosa. Gene expression study, in silico binding analysis, in vivo bacterial load and tissue histology observations also supported the antibiofilm activity of both the compounds. In vitro and in vivo study showed that both compounds were non-toxic. The study has explored the antibacterial and antibiofilm effect of two triterpenes isolated for the first time from S. pulcherrima.
Collapse
Affiliation(s)
- Chinmoy Ghosh
- Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, 799022, Tripura, India; Molecular Stress and Stem Cell Biology Lab, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, Odisha, India
| | - Joyanta Bhowmik
- Department of Chemistry, Tripura University, Suryamaninagar, 799022, Tripura, India
| | - Ranjit Ghosh
- Department of Chemistry, Tripura University, Suryamaninagar, 799022, Tripura, India
| | - Manash C Das
- Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, 799022, Tripura, India
| | - Padmani Sandhu
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur, District-Kangra, Himachal Pradesh, 176206, India
| | - Monika Kumari
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur, District-Kangra, Himachal Pradesh, 176206, India
| | - Shukdeb Acharjee
- Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, 799022, Tripura, India
| | - Akshay Vishnu Daware
- Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, 799022, Tripura, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Birendranath Banerjee
- Molecular Stress and Stem Cell Biology Lab, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, Odisha, India
| | - Utpal Chandra De
- Department of Chemistry, Tripura University, Suryamaninagar, 799022, Tripura, India.
| | - Surajit Bhattacharjee
- Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, 799022, Tripura, India.
| |
Collapse
|
24
|
PFA is superior to glyoxal in preserving oocyte, embryo, and stem cell proteins evidenced by super-resolution microscopical surveys of epitopes. J Assist Reprod Genet 2020; 37:369-384. [PMID: 31930433 DOI: 10.1007/s10815-019-01666-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/12/2019] [Indexed: 10/25/2022] Open
Abstract
PURPOSE Chemical fixation is a critical step to retaining cellular targets as naturally as possible. Recent developments in microscopy allow sophisticated detection and measuring techniques with which spatio-temporal molecular alterations are conceivable. In this study, we compare two members of aldehyde fixatives [i.e., glyoxal (Gly) and paraformaldehyde (PFA)] to determine whether Gly, a less toxic dialdehyde fixative that is considered to retain immunoreactivity could provide a successful and consistent cell fixation in favor of PFA in various cell preparations and types. METHODS We document the fixation competence of Gly and PFA side-by-side (with or without Triton X-100 permeabilization) in live- and fixed-cell preparations in mouse oocytes, embryos, and human somatic cells (human umbilical cord-derived mesenchymal stromal cells) using protein quantification by Western blot assay and super-resolution microscopy. RESULTS Although Gly seemed to act faster than PFA, catastrophic consequences were found not acceptable, especially in oocytes and embryos. Due to cell lysate and immunocytochemistry surveys, it was obvious that PFA is superior to Gly in retaining cellular proteins in situ with little/no background staining. In many samples, PFA revealed more reliable and consistent results regarding the protein quantity and cellular localization corresponding to previously defined patterns in the literature. CONCLUSION Although the use of Gly is beneficial as indicated by previous reports, we concluded that it does not meet the requirement for proper fixation, at least for the tested cell types and proteins. However, PFA alone with no addition of TX displayed a significant cytoplasmic loss by generating membrane blebs during fixation.
Collapse
|
25
|
Wang Y, Sun S, Luo J, Xiong Y, Ming T, Liu J, Ma Y, Yan S, Yang Y, Yang Z, Reboud J, Yin H, Cooper JM, Cai X. Low sample volume origami-paper-based graphene-modified aptasensors for label-free electrochemical detection of cancer biomarker-EGFR. MICROSYSTEMS & NANOENGINEERING 2020; 6:32. [PMID: 34567646 PMCID: PMC8433370 DOI: 10.1038/s41378-020-0146-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/04/2019] [Accepted: 02/18/2020] [Indexed: 05/08/2023]
Abstract
In this work, an electrochemical paper-based aptasensor was fabricated for label-free and ultrasensitive detection of epidermal growth factor receptor (EGFR) by employing anti-EGFR aptamers as the bio-recognition element. The device used the concept of paper-folding, or origami, to serve as a valve between sample introduction and detection, so reducing sampling volumes and improving operation convenience. Amino-functionalized graphene (NH2-GO)/thionine (THI)/gold particle (AuNP) nanocomposites were used to modify the working electrode not only to generate the electrochemical signals, but also to provide an environment conducive to aptamer immobilization. Electrochemical characterization revealed that the formation of an insulating aptamer-antigen immunocomplex would hinder electron transfer from the sample medium to the working electrode, thus resulting in a lower signal. The experimental results showed that the proposed aptasensor exhibited a linear range from 0.05 to 200 ngmL-1 (R 2 = 0.989) and a detection limit of 5 pgmL-1 for EGFR. The analytical reliability of the proposed paper-based aptasensor was further investigated by analyzing serum samples, showing good agreement with the gold-standard enzyme-linked immunosorbent assay.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Shuai Sun
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Ying Xiong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142 China
| | - Tao Ming
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| | - Yuanyuan Ma
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142 China
| | - Shi Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142 China
| | - Yue Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142 China
| | - Zhugen Yang
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Oakfield Avenue, Glasgow, G12 8LT United Kingdom
| | - Julien Reboud
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Oakfield Avenue, Glasgow, G12 8LT United Kingdom
| | - Huabing Yin
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Oakfield Avenue, Glasgow, G12 8LT United Kingdom
| | - Jonathan M. Cooper
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Oakfield Avenue, Glasgow, G12 8LT United Kingdom
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Institute of Electronics, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100190 China
| |
Collapse
|
26
|
Shang D, Xie C, Hu J, Tan J, Yuan Y, Liu Z, Yang Z. Pancreatic cancer cell-derived exosomal microRNA-27a promotes angiogenesis of human microvascular endothelial cells in pancreatic cancer via BTG2. J Cell Mol Med 2020; 24:588-604. [PMID: 31724333 PMCID: PMC6933412 DOI: 10.1111/jcmm.14766] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 08/14/2019] [Accepted: 09/17/2019] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer (PC) remains a primary cause of cancer-related deaths worldwide. Existing literature has highlighted the oncogenic role of microRNA-27a (miR-27a) in multiple cancers. Hence, the current study aimed to clarify the potential therapeutic role of PC cell-derived exosomal miR-27a in human microvascular endothelial cell (HMVEC) angiogenesis in PC. Initially, differentially expressed genes (DEGs) and miRs related to PC were identified by microarray analysis. Microarray analysis provided data predicting the interaction between miR-27a and BTG2 in PC, which was further verified by the elevation or depletion of miR-27a. Next, the expression of miR-27a and BTG2 in the PC tissues was quantified. HMVECs were exposed to exosomes derived from PC cell line PANC-1 to investigate the effects associated with PC cell-derived exosomes carrying miR-27a on HMVEC proliferation, invasion and angiogenesis. Finally, the effect of miR-27a on tumorigenesis and microvessel density (MVD) was analysed after xenograft tumour inoculation in nude mice. Our results revealed that miR-27a was highly expressed, while BTG2 was poorly expressed in both PC tissues and cell lines. miR-27a targeted BTG2. Moreover, miR-27a silencing inhibited PC cell proliferation and invasion, and promoted apoptosis through the elevation of BTG2. The in vitro assays revealed that PC cell-derived exosomes carrying miR-27a stimulated HMVEC proliferation, invasion and angiogenesis, while this effect was reversed in the HMVECs cultured with medium containing GW4869-treated PANC-1 cells. Furthermore, in vivo experiment revealed that miR-27a knockdown suppressed tumorigenesis and MVD. Taken together, cell-derived exosomes carrying miR-27a promotes HMVEC angiogenesis via BTG2 in PC.
