1
|
Glass SE, Bechard ME, Cao Z, Aramandla R, Zhao P, Ellis ST, Green EH, Fisher EG, Smith RT, Sievers CK, Irudayam MJ, Revetta F, Washington MK, Ayers GD, Heiser CN, Simmons AJ, Xu Y, Wang Y, Windon A, Shrubsole MJ, Markham NO, Liu Q, Lau KS, Coffey RJ. Dipeptidase-1-knockout mice develop invasive tumors with features of microsatellite-unstable colorectal cancer. JCI Insight 2025; 10:e186938. [PMID: 40178918 DOI: 10.1172/jci.insight.186938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
Dipeptidase-1 (DPEP1) is highly upregulated in colorectal cancer (CRC), with its enzymatic function linked to invasion and metastasis. More recently, DPEP1 was found to serve as a receptor for neutrophils when expressed by activated endothelial cells. It is unknown whether neutrophils bind to DPEP1-expressing CRC cells and whether this impacts features of CRC. Neutrophils have been shown to be tumor promoting in cancers including CRC, where they act to exclude CD8+ T cells. Herein, we show that neutrophils bind DPEP1-expressing CRC cells. In addition, DPEP1 is preferentially expressed in microsatellite-stable (MSS) CRCs, in which there are a paucity of CD8+ T cells, whereas DPEP1 is negatively correlated with microsatellite-unstable (MSI-H) CRCs, which are T cell rich and are more responsive to immunotherapy. Remarkably, carcinogen-treated Dpep1-null mice develop multiple, large, plaque-like, locally invasive adenocarcinomas and squamous cell cancers in the distal colon. These adenocarcinomas exhibit a marked reduction in neutrophils and an influx CD8+ T cells, along with reduced expression of mismatch repair proteins, consistent with features of MSI-H CRC. These results establish DPEP1's importance in maintaining MSS CRC and its ability to shape the tumor microenvironment.
Collapse
Affiliation(s)
- Sarah E Glass
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Medicine
- Epithelial Biology Center
| | | | - Zheng Cao
- Department of Medicine
- Epithelial Biology Center
| | | | - Ping Zhao
- Department of Medicine
- Epithelial Biology Center
| | | | - Emily H Green
- Epithelial Biology Center
- Vanderbilt Institute for Infection, Microbiology, and Immunology, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth G Fisher
- Epithelial Biology Center
- Cancer Biology Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | - Frank Revetta
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cody N Heiser
- Epithelial Biology Center
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Alan J Simmons
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center
| | - Yanwen Xu
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annika Windon
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Martha J Shrubsole
- Department of Medicine
- Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nicholas O Markham
- Department of Medicine
- Epithelial Biology Center
- Vanderbilt Institute for Infection, Microbiology, and Immunology, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Cancer Biology Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ken S Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
- Center for Computational Systems Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert J Coffey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Medicine
- Epithelial Biology Center
| |
Collapse
|
2
|
Li Y, Wang X, Zhang Q, Tian D, Bai Y, Feng Y, Liu W, Diao Z. Dipeptidase 1 promotes ferroptosis in renal tubular epithelial cells in diabetic nephropathy via inhibition of the GSH/GPX4 axis. Int Immunopharmacol 2024; 133:111955. [PMID: 38626544 DOI: 10.1016/j.intimp.2024.111955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024]
Abstract
Renal tubular injury is an important pathological change associated with diabetic nephropathy (DN), in which ferroptosis of renal tubular epithelial cells is critical to its pathogenesis. Inhibition of the glutathione/glutathione peroxidase 4 (GSH/GPX4) axis is the most important mechanism in DN tubular epithelial cell ferroptosis, but the underlying reason for this is unclear. Our biogenic analysis showed that a zinc-dependent metalloproteinase, dipeptidase 1 (DPEP1), is associated with DN ferroptosis. Here, we investigated the role and mechanism of DPEP1 in DN tubular epithelial cell ferroptosis. DPEP1 upregulation was observed in the renal tubular epithelial cells of DN patients and model mice, as well as in HK-2 cells stimulated with high glucose. Furthermore, the level of DPEP1 upregulation was associated with the degree of tubular injury in DN patients and HK-2 cell ferroptosis. Mechanistically, knocking down DPEP1 expression could alleviate the inhibition of GSH/GPX4 axis and reduce HK-2 cell ferroptosis levels in a high glucose environment. HK-2 cells with stable DPEP1 overexpression also showed GSH/GPX4 axis inhibition and ferroptosis, but blocking the GSH/GPX4 axis could mitigate these effects. Additionally, treatment with cilastatin, a DPEP1 inhibitor, could ameliorate GSH/GPX4 axis inhibition and relieve ferroptosis and DN progression in DN mice. These results revealed that DPEP1 can promote ferroptosis in DN renal tubular epithelial cells via inhibition of the GSH/GPX4 axis.
