1
|
Li X, Gu Q, Sun P, Yang L, Zhang X, Lu B, Ni Q. NSG2: a promising prognostic marker shaping the immune landscape of breast cancer. Front Immunol 2024; 15:1487447. [PMID: 39493764 PMCID: PMC11527618 DOI: 10.3389/fimmu.2024.1487447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background Breast cancer (BC) remains a significant health issue globally and most common cause of mortality in women. Enhancing our understanding on biomarkers may greatly improve both diagnostic and therapeutic approaches to this disease. Methods We retrospectively assessed tumor samples from 228 BC cases and 51 normal samples, alongside relevant clinical data. Neuronal vesicle trafficking associated 2(NSG2) expression was evaluated through bioinformatics and multiplex immunohistochemistry. Associations between NSG2 expression, tumor-infiltrating immune cells (TIICs), immune checkpoints, and clinical outcomes were investigated. Results NSG2 was present in both breast cancer cells and adjacent stromal cells. Increased NSG2 expression in cancer cells correlated with greater tumor size, distant metastasis, and more advanced clinical stages. Kaplan-Meier survival and multivariate analyses identified NSG2 expression in both cancer and stromal cells as an independent prognostic factor for breast cancer survival. Elevated NSG2 levels both in cancer and stroma cells were linked to increased CD4+ T, CD8+ T, and Lamp3+ dendritic cells infiltration in stromal regions (P < 0.05). Conversely, the expression of NSG2 in the stroma was negatively correlated with CD20+ B cells (P < 0.05). Additionally, NSG2 expression was found to be associated with CTLA-4 levels (P < 0.05). Conclusion NSG2 seems to be a significant component of the BC immune microenvironment and may serve as an important prognostic marker.
Collapse
Affiliation(s)
- Xuan Li
- Department of General Surgery, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Qiming Gu
- Department of General Surgery, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Pingping Sun
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lei Yang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xiaojing Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Bing Lu
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Qichao Ni
- Department of General Surgery, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| |
Collapse
|
2
|
Abomoelak B, Prather R, Pragya SU, Pragya SC, Mehta ND, Uddin P, Veeramachaneni P, Mehta N, Young A, Kapoor S, Mehta D. Cognitive Skills and DNA Methylation Are Correlating in Healthy and Novice College Students Practicing Preksha Dhyāna Meditation. Brain Sci 2023; 13:1214. [PMID: 37626570 PMCID: PMC10452635 DOI: 10.3390/brainsci13081214] [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: 06/21/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The impact of different meditation protocols on human health is explored at the cognitive and cellular levels. Preksha Dhyana meditation has been observed to seemingly affect the cognitive performance, transcriptome, and methylome of healthy and novice participant practitioners. In this study, we performed correlation analyses to investigate the presence of any relationships in the changes in cognitive performance and DNA methylation in a group of college students practicing Preksha Dhyāna (N = 34). Nine factors of cognitive performance were assessed at baseline and 8 weeks postintervention timepoints in the participants. Statistically significant improvements were observed in six of the nine assessments, which were predominantly relating to memory and affect. Using Illumina 850 K microarray technology, 470 differentially methylated sites (DMS) were identified between the two timepoints (baseline and 8 weeks), using a threshold of p-value < 0.05 and methylation levels beyond -3% to 3% at every site. Correlation analysis between the changes in performance on each of the nine assessments and every DMS unveiled statistically significant positive and negative relationships at several of these sites. The identified DMS were in proximity of essential genes involved in signaling and other important metabolic processes. Interestingly, we identified a set of sites that can be considered as biomarkers for Preksha meditation improvements at the genome level.