Collapse
Affiliation(s)
- Dan Shang
- Department of Vascular SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chao Xie
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Pancreatic Surgery CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jin Hu
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jinru Tan
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zhisu Liu
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Pancreatic Surgery CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
27
|
Yuan L, Liu Y, Qu Y, Liu L, Li H. Exosomes Derived From MicroRNA-148b-3p-Overexpressing Human Umbilical Cord Mesenchymal Stem Cells Restrain Breast Cancer Progression. Front Oncol 2019; 9:1076. [PMID: 31696054 PMCID: PMC6817568 DOI: 10.3389/fonc.2019.01076] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 09/30/2019] [Indexed: 01/08/2023] Open
Abstract
Exosomes derived from human umbilical cord mesenchymal stem cells (HUCMSCs) expressing microRNAs (miRs) have been highlighted as important carriers for gene or drug therapy. Hence, this study aimed to explore the role of exosomal miR-148b-3p from HUCMSCs in breast cancer. Clinical samples subjected to RT-qPCR detection revealed that miR-148b-3p was poorly expressed, while tripartite motif 59 (TRIM59) was highly expressed in breast cancer tissues. Online analyses available at miRanda, TargetScan, and miRbase databases revealed that miR-148b-3p could bind to TRIM59, while dual-luciferase reporter gene assay further verified that TRIM59 was a target gene of miR-148b-3p. Next, miR-148b-3p mimic or inhibitor and siRNA against TRIM59 were delivered into the breast cancer cells (MDA-MB-231) to alter the expression of miR-148b-3p and TRIM59 so as to evaluate their respective effects on breast cancer cellular processes. Evidence was obtained demonstrating that miR-148b-3p inhibited cell proliferation, invasion, and migration, but promoted cell apoptosis in breast cancer by down-regulating TRIM59. Next, MDA-MB-231 cells were co-cultured with the exosomes derived from HUCMSCs expressing miR-148b-3p. The results of co-culture experiments demonstrated that HUCMSCs-derived exosomes carrying miR-148b-3p exerted inhibitory effects on MDA-MB-231 progression in vitro. In vivo experimentation further confirmed the anti-tumor effects of HUCMSCs-derived exosomes carrying miR-148b-3p. Taken together, HUCMSC-derived exosomes carrying miR-148b-3p might suppress breast cancer progression, which highlights the potential of exosomes containing miR-148b-3p as a promising therapeutic approach for breast cancer treatment.
Collapse
Affiliation(s)
- Lei Yuan
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuqiong Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunhui Qu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huixiang Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Zeng SG, Lin X, Liu JC, Zhou J. Hypoxia‑induced internalization of connexin 26 and connexin 43 in pulmonary epithelial cells is involved in the occurrence of non‑small cell lung cancer via the P53/MDM2 signaling pathway. Int J Oncol 2019; 55:845-859. [PMID: 31485592 PMCID: PMC6741836 DOI: 10.3892/ijo.2019.4867] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Reports have highlighted an association between connexins (CXs) or gap junction proteins and non-small cell lung cancer (NSCLC). In the present study, it was aimed to elucidate the regulatory mechanism of CX26 and CX43 under hypoxic conditions in NSCLC. Clinical samples were collected for analysis of CX26 and CX43 expression and clinical cancerization followed by quantification of CX26 and CX43 expression. Following the establishment of an in vitro hypoxia model, P53/murine double minute-2 (MDM2) signaling pathway-, proliferation- and epithelial-mesenchymal transition (EMT)-related genes were quantified to evaluate the influence of CX26 and CX43 on the biological functions of pulmonary epithelial cells in NSCLC. In addition, the proliferation and tumorigenicity of cancer cells were assessed by EdU staining and xenograft tumors, respectively. Decreased expression of CX26 and CX43 was found in cancer tissues compared with surrounding normal tissue. Hypoxia was shown to activate the P53/MDM2 axis and stimulate the downregulation, ubiquitination and degradation of CX26 and CX43, which were translocated from the membrane to the cytoplasm. Low levels of CX26 and CX43 were demonstrated to further promote EMT and the induction of the proliferation and tumorigenicity of cancer cells. These results were reflected by decreased E-cadherin expression and increased N-cadherin expression, along with increased cell migration, promoted cell proliferation ability and elevated relative protein expression of Oct4 and Nanog, and accelerated tumor growth, accompanied by a higher number of metastatic nodes. Taken together, the key observations of the present study demonstrate that the internalization of CX26 and CX43 promoted proliferation, EMT and migration and thus induced NSCLC via aberrant activation of the P53/MDM2 signaling pathway under hypoxic conditions.