Collapse
Affiliation(s)
- Yan Li
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xueqi Wang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Qidong Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Dongli Tian
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yu Bai
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yiduo Feng
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Cvjetkovic A, Karimi N, Crescitelli R, Thorsell A, Taflin H, Lässer C, Lötvall J. Proteomic profiling of tumour tissue-derived extracellular vesicles in colon cancer. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e127. [PMID: 38939898 PMCID: PMC11080707 DOI: 10.1002/jex2.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 07/19/2023] [Accepted: 10/23/2023] [Indexed: 06/29/2024]
Abstract
Colon cancer is one of the most commonly occurring tumours among both women and men, and over the past decades the incidence has been on the rise. As such, the need for biomarker identification as well as an understanding of the underlying disease mechanism has never been greater. Extracellular vesicles are integral mediators of cell-to-cell communication and offer a unique opportunity to study the machinery that drives disease progression, and they also function as vectors for potential biomarkers. Tumour tissue and healthy mucosal tissue from the colons of ten patients were used to isolate tissue-resident EVs that were subsequently subjected to global quantitative proteomic analysis through LC-MS/MS. In total, more than 2000 proteins were identified, with most of the common EV markers being among them. Bioinformatics revealed a clear underrepresentation of proteins involved in energy production and cellular adhesion in tumour EVs, while proteins involved in protein biosynthesis were overrepresented. Additionally, 53 membrane proteins were found to be significantly upregulated in tumour EVs. Among them were several proteins with enzymatic functions that degrade the extracellular matrix, and three of these, Fibroblast activating factor (FAP), Cell surface hyaluronidase (CEMIP2), as well as Ephrin receptor B3 (EPHB3), were validated and found to be consistent with the global quantitative results. These stark differences in the proteomes between healthy and cancerous tissue emphasise the importance of the interstitial vesicle secretome as a major player of disease development.
Collapse
Affiliation(s)
- Aleksander Cvjetkovic
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Nasibeh Karimi
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Rossella Crescitelli
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University HospitalGothenburgSweden
| | - Annika Thorsell
- Proteomics Core Facility, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Helena Taflin
- Transplant Institute at Sahlgrenska University Hospital, Institute of Clinical SciencesSahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Wang Y, Zhang T, Du H, Yang M, Xie G, Liu T, Deng S, Yuan W, He S, Wu D, Xu Y. Dipeptidase‑2 is a prognostic marker in lung adenocarcinoma that is correlated with its sensitivity to cisplatin. Oncol Rep 2023; 50:161. [PMID: 37449493 PMCID: PMC10360146 DOI: 10.3892/or.2023.8598] [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: 02/17/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Lung cancer accounts for the highest percentage of cancer morbidity and mortality worldwide, and lung adenocarcinoma (LUAD) is the most prevalent subtype. Although numerous therapies have been developed for lung cancer, patient prognosis is limited by tumor metastasis and more effective treatment targets are urgently required. In the present study, gene expression profiles were extracted from the Gene Expression Omnibus database and mRNA expression data were downloaded from The Cancer Genome Atlas database. In addition, TIMER 2.0 database was used to analyze the expression of genes in normal and multiple tumor tissues. Protein expression was confirmed using the Human Protein Atlas database and LUAD cell lines, sphere formation assay, western blotting, and a xenograft mouse model were used to confirm the bioinformatics analysis. Dipeptidase‑2 (DPEP2) expression was significantly decreased in LUAD and was negatively associated with prognosis. DPEP2 overexpression substantially inhibited epithelial‑mesenchymal transition (EMT) as well as LUAD cell metastasis, and limited the expression of the cancer stem cell transformation markers, CD44 and CD133. In addition, DPEP2 improved LUAD sensitivity to cisplatin by inhibiting EMT; this was verified in vitro and in vivo. These data indicated that DPEP2 upregulates E‑cadherin, thereby regulating cell migration, cancer stem cell transformation, and cisplatin resistance, ultimately affecting the survival of patients with LUAD. Overall, the findings of the present suggest that DPEP2 is important in the development of LUAD and can be used both as a prognostic marker and a target for future therapeutic research.