Collapse
Affiliation(s)
- Bassam Abomoelak
- Gastrointestinal Translational Laboratory, Arnold Palmer Hospital for Children, Orlando, FL 32806, USA;
| | - Ray Prather
- Pediatric Cardiothoracic Surgery Department, Arnold Palmer Hospital for Children, Orlando, FL 32806, USA;
| | - Samani U. Pragya
- Department of Religions and Philosophies, University of London, London WC1H 0XG, UK;
| | - Samani C. Pragya
- Department of Biostatistics, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA;
| | - Neelam D. Mehta
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Parvin Uddin
- College of Arts, Sciences and Education, Florida International University, Miami, FL 33199, USA;
| | | | - Naina Mehta
- Neurodevelopmental Pediatrician, Behavioral and Developmental Center, Orlando Health, Orlando, FL 32805, USA;
| | - Amanda Young
- Institute for Simulation and Training, University of Central Florida, Orlando, FL 32765, USA;
| | - Saumya Kapoor
- Medical School, University of Central Florida, Orlando, FL 32827, USA;
| | - Devendra Mehta
- Gastrointestinal Translational Laboratory, Arnold Palmer Hospital for Children, Orlando, FL 32806, USA;
| |
Collapse
|
3
|
Bhattacharya A, Santhoshkumar A, Kurahara H, Harihar S. Metastasis Suppressor Genes in Pancreatic Cancer: An Update. Pancreas 2021; 50:923-932. [PMID: 34643607 DOI: 10.1097/mpa.0000000000001853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT Pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), has for long remained a deadly form of cancer characterized by high mortality rates resulting from metastasis to multiple organs. Several factors, including the late manifestation of the disease, partly amplified by lack of efficient screening methods, have hampered the drive to design an effective therapeutic strategy to treat this deadly cancer. Understanding the biology of PDAC progression and identifying critical genes regulating these processes are essential to overcome the barriers toward effective treatment. Metastasis suppressor genes have been shown to inhibit multiple steps in the metastatic cascade without affecting primary tumor formation and are considered to hold promise for treating metastatic cancers. In this review, we catalog the bona fide metastasis suppressor genes reported in PDAC and discuss their known mechanism of action.
Collapse
Affiliation(s)
- Arnav Bhattacharya
- From the Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Anirudh Santhoshkumar
- From the Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Hiroshi Kurahara
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Sitaram Harihar
- From the Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
4
|
Young ED, Manley SJ, Beadnell TC, Shearin AE, Sasaki K, Zimmerman R, Kauffman E, Vivian CJ, Parasuram A, Iwakuma T, Grandgenett PM, Hollingsworth MA, O'Neil M, Welch DR. Suppression of pancreatic cancer liver metastasis by secretion-deficient ITIH5. Br J Cancer 2021; 124:166-175. [PMID: 33024269 PMCID: PMC7782545 DOI: 10.1038/s41416-020-01093-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/14/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Previously, we identified ITIH5 as a suppressor of pancreatic ductal adenocarcinoma (PDAC) metastasis in experimental models. Expression of ITIH5 correlated with decreased cell motility, invasion and metastasis without significant inhibition of primary tumour growth. Here, we tested whether secretion of ITIH5 is required to suppress liver metastasis and sought to understand the role of ITIH5 in human PDAC. METHODS We expressed mutant ITIH5 with deletion of the N-terminal secretion sequence (ITIH5Δs) in highly metastatic human PDAC cell lines. We used a human tissue microarray (TMA) to compare ITIH5 levels in uninvolved pancreas, primary and metastatic PDAC. RESULTS Secretion-deficient ITIH5Δs was sufficient to suppress liver metastasis. Similar to secreted ITIH5, expression of ITIH5Δs was associated with rounded cell morphology, reduced cell motility and reduction of liver metastasis. Expression of ITIH5 is low in both human primary PDAC and matched metastases. CONCLUSIONS Metastasis suppression by ITIH5 may be mediated by an intracellular mechanism. In human PDAC, loss of ITIH5 may be an early event and ITIH5-low PDAC cells in primary tumours may be selected for liver metastasis. Further defining the ITIH5-mediated pathway in PDAC could establish future therapeutic exploitation of this biology and reduce morbidity and mortality associated with PDAC metastasis.