Collapse
Affiliation(s)
- Shang-Gan Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiang Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji-Chun Liu
- Departments of Cardio‑Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jin Zhou
- Department of Respiratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
29
|
CDC20 expression in oestrogen receptor positive breast cancer predicts poor prognosis and lack of response to endocrine therapy. Breast Cancer Res Treat 2019; 178:535-544. [DOI: 10.1007/s10549-019-05420-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/23/2022]
|
30
|
Zhang YP, Liu KL, Yang Z, Lu BS, Qi JC, Han ZW, Yin YW, Zhang M, Chen DM, Wang XW, Li W, Xin H. The involvement of FBP1 in prostate cancer cell epithelial mesenchymal transition, invasion and metastasis by regulating the MAPK signaling pathway. Cell Cycle 2019; 18:2432-2446. [PMID: 31448674 DOI: 10.1080/15384101.2019.1648956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is a frequently occurring malignancy in males, and epithelial mesenchymal transition (EMT) plays a critical role in PCa metastasis. Thus, developing biomarkers inhibiting EMT may provide significance for treatment of PCa. Hence, the aim of the current study was to investigate the mechanism by which FBP1 gene silencing influences PCa cell EMT, invasion and metastasis by mediating the MAPK pathway. PCa cell lines exhibiting the highest FBP1 expression were selected and treated with plasmids of siRNA-FBP1 sequence 1 and 2, pcDNA3.1-Flag-FBP1 (over-expression plasmid of FBP1), U0126 (an inhibitor of the ERK signaling pathway) and PD98059 (an inhibitor of the MEK signaling pathway). Cell proliferation, migration and invasion were detected by MTT assay, wound healing assay and Transwell assay, respectively. The mRNA and protein expression of related factors of EMT and MAPK signaling were determined by RT-qPCR and western blot analysis, respectively. Xenograft tumor growth after inoculation of DU145 cells was regularly analyzed in the nude mice. The positive expression of EMT markers was determined by immunohistochemistry. DU-145 and PC-3 cells displaying the highest FBP1 expression were selected for further analysis. The PCa cells treated with siRNA-FBP1 exhibited increased proliferation, migration rate and invasion, in addition to facilitated xenograft tumor growth. Notably, siRNA-FBP1 was identified to accelerate PCa cell EMT by elevating the expression of Vimentin and N-cadherin while diminishing E-cadherin expression via activation of the MAPK signaling pathway. The aforementioned results were reversed in PCa cells treated by pcDNA3.1-Flag-FBP1. Evidence has been provided in this study that FBP1 gene silencing activates the MAPK pathway, which ultimately promotes cell EMT, invasion and metastasis in PCa.
Collapse
Affiliation(s)
- Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhan Yang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Jin-Chun Qi
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhen-Wei Han
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Ming Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - De-Min Chen
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Xiao-Wei Wang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| |
Collapse
|
31
|
Prognostic Impact of Melatonin Receptors MT1 and MT2 in Non-Small Cell Lung Cancer (NSCLC). Cancers (Basel) 2019; 11:cancers11071001. [PMID: 31319607 PMCID: PMC6679108 DOI: 10.3390/cancers11071001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Several studies have investigated the inhibitory effect of melatonin on lung cancer cells. There are no data available on the prognostic impact of melatonin receptors MT1 and MT2 in non-small cell lung cancer (NSCLC). Materials and Methods: Immunohistochemical studies of MT1 and MT2 were conducted on NSCLC (N = 786) and non-malignant lung tissue (NMLT) (N = 120) using tissue microarrays. Molecular studies were performed on frozen fragments of NSCLC (N = 62; real time PCR), NMLT (N = 24) and lung cancer cell lines NCI-H1703, A549 and IMR-90 (real time PCR, western blot). Results: The expression of both receptors was higher in NSCLC than in NMLT. Higher MT1 and MT2 expression levels (at protein and mRNA) were noted in squamous cell carcinomas (SCC) compared to adenocarcinomas (AC). MT1 immunoexpression decreased as both the tumour size and the cancer stage increased in the whole cohort, while MT2 decreased as the cancer stage increased, with lymph node involvement (in the whole study group) and increasing malignancy grade (in SCC). Higher expression of MT2 was associated with a favorable prognosis. MT2 was an independent prognostic factor for overall survival (OS) in all analyzed NSCLC and in smoking patients. Conclusions: Our observations may point to the potential prognostic significance of MT2 in NSCLC.
Collapse
|
32
|
Liu Z, Zhao X, Shan H, Gao H, Wang P. microRNA-520c-3p suppresses NLRP3 inflammasome activation and inflammatory cascade in preeclampsia by downregulating NLRP3. Inflamm Res 2019; 68:643-654. [PMID: 31143973 DOI: 10.1007/s00011-019-01246-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/22/2019] [Accepted: 05/06/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The pathogenesis of preeclampsia (PE) is suggested to be a consequence of inflammation. Previously conducted investigations on nod-like receptor pyrin domain-containing 3 (NLRP3) have shed light to its crucial role in PE. Furthermore, microRNA-520c-3p (miR-520c-3p) is observed to be implicated in inflammation. Therefore, the current study aimed to explore the role of miR-520c-3p in inflammatory cascade of PE by targeting NLRP3. METHODS Microarray analyses were performed to screen differentially expressed genes associated with PE, and the potential relationship between miR-520c-3p and NLRP3 was analyzed. PE and normal placenta tissues were collected to determine the levels of inflammatory cytokines (IL-18, IL-33, IL-1β, IL-10, and TNF-α), miR-520c-3p and NLRP3. Hypoxic HTR8/SVneo cells were transfected with oe-NLRP3, si-NLRP3 or miR-520c-3p mimic to elucidate the functional role of NLRP3 or miR-520c-3p in the inflammatory cascade in PE, followed by the evaluation of levels of inflammatory cytokines and NLRP3 inflammasomes (NLRP3, ASC and caspase-1). Additionally, the HTR8/SVneo cell migration and invasion were evaluated. RESULTS An upregulation of NLRP3, IL-18, IL-1β and TNF-α, and downregulation of miR-520c-3p, IL-33 and IL-10 were observed in PE placenta tissues. NLRP3 was found to be a target gene of miR-520c-3p. HTR8/SVneo cells after hypoxia transfected with si-NLRP3 or miR-520c-3p mimic exhibited decreased levels of inflammatory cytokines and NLRP3 inflammasomes, in addition to increased IL-10 and IL-33 levels. Moreover, enhanced migration and invasion abilities were observed in cells transfected with si-NLRP3. CONCLUSION Collectively, miR-520c-3p could potentially inhibit NLRP3 inflammasome activation and inflammatory cascade in PE by downregulating NLRP3, highlighting the potential of miR-520c-3p as a therapeutic target for PE treatment.
Collapse
Affiliation(s)
- Zhaochun Liu
- Obstetrics Department, The First Affiliated Hospital of the Medical College, Shihezi University, No. 107, North 2nd Road, Shihezi, 832008, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Xia Zhao
- Obstetrics Department, The First Affiliated Hospital of the Medical College, Shihezi University, No. 107, North 2nd Road, Shihezi, 832008, The Xinjiang Uygur Autonomous Region, People's Republic of China.