Collapse
Affiliation(s)
- Yuanyi Wang
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ting Zhang
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Hongfei Du
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Min Yang
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Guangsu Xie
- Clinical Laboratory, Xindu District People's Hospital of Chengdu, Chengdu, Sichuan 610500, P.R. China
| | - Teng Liu
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Shihua Deng
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Wei Yuan
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Shuang He
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Dongming Wu
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ying Xu
- College of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
5
|
Ishinaga H, Okugawa Y, Hou B, He F, Yin C, Murata M, Toiyama Y, Takeuchi K. The role of miR-21 as a predictive biomarker and a potential target to improve the effects of chemoradiotherapy against head and neck squamous cell carcinoma. JOURNAL OF RADIATION RESEARCH 2023:rrad043. [PMID: 37312615 DOI: 10.1093/jrr/rrad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Indexed: 06/15/2023]
Abstract
This study aimed to clarify whether circulating miR-21 represents a predictive biomarker in patients with head and neck squamous cell carcinoma (HNSCC) undergoing chemoradiotherapy, and to investigate the effect of miR-21 inhibitor for chemoradiation in human SCC cells. Plasma samples were obtained from 22 patients with HNSCC and 25 non-cancer volunteers. Plasma miR-21 expression was measured using real-time quantitative reverse transcription polymerase chain reaction. The effects of miR-21 inhibitor in human SCC cells were investigated by performing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry and western blot analysis. As a result, plasma miR-21 expression was higher in HNSCC patients than in control patients (P < 0.001). Seven patients with recurrence showed significantly higher plasma miR-21 than the 15 patients without recurrence. And high miR-21 expression group showed poor overall survival. Moreover, miR-21 inhibition significantly enhanced cisplatin- or radiation-induced apoptosis. Western blot analysis suggested the programmed cell death 4 protein as a potential target of miR-21 in relation to apoptosis. In conclusion, this study provides new insights into the role of miR-21 as a predictive biomarker for HNSCC treated with chemoradiotherapy and suggests a potential target to improve the effects of chemoradiotherapy against HNSCC.
Collapse
Affiliation(s)
- Hajime Ishinaga
- Department of Otorhinolaryngology - Head & Neck Surgery, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Edobashi, Tsu, Mie 514-8507, Japan
| | - Bo Hou
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Feng He
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology - Head & Neck Surgery, Mie University Graduate School of Medicine, Edobashi, Tsu, Mie 514-8507, Japan
| |
Collapse
|
6
|
Zeng C, Qi G, Shen Y, Li W, Zhu Q, Yang C, Deng J, Lu W, Liu Q, Jin J. DPEP1 promotes drug resistance in colon cancer cells by forming a positive feedback loop with ASCL2. Cancer Med 2022; 12:412-424. [PMID: 35670012 PMCID: PMC9844606 DOI: 10.1002/cam4.4926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/12/2022] [Accepted: 05/24/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Drug resistance is an important factor affecting the efficacy of chemotherapy in patients with colon cancer. However, clinical markers for diagnosing drug resistance of tumor cells are not only a few in number, but also low in specificity, and the mechanism of action of tumor cell drug resistance remains unclear. METHODS Dipeptidase 1 (DPEP1) expression was analyzed using the cancer genome atlas (TCGA) and genotype-Tissue Expression pan-cancer data. Survival analysis was performed using the survival package in R software to assess the prognostic value of DPEP1 expression in colon cancer. Correlation and Venn analyses were adopted to identify key genes. Immunohistochemistry, western