Collapse
Affiliation(s)
- Eric D Young
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sharon J Manley
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Thomas C Beadnell
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexander E Shearin
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ken Sasaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Rosalyn Zimmerman
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Evan Kauffman
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carolyn J Vivian
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Aishwarya Parasuram
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul M Grandgenett
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Maura O'Neil
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
5
|
Zhang Y, Zhu L, Wang X. A Network-Based Approach for Identification of Subtype-Specific Master Regulators in Pancreatic Ductal Adenocarcinoma. Genes (Basel) 2020; 11:genes11020155. [PMID: 32024063 PMCID: PMC7074188 DOI: 10.3390/genes11020155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the predominant subtype of pancreatic cancer, has been reported with equal mortality and incidence for decades. The lethality of PDAC is largely due to its late presentation, when surgical resection is no longer an option. Similar to other major malignancies, it is now clear that PDAC is not a single disease, posing a great challenge to precise selection of patients for optimized adjuvant therapy. A representative study found that PDAC comprises four distinct molecular subtypes: squamous, pancreatic progenitor, immunogenic, and aberrantly differentiated endocrine exocrine (ADEX). However, little is known about the molecular mechanisms underlying specific PDAC subtypes, hampering the design of novel targeted agents. In this study we performed network inference that integrates miRNA expression and gene expression profiles to dissect the miRNA regulatory mechanism specific to the most aggressive squamous subtype of PDAC. Master regulatory analysis revealed that the particular subtype of PDAC is predominantly influenced by miR-29c and miR-192. Further integrative analysis found miR-29c target genes LOXL2, ADAM12 and SERPINH1, which all showed strong association with prognosis. Furthermore, we have preliminarily revealed that the PDAC cell lines with high expression of these miRNA target genes showed significantly lower sensitivities to multiple anti-tumor drugs. Together, our integrative analysis elucidated the squamous subtype-specific regulatory mechanism, and identified master regulatory miRNAs and their downstream genes, which are potential prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China; (Y.Z.); (L.Z.)
| | - Lina Zhu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China; (Y.Z.); (L.Z.)
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China; (Y.Z.); (L.Z.)
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Correspondence: ; Tel.: +852-3442-2367
| |
Collapse
|
6
|
Rose M, Meurer SK, Kloten V, Weiskirchen R, Denecke B, Antonopoulos W, Deckert M, Knüchel R, Dahl E. ITIH5 induces a shift in TGF-β superfamily signaling involving Endoglin and reduces risk for breast cancer metastasis and tumor death. Mol Carcinog 2017; 57:167-181. [PMID: 28940371 DOI: 10.1002/mc.22742] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/07/2017] [Accepted: 09/18/2017] [Indexed: 12/26/2022]
Abstract
ITIH5 has been proposed being a novel tumor suppressor in various tumor entities including breast cancer. Recently, ITIH5 was furthermore identified as metastasis suppressor gene in pancreatic carcinoma. In this study we aimed to specify the impact of ITIH5 on metastasis in breast cancer. Therefore, DNA methylation of ITIH5 promoter regions was assessed in breast cancer metastases using the TCGA portal and methylation-specific PCR (MSP). We reveal that the ITIH5 upstream promoter region is particularly responsible for ITIH5 gene inactivation predicting shorter survival of patients. Notably, methylation of this upstream ITIH5 promoter region was associated with disease progression, for example, abundantly found in distant metastases. In vitro, stably ITIH5-overexpressing MDA-MB-231 breast cancer clones were used to analyze cell invasion and to identify novel ITIH5-downstream targets. Indeed, ITIH5 re-expression suppresses invasive growth of MDA-MB-231 breast cancer cells while modulating expression of genes involved in metastasis including Endoglin (ENG), an accessory TGF-β receptor, which was furthermore co-expressed with ITIH5 in primary breast tumors. By performing in vitro stimulation of TGF-β signaling using TGF-β1 and BMP-2 we show that ITIH5 triggered a TGF-β superfamily signaling switch contributing to downregulation of targets like Id1, known to endorse metastasis. Moreover, ITIH5 predicts longer overall survival (OS) only in those breast tumors that feature high ENG expression or inversely regulated ID1 suggesting a clinical and functional impact of an ITIH5-ENG axis for breast cancer progression. Hence, we provide evidence that ITIH5 may represent a novel modulator of TGF-β superfamily signaling involved in suppressing breast cancer metastasis.