| | - HongYing Shan
- Obstetrics Department, The First Affiliated Hospital of the Medical College, Shihezi University, No. 107, North 2nd Road, Shihezi, 832008, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Huan Gao
- Obstetrics Department, The First Affiliated Hospital of the Medical College, Shihezi University, No. 107, North 2nd Road, Shihezi, 832008, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Ping Wang
- Obstetrics Department, The First Affiliated Hospital of the Medical College, Shihezi University, No. 107, North 2nd Road, Shihezi, 832008, The Xinjiang Uygur Autonomous Region, People's Republic of China
| |
Collapse
|
33
|
Liao YJ, Yin XL, Deng Y, Peng XW. PRC1 gene silencing inhibits proliferation, invasion, and angiogenesis of retinoblastoma cells through the inhibition of the Wnt/β-catenin signaling pathway. J Cell Biochem 2019; 120:16840-16852. [PMID: 31144388 DOI: 10.1002/jcb.28942] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
Retinoblastoma is an ocular malignancy occurring in childhood. The current study evaluates the ability of silenced PRC1 on retinoblastoma cell proliferation, and angiogenesis via the Wnt/β-catenin signaling pathway. A total of 36 cases of retinoblastoma tissues (n = 36) and normal retinal tissues (n = 10) were selected in the current study. Retinoblastoma cells presenting with the high PRC1 messenger RNA (mRNA) expression were selected among the WERI-Rb-1, HXO-RB44, Y79, SO-Rb50, and SO-Rb70 cells lines, and were transfected with siRNA-PRC1 and LiCl (the activator of the Wnt/β-catenin pathway). The expressions of PRC1, VEGF, Wnt1, β-catenin, CyclinD1, extent of β-catenin, and GSK-3β phosphorylation were evaluated. Cell proliferation, cell-cycle distribution, and cell invasion of retinoblastoma cells were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, and Transwell assay. The angiogenesis of retinoblastoma cells was detected by tube formation assay. HXO-RB44 and WERI-Rb-1 cells were selected owing to the highest PRC1 mRNA expression. Meanwhile, PRC2 gene silencing presented lower expression levels of PRC1, VEGF, Wnt1, β-catenin, CyclinD1, extent of β-catenin and GSK-3β phosphorylation, decreased proliferation and invasion abilities, extended G0/G1 phase, and shortened S and G2/M phases of HXO-RB44 and WERI-Rb-1 cells, suggesting the silenced PRC2 inactivated Wnt/β-catenin pathway, so as to further restrain the retinoblastoma cell proliferation, invasion, and angiogenesis. These results support the view that PRC1 gene silencing could suppress the proliferation, and angiogenesis of retinoblastoma cells by repressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yu-Jun Liao
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiao-Long Yin
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Yan Deng
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiao-Wei Peng
- Department of Pediatric Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
34
|
Wen Y, Liu G, Zhang Y, Li H. MicroRNA-205 is associated with diabetes mellitus-induced erectile dysfunction via down-regulating the androgen receptor. J Cell Mol Med 2019; 23:3257-3270. [PMID: 30729682 PMCID: PMC6484320 DOI: 10.1111/jcmm.14212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/12/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
As a major class of regulatory genes in majority metazoans, microRNAs (miRs) play an important role in various diseases including diabetes mellitus (DM). Lack of androgens has previously been associated with DM-induced erectile dysfunction (DMED). In addition, the biological functioning of androgen is mediated by androgen receptor (AR). Herein, we sought to investigate whether miRs participate in AR-associated DMED. Sprague-Dawlay rats were employed to establish DMED models. After modelling, levels of miR-205 and AR in their cavernous bodies were measured. The relationship between miR-205 and AR was verified using a dual-luciferase reporter gene assay. The underlying regulatory mechanisms of miR-205 were investigated in concert with the treatment of mimics or inhibitors of miR-205, or AR overexpression in the cavernous smooth muscle cells (CSMCs) isolated from rats with DMED. Meanwhile, the effects of miR-205 and AR on cell proliferation and apoptosis were evaluated using MTT assay and flow cytometry respectively. Rats with DMED presented with increased miR-205 and decreased AR levels in the cavernous bodies. AR was identified as a target gene of miR-205. Down-regulation of miR-205 or up-regulation of AR could increase proliferation and inhibits apoptosis of CSMCs in addition to improvements in the erectile functioning of rats with DMED. In summary, miR-205 may contribute to the pathogenesis of DMED via down-regulation of AR expressions.
Collapse
Affiliation(s)
- Yan Wen
- Department of EndocrinologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Guohui Liu
- Department of CardiologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Yun Zhang
- Department of UrologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Hai Li
- Department of UrologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
35
|
Yang LX, Wu J, Guo ML, Zhang Y, Ma SG. Suppression of long non-coding RNA TNRC6C-AS1 protects against thyroid carcinoma through DNA demethylation of STK4 via the Hippo signalling pathway. Cell Prolif 2019; 52:e12564. [PMID: 30938030 PMCID: PMC6536409 DOI: 10.1111/cpr.12564] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/28/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023] Open
Abstract
Objectives Thyroid carcinoma (TC) represents a malignant neoplasm affecting the thyroid. Current treatment strategies include the removal of part of the thyroid; however, this approach is associated with a significant risk of developing hypothyroidism. In order to adequately understand the expression profiles of TNRC6C‐AS1 and STK4 and their potential functions in TC, an investigation into their involvement with Hippo signalling pathway and the mechanism by which they influence TC apoptosis and autophagy were conducted. Methods A microarray analysis was performed to screen differentially expressed lncRNAs associated with TC. TC cells were employed to evaluate the role of TNRC6C‐AS1 by over‐expression or silencing means. The interaction of TNRC6C‐AS1 with methylation of STK4 promoter was evaluated to elucidate its ability to elicit autophagy, proliferation and apoptosis. Results TNRC6C‐AS1 was up‐regulated while STK4 was down‐regulated, where methylation level was elevated. STK4 was verified as a target gene of TNRC6C‐AS1, which was enriched by methyltransferase. Methyltransferase’s binding to STK4 increased expression of its promoter. Over‐expressed TNRC6C‐AS1 inhibited STK4 by promoting STK4 methylation and reducing the total protein levels of MST1 and LATS1/2. The phosphorylation of YAP1 phosphorylation was decreased, which resulted in the promotion of SW579 cell proliferation and tumorigenicity. Conclusion Based on our observations, we subsequently confirmed the anti‐proliferative, pro‐apoptotic and pro‐autophagy capabilities of TNRC6C‐AS1 through STK4 methylation via the Hippo signalling pathway in TC.