blot, qRT-PCR, Co-immunoprecipitation, and dual-luciferase reporter experiments were carried out to explore the underlying associations between DPEP1 and Achaete scute-like 2 (ASCL2). MTT assays were used to evaluate the role of DPEP1 and ASCL2 in colon cancer drug resistance. RESULTS DPEP1 was highly expressed in colon cancer tissues. DPEP1 expression correlated negatively with disease-specific survival but not with overall survival. Bioinformatics analysis and experiments showed that the expressions of DPEP1 and ASCL2 in colon cancer tissues were markedly positively correlated. Mechanistic research indicated that DPEP1 enhanced the stability of protein ASCL2 by inhibiting its ubiquitination-mediated degradation. In turn, ASCL2 functioned as a transcription factor to activate the transcriptional activity of the DPEP1 gene and boost its expression. Furthermore, DPEP1 also could enhance the expression of colon cancer stem cell markers (LGR5, CD133, and CD44), which strengthened the tolerance of colon cancer cells to chemotherapy drugs. CONCLUSIONS Our findings reveal that the DPEP1 enhances the stemness of tumor cells by forming a positive feedback loop with ASCL2 to improve resistance to chemotherapy drugs.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina
| | - Guoping Qi
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina
| | - Ying Shen
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Wenjing Li
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Qi Zhu
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Chunxia Yang
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Jianzhong Deng
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Wenbin Lu
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Qian Liu
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| | - Jianhua Jin
- Department of OncologyWujin Hospital Affiliated with Jiangsu UniversityChangzhouJiangsu ProvinceChina,Department of OncologyWujin Clinical College of Xuzhou Medical UniversityChangzhouJiangsu ProvinceChina
| |
Collapse
|
7
|
Stratmann S, Yones SA, Garbulowski M, Sun J, Skaftason A, Mayrhofer M, Norgren N, Herlin MK, Sundström C, Eriksson A, Höglund M, Palle J, Abrahamsson J, Jahnukainen K, Munthe-Kaas MC, Zeller B, Tamm KP, Cavelier L, Komorowski J, Holmfeldt L. Transcriptomic analysis reveals proinflammatory signatures associated with acute myeloid leukemia progression. Blood Adv 2022; 6:152-164. [PMID: 34619772 PMCID: PMC8753201 DOI: 10.1182/bloodadvances.2021004962] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/23/2021] [Indexed: 11/20/2022] Open
Abstract
Numerous studies have been performed over the last decade to exploit the complexity of genomic and transcriptomic lesions driving the initiation of acute myeloid leukemia (AML). These studies have helped improve risk classification and treatment options. Detailed molecular characterization of longitudinal AML samples is sparse, however; meanwhile, relapse and therapy resistance represent the main challenges in AML care. To this end, we performed transcriptome-wide RNA sequencing of longitudinal diagnosis, relapse, and/or primary resistant samples from 47 adult and 23 pediatric AML patients with known mutational background. Gene expression analysis revealed the association of short event-free survival with overexpression of GLI2 and IL1R1, as well as downregulation of ST18. Moreover, CR1 downregulation and DPEP1 upregulation were associated with AML relapse both in adults and children. Finally, machine learning-based and network-based analysis identified overexpressed CD6 and downregulated INSR as highly copredictive genes depicting important relapse-associated characteristics among adult patients with AML. Our findings highlight the importance of a tumor-promoting inflammatory environment in leukemia progression, as indicated by several of the herein identified differentially expressed genes. Together, this knowledge provides the foundation for novel personalized drug targets and has the potential to maximize the benefit of current treatments to improve cure rates in AML.