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Steffen K Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Vera Kloten
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- IZKF Aachen, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Wiebke Antonopoulos
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Martina Deckert
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Ruth Knüchel
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Huang X, Ta N, Zhang Y, Gao Y, Hu R, Deng L, Zhang B, Jiang H, Zheng J. Microarray Analysis of the Expression Profile of Long Non-Coding RNAs Indicates lncRNA RP11-263F15.1 as a Biomarker for Diagnosis and Prognostic Prediction of Pancreatic Ductal Adenocarcinoma. J Cancer 2017; 8:2740-2755. [PMID: 28928863 PMCID: PMC5604206 DOI: 10.7150/jca.18073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/16/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognostic outcomes. Accumulating evidence has demonstrated that long non-coding RNAs (lncRNAs) play an important role in the development and progression of carcinogenesis. Nevertheless, little is known about the role of lncRNAs in PDAC. The aim of the current study was to find differentially expressed lncRNAs and related mRNAs in human PDAC tissues and adjacent normal tissues by microarray analysis, and investigate the relationship between lncRNA RP11-263F15.1 levels and the clinicaopathological features of PDAC patients. It was found that 4364 lncRNAs and 4862 related mRNAs were significantly dysregulated in PDAC tissues as compared with adjacent normal tissues with a fold change ≥2.0 (P<0.05). GO and pathway analyses showed that the up-regulated gene profiles were related to several pathways associated with carcinogenesis, while the down-regulated gene profiles were closely correlated with nutrient metabolism. RP11-263F15.1 levels were associated with histologic differentiation (P=0.001). Besides, Kaplan-Meier analysis showed that high expression of RP11-263F15.1 was associated with poor outcomes, but multivariate analysis suggested that RP11-263F15.1 was not an independent factor for predicting prognosis of PDAC. In conclusion, these data indicate that differentially expressed lncRNAs and mRNAs were involved in the carcinogenesis of PDAC, and RP11-263F15.1 may prove to be a potential biomarker for the diagnosis and prognostic prediction of PDAC.
Collapse
Affiliation(s)
- Xiaoyi Huang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Na Ta
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yunshuo Zhang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yisha Gao
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ronglei Hu
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Lulu Deng
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Bingbing Zhang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jianming Zheng
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
8
|
Genome-wide in vivo RNAi screen identifies ITIH5 as a metastasis suppressor in pancreatic cancer. Clin Exp Metastasis 2017; 34:229-239. [PMID: 28289921 DOI: 10.1007/s10585-017-9840-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 02/13/2017] [Indexed: 12/18/2022]
Abstract
The overwhelming majority of pancreatic ductal adenocarcinoma (PDAC) is not diagnosed until the cancer has metastasized, leading to an abysmal average life expectancy (3-6 months post-diagnosis). Earlier detection and more effective treatments have been hampered by inadequate understanding of the underlying molecular mechanisms controlling metastasis. We hypothesized that metastasis suppressors are involved in controlling metastasis in pancreatic cancer. Using an unbiased genome-wide shRNA screen, an shRNA library was transduced into the non-metastatic PDAC line S2-028 followed by intrasplenic injection. Resulting liver metastases were individually isolated from these mice. One liver metastatic nodule contained shRNA for ITIH5 (Inter-alpha-trypsin inhibitor heavy chain 5), suggesting that ITIH5 may act as a metastasis suppressor. Consistent with this notion, metastatic PDAC cell lines had significantly lower protein expression of ITIH5 compared to immortalized pancreatic ductal epithelial cells and non-/poorly-metastatic PDAC cell lines. By manipulating expression of ITIH5 in different PDAC cell lines (over-expression in metastatic, knockdown in non-metastatic) functional and selective regulation of metastasis was observed for ITIH5. Orthotopic tumor growth of PDAC cells was not blocked following orthotopic injection. In vitro ITIH5 over-expression inhibited motility and invasion. Immunohistochemical analysis of a human PDAC tissue microarray revealed that ITIH5 expression inversely correlated with both survival and invasion/metastasis. ITIH5 is, therefore, functionally validated as a PDAC metastasis suppressor and shows promise as a prognostic biomarker.
Collapse
|