Collapse
Affiliation(s)
- Liu-Xue Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ji Wu
- Department of Thyroid and Breast Surgery, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, China.,Department of Thyroid and Breast Surgery, Nanjing Drum Tower Hospital, Suqian, China
| | - Man-Li Guo
- Department of Endocrinology and Metabolism, Suqian People's Hospital, Nanjing Drum Tower Hospital, Suqian, China
| | - Yong Zhang
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, China.,Department of Endocrinology and Metabolism, Suqian First Hospital, Suqian, China
| | - Shao-Gang Ma
- Department of Endocrinology and Metabolism, Suqian First Hospital, Suqian, China.,Department of Endocrinology and Metabolism, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
36
|
Song S, Lin S, Liu J, Zhang M, Du Y, Zhang D, Xu W, Wang H. Retracted
: Targeting of SPP1 by microRNA‐340 inhibits gastric cancer cell epithelial–mesenchymal transition through inhibition of the PI3K/AKT signaling pathway. J Cell Physiol 2019; 234:18587-18601. [DOI: 10.1002/jcp.28497] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Su‐Zhen Song
- Department of Internal Medicine Shandong University of Traditional Chinese Medicine Jinan Shandong People's Republic of China
| | - Sen Lin
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Jia‐Ning Liu
- Department of Thyroid and Pancreatic Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Ming‐Bao Zhang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Ya‐Ting Du
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Dong‐Dong Zhang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Wei‐Hua Xu
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Hong‐Bo Wang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| |
Collapse
|
37
|
Zhu H, Cao XX, Liu J, Hua H. MicroRNA-488 inhibits endometrial glandular epithelial cell proliferation, migration, and invasion in endometriosis mice via Wnt by inhibiting FZD7. J Cell Mol Med 2019; 23:2419-2430. [PMID: 30729701 PMCID: PMC6433721 DOI: 10.1111/jcmm.14078] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/07/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022] Open
Abstract
Endometriosis is a chronic inflammatory syndrome and nearly 6%‐10% of women are affected by it during the reproductive period. Previous studies have proved that microRNAs (miRNAs) are implicated in the pathogenesis of ovarian endometriosis. In this study, we aimed to investigate that restored miR‐488 would effectively inhibit the development of endometriosis. The microarray‐based data analysis was performed to screen endometriosis‐related differentially expressed genes (DEGs). The mouse model in endometriosis syndrome was established by being subcutaneously injected with Estradiol benzoate, and the ectopic endometrial tissues and normal endometrial tissues were collected. Additionally, the endometrial glandular epithelial cells were extracted from the endometrial glandular epithelial tissues from normal and endometriosis mice. In order to examine the role of miR‐488 in mice with endometriosis, we measured miR‐488 expression and expression levels of Frizzled‐7 (FZD7), cyclinD1, β‐catenin, and c‐Myc in vivo and in vitro. Finally, we detected the effect of miR‐488 on cell proliferation, apoptosis, migration and invasion in vitro. FZD7 was upregulated in human endometriosis. The data showed higher expression levels of FZD7, β‐catenin, c‐Myc and cyclinD1, and lower miR‐488 expression in mouse endometrial tissues. FZD7 was the target gene of miR‐488. Furthermore, elevated miR‐488 in isolated mouse endometrial glandular endometrial cells inhibited FZD7, the translocation of β‐catenin to nucleus, the activation of Wnt pathway, and the cell proliferation, migration and invasion. Collectively, these findings indicated that up‐regulated miR‐488 may reduce the proliferation, migration and invasion of endometrial glandular epithelial cells through inhibiting the activation of Wnt pathway by down‐regulating FZD7.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Reproductive Health, Nanjing Maternity and Child Health Care Hospital, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Xi-Xia Cao
- Department of Reproductive Health, Nanjing Maternity and Child Health Care Hospital, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Juan Liu
- Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Hua Hua
- Department of Reproductive Health, Nanjing Maternity and Child Health Care Hospital, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing, P. R. China
| |
Collapse
|
38
|
Wang HB, Wei H, Wang JS, Li L, Chen AY, Li ZG. Down-regulated expression of LINC00518 prevents epithelial cell growth and metastasis in breast cancer through the inhibition of CDX2 methylation and the Wnt signaling pathway. Biochim Biophys Acta Mol Basis Dis 2019; 1865:708-723. [PMID: 30611858 DOI: 10.1016/j.bbadis.2019.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/07/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023]
Abstract
Breast cancer (BC)-related mortality is associated with the potential metastatic properties of the primary breast tumors. The following study was conducted with the main focus on the effect of LINC00518 on the growth and metastasis of BC epithelial cells via the Wnt signaling pathway through regulation of the methylation of CDX2 gene. Initially, differentially expressed long intergenic non-protein coding RNAs (lincRNAs) related to BC were screened out in the Cancer Genome Atlas (TCGA) database, after which we detected the LINC00518 expression and localization in BC tissues and cells. Then the CDX2 positive expression and methylation level were identified. The targeting relationship of LINC00518 and CDX2, and binding methyltransferase in the promoter region were examined. BC epithelial cell proliferation, colony formation ability, invasion, migration and apoptosis were further evaluated. The lincRNA expression data related to BC downloaded from the TCGA database revealed that there was a high expression of LINC00518 in BC, and a negative correlation between LINC00518 and CDX2. In addition, LINC00518 promotes CDX2 methylation by recruiting DNA methyltransferase through activating the Wnt signaling pathway. The down-regulation of LINC00518 inhibited proliferation, invasion, migration, and EMT of BC epithelial cells while enhancing apoptosis. The inhibitory effects of LINC00518 down-regulation was reversed by CDX2 down-regulation. In conclusion, our findings revealed that down-regulation of LINC00518 might have the ability to suppress BC progression by up-regulating CDX2 expression through the reduction of methylation and blockade of the Wnt signaling pathway, resulting in the inhibition of proliferation and promotion of apoptosis of BC epithelial cells.
Collapse
Affiliation(s)
- Hong-Bin Wang
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Hong Wei
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Jin-Song Wang
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Lin Li
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - An-Yue Chen
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Zhi-Gao Li
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China.
| |
Collapse
|
39
|
Qi AQ, Zhang YH, Qi QD, Liu YH, Zhu JL. Overexpressed HspB6 Underlines a Novel Inhibitory Role in Kainic Acid-Induced Epileptic Seizure in Rats by Activating the cAMP-PKA Pathway. Cell Mol Neurobiol 2019; 39:111-122. [PMID: 30511325 PMCID: PMC11479602 DOI: 10.1007/s10571-018-0637-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022]
Abstract
Epilepsy is a commonly occurring neurological disease that has a large impact on the patient's daily life. Phosphorylation of heat shock protein B6 (HspB6) has been reported to protect the central nervous system. In this investigation, we explored whether HspB6 played a positive effect on epilepsy with the involvement of the cyclic adenosine monophosphate-protein kinase A (cAMP-PKA) pathway. The epileptic seizure was induced in rats by intraperitoneal injection of kainic acid (KA). The extent of HspB6 phosphorylation and expressions of HspB6, PKA, and inflammatory factors TNF-α, IL-1β, and IL-6 were quantified along with neuronal apoptosis. To further understand the regulatory mechanism of the HspB6 in the hippocampus, we altered the expression and the extent of HspB6 phosphorylation to see whether the cAMP-PKA pathway was inactivated or not in hippocampal neurons of rats post KA. Results showed that HspB6 was poorly expressed, resulting in the inactivation of the cAMP-PKA pathway in rats post KA, as well as an aggravated inflammatory response and hippocampal neuronal apoptosis. HspB6 overexpression and the cAMP-PKA pathway activation decreased the expression of inflammatory factors and inhibited hippocampal neuronal apoptosis. Additionally, HspB6 phosphorylation further augments the inhibitory effects of HspB6 on the inflammatory response and hippocampal neuronal apoptosis. The cAMP-PKA pathway activation was found to result in increased HspB6 phosphorylation. HspB6 decreased apoptosis signal-regulating kinase 1 (ASK1) expression to inhibit inflammatory response and hippocampal neuronal apoptosis. Collectively, our findings demonstrate that activation of the cAMP-PKA pathway induces overexpression and partial phosphorylation of HspB6 lead to the inhibition of ASK1 expression. This in turn protects rats against epilepsy and provides a potential approach to prevent the onset of epileptic seizure in a clinical setting.