Collapse
Affiliation(s)
| | - Sara A. Yones
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mateusz Garbulowski
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jitong Sun
- Department of Immunology, Genetics and Pathology and
| | - Aron Skaftason
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Markus Mayrhofer
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nina Norgren
- Department of Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Umeå University, Umeå, Sweden
| | - Morten Krogh Herlin
- Department of Clinical Medicine and
- Department of Pediatrics and Adolescent Medicine, Aarhus University, Aarhus, Denmark
| | | | | | | | - Josefine Palle
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Jonas Abrahamsson
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Kirsi Jahnukainen
- Children’s Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Monica Cheng Munthe-Kaas
- Norwegian Institute of Public Health, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Bernward Zeller
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Katja Pokrovskaja Tamm
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | - Jan Komorowski
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Umeå University, Umeå, Sweden
- Department of Clinical Medicine and
- Department of Pediatrics and Adolescent Medicine, Aarhus University, Aarhus, Denmark
- Department of Medical Sciences and
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Children’s Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Norwegian Institute of Public Health, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Swedish Collegium for Advanced Study, Uppsala, Sweden
- Institute of Computer Science, Polish Academy of Sciences, Warsaw, Poland
- Washington National Primate Research Center, Seattle, WA; and
| | - Linda Holmfeldt
- Department of Immunology, Genetics and Pathology and
- The Beijer Laboratory, Uppsala, Sweden
| |
Collapse
|
8
|
Ren X, Ma L, Wang N, Zhou R, Wu J, Xie X, Zhang H, Liu D, Ma X, Dang C, Kang H, Zhou Z. Antioxidant Gene Signature Impacts the Immune Infiltration and Predicts the Prognosis of Kidney Renal Clear Cell Carcinoma. Front Genet 2021; 12:721252. [PMID: 34490047 PMCID: PMC8416991 DOI: 10.3389/fgene.2021.721252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Oxidative stress is related to oncogenic transformation in kidney renal clear cell carcinoma (KIRC). We intended to identify a prognostic antioxidant gene signature and investigate its relationship with immune infiltration in KIRC. Methods: With the support of The Cancer Genome Atlas (TCGA) database, we researched the gene expression and clinical data of KIRC patients. Antioxidant related genes with significant differences in expression between KIRC and normal samples were then identified. Through univariate and multivariate Cox analysis, a prognostic gene model was established and all patients were divided into high- and low-risk subgroups. Single sample gene set enrichment analysis was adopted to analyze the immune infiltration, HLA expression, and immune checkpoint genes in different risk groups. Finally, the prognostic nomogram model was established and evaluated. Results: We identified six antioxidant genes significantly correlated with the outcome of KIRC patients as independent predictors, namely DPEP1 (HR = 0.97, P < 0.05), GSTM3 (HR = 0.97, P < 0.05), IYD (HR = 0.33, P < 0.05), KDM3B (HR = 0.96, P < 0.05), PRDX2 (HR = 0.99, P < 0.05), and PRXL2A (HR = 0.96, P < 0.05). The high- and low-risk subgroups of KIRC patients were grouped according to the six-gene signature. Patients with higher risk scores had poorer prognosis, more advanced grade and stage, and more abundance of M0 macrophages, regulatory T cells, and follicular helper T cells. There were statistically significant differences in HLA and checkpoint gene expression between the two risk subgroups. The performance of the nomogram was favorable (concordance index = 0.766) and reliably predicted the 3-year (AUC = 0.792) and 5-year (AUC = 0.766) survival of patients with KIRC. Conclusion: The novel six antioxidant related gene signature could effectively forecast the prognosis of patients with KIRC, supply insights into the interaction between cellular antioxidant mechanisms and cancer, and is an innovative tool for selecting potential patients and targets for immunotherapy.
Collapse
Affiliation(s)
- Xueting Ren
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nan Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruina Zhou
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianhua Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Xie
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Di Liu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhangjian Zhou
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Li ZX, Chen W, Qin M, Wang LX, Zeng YQ. Characteristics of circRNAs expression profiles in the piglets intestine induced by oxidative stress. Genes Genomics 2021; 44:425-433. [PMID: 34449066 DOI: 10.1007/s13258-021-01154-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUD Oxidative stress (OS) can affect the expression of key genes and destroy the intestinal structure. However, it is unclear how OS regulates the expression of circular RNAs (circRNAs), microRNAs (miRNAs) and mRNAs. OBJECTIVE The aim of this study was to examine the expression of circRNAs, miRNAs and mRNAs exposed to OS. METHODS Piglets were exposed to diquat (DQ), a herbicide, and the activity of antioxidant enzymes and the morphology of the intestine were investigated. We utilized whole transcriptome sequencing to examine the global expression of circRNAs, miRNAs and mRNAs in the jejunum. RESULTS Compared to controls, 751 circRNAs, 731 miRNAs and 164 mRNAs were differentially expressed in diquat-treated piglets. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that oxidative phosphorylation, RNA degradation and ubiquitin-mediated proteolysis were closely associated with OS. CONCLUSIONS Our results indicated that diquat-induced OS alters the intestinal structure, resulting in the differential expression of circRNAs, miRNAs and mRNAs in the jejunum of piglets. Meanwhile, OS weakened the enzyme antioxidant system in serum of piglets. Our results provide a foundation for further studies on the mechanisms involved in the response to OS in the jejunum.