Collapse
Affiliation(s)
- Ai-Qin Qi
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, No. 001, Xuehu Street, Changshao North Road, Laicheng District, Laiwu, 271199, Shandong, People's Republic of China
| | - Yan-Hui Zhang
- Department of Neurology, Beijing Haidian Hospital, Beijing, 100080, People's Republic of China
| | - Qin-De Qi
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, No. 001, Xuehu Street, Changshao North Road, Laicheng District, Laiwu, 271199, Shandong, People's Republic of China
| | - Ye-Hui Liu
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, No. 001, Xuehu Street, Changshao North Road, Laicheng District, Laiwu, 271199, Shandong, People's Republic of China
| | - Jun-Ling Zhu
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, No. 001, Xuehu Street, Changshao North Road, Laicheng District, Laiwu, 271199, Shandong, People's Republic of China.
| |
Collapse
|
40
|
Yang Y, Gong B, Wu Z, Shuai P, Li D, Liu L, Yu M. Inhibition of microRNA‐129‐5p expression ameliorates ultraviolet ray‐induced corneal epithelial cell injury via upregulation of EGFR. J Cell Physiol 2018; 234:11692-11707. [PMID: 30515795 DOI: 10.1002/jcp.27837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/06/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Ying Yang
- Department of Ophthalmology Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Bo Gong
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Zheng‐Zheng Wu
- Department of Ophthalmology Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Ping Shuai
- Health Management Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Dong‐Feng Li
- Department of Ophthalmology Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Ling‐Lin Liu
- Department of Ophthalmology Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| | - Man Yu
- Department of Ophthalmology Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital Chengdu China
| |
Collapse
|
41
|
Meng H, Zhang Y, An S, Chen Y. Annexin A3 gene silencing promotes myocardial cell repair through activation of the PI3K/Akt signaling pathway in rats with acute myocardial infarction. J Cell Physiol 2018; 234:10535-10546. [PMID: 30456911 DOI: 10.1002/jcp.27717] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/17/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Hua Meng
- Department of Cardiology Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital Zhengzhou China
| | - Yan Zhang
- Department of Cardiology Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital Zhengzhou China
| | - Song‐Tao An
- Department of Cardiology Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital Zhengzhou China
| | - Yan Chen
- Department of Cardiology Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital Zhengzhou China
| |
Collapse
|
42
|
Somuncu ÖS, Coşkun Y, Ballica B, Temiz AF, Somuncu D. In vitro artificial skin engineering by decellularized placental scaffold for secondary skin problems of meningomyelocele. J Clin Neurosci 2018; 59:291-297. [PMID: 30385168 DOI: 10.1016/j.jocn.2018.10.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/07/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Meningomyelocele (MMC) is a condition that is originated by the fusion defect of the neural tube. It is a congenital anomaly and can be characterized by spinal cord defects and impaired skin integrity. It is very important to close the skin openings via three-dimensional artificial skin like construction for preventing infection and maintaining the healthy skin structure. Therefore, we aim to generate artificial skin like structures formed by the own cells of donor for treating the MMC-related skin disorder. METHODS In this study, waste placental tissues were collected and decellularization process was applied. Decellularized and normal placental tissues were compared by immunohistochemistry (IHC). Donor's own placental stem cells were seeded onto biological scaffold and were differentiated into skin related cell types. Finally, gene expressions were evaluated, and the structural integrity were analyzed with IHC. Tube formation assay was also performed for examining the angiogenesis formation of the tissue in order to evaluate the possibility of a healthy organ development. RESULTS Characterization experiments proved that the decellularized skin preserved a normal skin 3D construction and vasculature along with significant ECM arrangements. The well-kept placental ECM scaffold was cytocompatible, supportive of mesenchymal cell types. Native organ related scaffold is expected to carry a huge influence in skin tissue engineering via delivering a niche for skin-based cells and even for stem/progenitor cells. Regarding to the data obtained from this study, in vivo investigation the skin-like structure in animal models is thought to be the next step as a future prospect. CONCLUSION This study is a reference investigation for skin engineering based on placental stem cells and biological scaffolds.
Collapse
Affiliation(s)
| | - Yeşim Coşkun
- Bahçeşehir University, Faculty of Medicine, Department of Pediatrics, Turkey
| | | | | | | |
Collapse
|
43
|
Ben Brahim E, Ayari I, Jouini R, Atafi S, Koubaa W, Elloumi H, Chadli A. Expression of epidermal growth factor receptor (EGFR) in colorectal cancer: An immunohistochemical study. Arab J Gastroenterol 2018; 19:121-124. [PMID: 30243897 DOI: 10.1016/j.ajg.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 04/14/2018] [Accepted: 08/06/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND STUDY AIM The epidermal growth factor receptor (EGFR) plays an important role in tumourigenesis and tumour progression of colorectal cancer (CRC) and leads to the activation of intracellular signaling pathways. The use of anti-EGFR-targeted therapy has increased for patients with metastatic CRC. Today, the clinical utility of immunohistochemistry has remained somewhat inconclusive. It is based on EGFR screening methods using paraffin-embedded tumour specimen to select patients eligible for treatment. There is still lack of agreement on reproducible scoring criteria for EGFR immunohistochemistry has in various clinical trials. PATIENTS AND METHODS We retrospectively reviewed 36 CRC patients who underwent surgeries during 2011 in Habib Thameur hospital in Tunis. We analyzed the immunohistochemical overexpression of EGFR using a score based on immunostaining intensity. In addition, we analyzed the correlation between this overexpression and patients' clinicopathologic parameters. RESULTS The positive expression rate of EGFR was 78% (28/36). Using the immunoreactivity score, 21 cases were considered low grade expression and 15 tumours were high grade. Immunohistochemical expression of EGFR showed a significant difference with tumour's location (p = 0.034) and vascular invasion (p = 0.03). This expression was not significantly associated with age, gender, tumour size, histological type, grade, TNM staging and perineural invasion. CONCLUSIONS EGFR expression by immunohistochemistry in CRC is variably correlated with clinicopathological parameters. Its assessment by this method has still not proved its predictive value.
Collapse
Affiliation(s)
- Ehsen Ben Brahim
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia.
| | - Imen Ayari
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Raja Jouini
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Salsabil Atafi
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Wafa Koubaa
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Hela Elloumi
- Gastroenterology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Aschraf Chadli
- Pathology Department in Habib Thameur Hospital and Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
44
|
Neumeister VM, Juhl H. Tumor Pre-Analytics in Molecular Pathology: Impact on Protein Expression and Analysis. CURRENT PATHOBIOLOGY REPORTS 2018; 6:265-274. [PMID: 30595971 PMCID: PMC6290693 DOI: 10.1007/s40139-018-0179-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Purpose of Review Precision medicine promises patient tailored, individualized diagnosis and treatment of diseases and relies on clinical specimen integrity and accuracy of companion diagnostic testing. Therefore, pre-analytics, which are defined as the collection, processing, and storage of clinical specimens, are critically important to enable optimal diagnostics, molecular profiling, and clinical decision-making around harvested specimens. This review article discusses the impact of tumor pre-analytics on molecular pathology focusing on biospecimen protein expression and analysis. Recent Findings Due to busy clinical schedules and workflows that have been established for many years and to lack of standardization and limited assessment tools to quantify variability in pre-analytical processing, the effects of pre-analytics on biospecimen integrity are often overlooked. Several studies have recently emphasized an emerging crisis in science and reproducibility of results. Summary Biomarker instability due to pre-analytical variables affects comprehensive analysis and molecular phenotyping of patients’ tissue. This problematic emphasizes the critical need for standardized protocols and technologies to be applied in the clinical and research setting.
Collapse
Affiliation(s)
| | - Hartmut Juhl
- Indivumed, GmbH, Falkenried 88, D-20251 Hamburg, Germany
| |
Collapse
|
45
|
Yun S, Kwak Y, Nam SK, Seo AN, Oh HK, Kim DW, Kang SB, Lee HS. Ligand-Independent Epidermal Growth Factor Receptor Overexpression Correlates with Poor Prognosis in Colorectal Cancer. Cancer Res Treat 2018; 50:1351-1361. [PMID: 29361822 PMCID: PMC6192927 DOI: 10.4143/crt.2017.487] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/15/2018] [Indexed: 12/14/2022] Open
Abstract
Purpose Molecular treatments targeting epidermal growth factor receptors (EGFRs) are important strategies for advanced colorectal cancer (CRC). However, clinicopathologic implications of EGFRs and EGFR ligand signaling have not been fully evaluated. We evaluated the expression of EGFR ligands and correlation with their receptors, clinicopathologic factors, and patients’ survival with CRC. Materials and Methods The expression of EGFR ligands, including heparin binding epidermal growth factor-like growth factor (HBEGF), transforming growth factor (TGF), betacellulin, and epidermal growth factor (EGF), were evaluated in 331 consecutive CRC samples using mRNA in situ hybridization (ISH). We also evaluated the expression status of EGFR, human epidermal growth factor receptor 2 (HER2), HER3, and HER4 using immunohistochemistry and/or silver ISH. Results Unlike low incidences of TGF (38.1%), betacellulin (7.9%), and EGF (2.1%), HBEGF expression was noted in 62.2% of CRC samples. However, the expression of each EGFR ligand did not reveal significant correlations with survival. The combined analyses of EGFR ligands and EGFR expression indicated that the ligands‒/EGFR+ group showed a significant association with the worst disease-free survival (DFS; p=0.018) and overall survival (OS; p=0.005). It was also an independent, unfavorable prognostic factor for DFS (p=0.026) and OS (p=0.007). Additionally, HER4 nuclear expression, regardless of ligand expression, was an independent, favorable prognostic factor for DFS (p=0.034) and OS (p=0.049), by multivariate analysis. Conclusion Ligand-independent EGFR overexpression was suggested to have a significant prognostic impact; thus, the expression status of EGFR ligands, in addition to EGFR, might be necessary for predicting patients' outcome in CRC.
Collapse
Affiliation(s)
- Sumi Yun
- Department of Diagnostic Pathology, Samkwang Medical Laboratories, Seoul, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Soo Kyung Nam
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - An Na Seo
- Department of Pathology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Heung-Kwon Oh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Duck-Woo Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung-Bum Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.,Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
46
|
Li R, Zhou M, Li J, Wang Z, Zhang W, Yue C, Ma Y, Peng H, Wei Z, Hu Z. Identifying EGFR-Expressed Cells and Detecting EGFR Multi-Mutations at Single-Cell Level by Microfluidic Chip. NANO-MICRO LETTERS 2018; 10:16. [PMID: 30393665 PMCID: PMC6199061 DOI: 10.1007/s40820-017-0168-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/14/2017] [Indexed: 05/20/2023]
Abstract
EGFR mutations companion diagnostics have been proved to be crucial for the efficacy of tyrosine kinase inhibitor targeted cancer therapies. To uncover multiple mutations occurred in minority of EGFR-mutated cells, which may be covered by the noises from majority of un-mutated cells, is currently becoming an urgent clinical requirement. Here we present the validation of a microfluidic-chip-based method for detecting EGFR multi-mutations at single-cell level. By trapping and immunofluorescently imaging single cells in specifically designed silicon microwells, the EGFR-expressed cells were easily identified. By in situ lysing single cells, the cell lysates of EGFR-expressed cells were retrieved without cross-contamination. Benefited from excluding the noise from cells without EGFR expression, the simple and cost-effective Sanger's sequencing, but not the expensive deep sequencing of the whole cell population, was used to discover multi-mutations. We verified the new method with precisely discovering three most important EGFR drug-related mutations from a sample in which EGFR-mutated cells only account for a small percentage of whole cell population. The microfluidic chip is capable of discovering not only the existence of specific EGFR multi-mutations, but also other valuable single-cell-level information: on which specific cells the mutations occurred, or whether different mutations coexist on the same cells. This microfluidic chip constitutes a promising method to promote simple and cost-effective Sanger's sequencing to be a routine test before performing targeted cancer therapy.
Collapse
Affiliation(s)
- Ren Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Mingxing Zhou
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Jine Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Zihua Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Weikai Zhang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Chunyan Yue
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Yan Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Hailin Peng
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China
| | - Zewen Wei
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
- Yangtze River Delta Academy of Nanotechnology and Industry Development Research, Jiaxing, 314000, Zhejiang Province, People's Republic of China.
| |
Collapse
|
47
|
Intartaglia M, Sabetta R, Gargiulo M, Roncador G, Marino FZ, Franco R. Immunohistochemistry for Cancer Stem Cells Detection: Principles and Methods. Methods Mol Biol 2018; 1692:195-211. [PMID: 28986898 DOI: 10.1007/978-1-4939-7401-6_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are rare immortal cells within a tumor that can self-renew and drive tumorigenesis. CSCs play a pivotal role in the tumor development, progression and relapse, as well as in the resistance of anticancer therapy. Different tools could help in the analysis of CSCs, especially Immunohistochemistry (IHC) represents a useful technique able to identify several specific CSC markers. The main aims of this chapter are the description of the explain immunohistochemical methods used in the characterization of CSCs. Furthermore, focus on the most common troubleshooting in CSCs IHC is provided, especially the pitfalls of the CSCs markers IHC on tissue microarrays.