Collapse
Affiliation(s)
- Zhi-Xin Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, No. 61 DaiZong Street, Tai'an City, 271018, Shandong Province, People's Republic of China
| | - Wei Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, No. 61 DaiZong Street, Tai'an City, 271018, Shandong Province, People's Republic of China
| | - Ming Qin
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, No. 61 DaiZong Street, Tai'an City, 271018, Shandong Province, People's Republic of China
| | - Li-Xue Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, No. 61 DaiZong Street, Tai'an City, 271018, Shandong Province, People's Republic of China
| | - Yong-Qing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, No. 61 DaiZong Street, Tai'an City, 271018, Shandong Province, People's Republic of China.
| |
Collapse
|
10
|
Choong WK, Sung TY. Somatic mutation subtypes of lung adenocarcinoma in East Asian reveal divergent biological characteristics and therapeutic vulnerabilities. iScience 2021; 24:102522. [PMID: 34142036 PMCID: PMC8188494 DOI: 10.1016/j.isci.2021.102522] [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: 12/23/2020] [Revised: 03/25/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Lung adenocarcinoma (LUAD) patients in East Asia predominantly harbor oncogenic EGFR mutations. However, there remains a limited understanding of the biological characteristics and therapeutic vulnerabilities of the concurrent mutations of EGFR and other genes in LUAD. Here, we performed comprehensive bioinformatics analyses on 88 treatment-naïve East Asian LUAD patients. Based on somatic mutation clustering, we identified three somatic mutation subtypes: EGFR + TP53 co-mutation, EGFR mutation, and multiple-gene mutation. A proteogenomic analysis among subtypes revealed varying degrees of dysregulation in cell-cycle-related and immune-related processes. An immune-characteristic analysis revealed higher PDL1 protein expression in the EGFR + TP53 co-mutation subtype than in the EGFR mutation subtype, which may affect the therapeutic efficacy of anti-PD-L1 therapy. Moreover, integrating known and potential therapeutic target analysis reveals therapeutic vulnerabilities of specific subtypes and nominates candidate biomarkers for therapeutic intervention. This study provides new biological insight and therapeutic opportunities with respect to EGFR-mutant LUAD subtypes. Comprehensive clustering analysis reveals three somatic mutation subtypes Prognosis of EGFRmut/TP53mut subtype is worse than EGFRmut subtype EGFRmut/TP53mut subtype shows IFN signaling and antigen processing pathway signatures Proteome analysis identifies druggable proteins and candidates for drug repositioning
Collapse
Affiliation(s)
- Wai-Kok Choong
- Institute of Information Science, Academia Sinica, Taipei, 115, Taiwan
| | - Ting-Yi Sung
- Institute of Information Science, Academia Sinica, Taipei, 115, Taiwan
| |
Collapse
|
11
|
Rossouw SC, Bendou H, Blignaut RJ, Bell L, Rigby J, Christoffels A. Evaluation of Protein Purification Techniques and Effects of Storage Duration on LC-MS/MS Analysis of Archived FFPE Human CRC Tissues. Pathol Oncol Res 2021; 27:622855. [PMID: 34257588 PMCID: PMC8262168 DOI: 10.3389/pore.2021.622855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/01/2021] [Indexed: 12/17/2022]
Abstract
To elucidate cancer pathogenesis and its mechanisms at the molecular level, the collecting and characterization of large individual patient tissue cohorts are required. Since most pathology institutes routinely preserve biopsy tissues by standardized methods of formalin fixation and paraffin embedment, these archived FFPE tissues are important collections of pathology material that include patient metadata, such as medical history and treatments. FFPE blocks can be stored under ambient conditions for decades, while retaining cellular morphology, due to modifications induced by formalin. However, the effect of long-term storage, at resource-limited institutions in developing countries, on extractable protein quantity/quality has not yet been investigated. In addition, the optimal sample preparation techniques required for accurate and reproducible results