Collapse
Affiliation(s)
- Martina Intartaglia
- Pathology Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni, 20, 80138, Naples, Italy
| | - Rosalaura Sabetta
- Pathology Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni, 20, 80138, Naples, Italy
| | - Monica Gargiulo
- Pathology Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni, 20, 80138, Naples, Italy
| | - Giovanna Roncador
- Monoclonal Antibody Unit, Spanish National Cancer Research Center, Madrid, Spain
| | - Federica Zito Marino
- Pathology Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni, 20, 80138, Naples, Italy
| | - Renato Franco
- Pathology Unit, University of Campania "Luigi Vanvitelli", Via Luciano Armanni, 20, 80138, Naples, Italy.
| |
Collapse
|
48
|
Yaghjyan L, Stoll E, Ghosh K, Scott CG, Jensen MR, Brandt KR, Visscher D, Vachon CM. Tissue-based associations of mammographic breast density with breast stem cell markers. Breast Cancer Res 2017; 19:100. [PMID: 28851411 PMCID: PMC5576318 DOI: 10.1186/s13058-017-0889-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022] Open
Abstract
Background Mammographic breast density is a well-established, strong breast cancer risk factor but the biology underlying this association remains unclear. Breast density may reflect underlying alterations in the size and activity of the breast stem cell pool. We examined, for the first time, associations of CD44, CD24, and aldehyde dehydrogenase family 1 member A1 (ALDH1A1) breast stem cell markers with breast density. Methods We included in this study 64 asymptomatic healthy women who previously volunteered for a unique biopsy study of normal breast tissue at the Mayo Clinic (2006-2008). Mammographically identified dense and non-dense areas were confirmed/localized by ultrasound and biopsied. Immunohistochemical analysis of the markers was performed according to a standard protocol and the staining was assessed by a single blinded pathologist. In core biopsy samples retrieved from areas of high vs. low density within the same woman, we compared staining extent and an expression score (the product of staining intensity and extent), using the signed rank test. All tests of statistical significance were two-sided. Results A total of 64, 28, and 10 women were available for CD44, CD24, and ALDH1A1 staining, respectively. For all three markers, we found higher levels of staining extent in dense as compared to non-dense tissue, though for CD24 and ALDH1A1 the difference did not reach statistical significance (CD44, 6.3% vs. 2.0%, p < 0.001; CD24, 8.0% vs. 5.6%, p = 0.10; and ALDH1A1, 0.5% vs. 0.3%, p = 0.12). The expression score for CD44 was significantly greater in dense as compared to non-dense tissue (9.8 vs.3.0, p < 0.001). Conclusions Our findings suggest an increased presence and/or activity of stem cells in dense as compared to non-dense breast tissue. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0889-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lusine Yaghjyan
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd, Gainesville, FL, 32610, USA.
| | - Ethan Stoll
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610, USA
| | - Karthik Ghosh
- Division of General Internal Medicine, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55902, USA
| | - Christopher G Scott
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA
| | - Matthew R Jensen
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA
| | - Kathleen R Brandt
- Department of Radiology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA
| | - Daniel Visscher
- Department of Anatomic Pathology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA
| | - Celine M Vachon
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, USA
| |
Collapse
|
49
|
Beca F, Kensler K, Glass B, Schnitt SJ, Tamimi RM, Beck AH. EZH2 protein expression in normal breast epithelium and risk of breast cancer: results from the Nurses' Health Studies. Breast Cancer Res 2017; 19:21. [PMID: 28253895 PMCID: PMC5335498 DOI: 10.1186/s13058-017-0817-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Enhancer of zeste homolog 2 (EZH2) is a polycomb-group protein that is involved in stem cell renewal and carcinogenesis. In breast cancer, increased EZH2 expression is associated with aggressiveness and has been suggested to identify normal breast epithelium at increased risk of breast cancer development. However, the association between EZH2 expression in benign breast tissue and breast cancer risk has not previously been evaluated in a large prospective cohort. METHODS We examined the association between EZH2 protein expression and subsequent breast cancer risk using logistic regression in a nested case-control study of benign breast disease (BBD) and breast cancer within the Nurses' Health Studies. EZH2 immunohistochemical expression in normal breast epithelium and stroma was evaluated by computational image analysis and its association with breast cancer risk was analyzed after adjusting for matching factors between cases and controls, the concomitant BBD diagnosis, and the Ki67 proliferation index. RESULTS Women with a breast biopsy in which more than 20% of normal epithelial cells expressed EZH2 had a significantly increased risk of developing breast cancer (odds ratio (OR) 2.95, 95% confidence interval (CI) 1.11-7.84) compared to women with less than 10% EZH2 epithelial expression. The risk of developing breast cancer increased for each 5% increase in EZH2 expression (OR 1.22, 95% CI 1.02-1.46, p value 0.026). Additionally, women with high EZH2 expression and low estrogen receptor (ER) expression had a 4-fold higher risk of breast cancer compared to women with low EZH2 and low ER expression (OR 4.02, 95% CI 1.29-12.59). CONCLUSIONS These results provide further evidence that EZH2 expression in the normal breast epithelium is independently associated with breast cancer risk and might be used to assist in risk stratification for women with benign breast biopsies.
Collapse
Affiliation(s)
- Francisco Beca
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, 02215, MA, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, MA, USA. .,Harvard Medical School, Boston, 02215, MA, USA.
| | - Kevin Kensler
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Benjamin Glass
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, 02215, MA, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, MA, USA
| | - Stuart J Schnitt
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, 02215, MA, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, MA, USA.,Harvard Medical School, Boston, 02215, MA, USA
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew H Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, 02215, MA, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, MA, USA.,Harvard Medical School, Boston, 02215, MA, USA
| |
Collapse
|
50
|
Abstract
In the last 20 years, improvements in metastatic colorectal cancer treatment lead to a radical raise of outcomes with median survival reaching now more than 30 months. Despite that, the identification of predictive and/or prognostic biomarker still represents a challenging issue, and until today, although clinician and researchers might face with a deeper knowledge of biological mechanisms related to colorectal cancer, many pieces of evidence underline the heterogeneity and the dynamism of such disease. In the present review, we describe the road leading to the discovery of RAS mutations, BRAF V600E mutation, and microsatellite instability role in colorectal cancer; second, we discuss some of the possible major pitfalls of biomarker research, and lastly, we give new suggestions for future research in this field.
Collapse
|