from label-free LC-MS/MS analysis across block ages remains unclear. This study investigated protein extraction efficiency of 1, 5, and 10-year old human colorectal carcinoma resection tissue and assessed three different gel-free protein purification methods for label-free LC-MS/MS analysis. A sample size of n = 17 patients per experimental group (with experiment power = 0.7 and α = 0.05, resulting in 70% confidence level) was selected. Data were evaluated in terms of protein concentration extracted, peptide/protein identifications, method reproducibility and efficiency, sample proteome integrity (due to storage time), as well as protein/peptide distribution according to biological processes, cellular components, and physicochemical properties. Data are available via ProteomeXchange with identifier PXD017198. The results indicate that the amount of protein extracted is significantly dependent on block age (p < 0.0001), with older blocks yielding less protein than newer blocks. Detergent removal plates were the most efficient and overall reproducible protein purification method with regard to number of peptide and protein identifications, followed by the MagReSyn® SP3/HILIC method (with on-bead enzymatic digestion), and lastly the acetone precipitation and formic acid resolubilization method. Overall, the results indicate that long-term storage of FFPE tissues (as measured by methionine oxidation) does not considerably interfere with retrospective proteomic analysis (p > 0.1). Block age mainly affects initial protein extraction yields and does not extensively impact on subsequent label-free LC-MS/MS analysis results.
Collapse
Affiliation(s)
- Sophia C. Rossouw
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Hocine Bendou
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Renette J. Blignaut
- Department of Statistics and Population Studies, University of the Western Cape, Bellville, South Africa
| | - Liam Bell
- Centre for Proteomic and Genomic Research, Observatory, Cape Town, South Africa
| | - Jonathan Rigby
- Division of Anatomical Pathology, Department of Pathology, Faculty of Health Sciences, University of Stellenbosch, National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| | - Alan Christoffels
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
12
|
Abstract
Hepatoblastoma (HB) is the predominant primary liver tumor in children. While the prognosis is favorable when the tumor can be resected, the outcome is dismal for patients with progressed HB. Therefore, a better understanding of the molecular mechanisms responsible for HB is imperative for early detection and effective treatment. Sequencing analysis of human HB specimens unraveled the pivotal role of Wnt/β-catenin pathway activation in this disease. Nonetheless, β-catenin activation alone does not suffice to induce HB, implying the need for additional alterations. Perturbations of several pathways, including Hippo, Hedgehog, NRF2/KEAP1, HGF/c-Met, NK-1R/SP, and PI3K/AKT/mTOR cascades and aberrant activation of c-MYC, n-MYC, and EZH2 proto-oncogenes, have been identified in HB, although their role requires additional investigation. Here, we summarize the current knowledge on HB molecular pathogenesis, the relevance of the preclinical findings for the human disease, and the innovative therapeutic strategies that could be beneficial for the treatment of HB patients.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Antonio Solinas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefano Cairo
- XenTech, Evry, France,Istituto di Ricerca Pediatrica, Padova, Italy
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
13
|
Liu Q, Deng J, Yang C, Wang Y, Shen Y, Zhang H, Ding Z, Zeng C, Hou Y, Lu W, Jin J. DPEP1 promotes the proliferation of colon cancer cells via the DPEP1/MYC feedback loop regulation. Biochem Biophys Res Commun 2020; 532:520-527. [PMID: 32896379 DOI: 10.1016/j.bbrc.2020.08.063] [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: 08/12/2020] [Accepted: 08/15/2020] [Indexed: 12/18/2022]
Abstract
DPEP1 is highly expressed in the colorectal carcinoma tissues and colon cancer cells. However, the function and underlying mechanism of DPEP1 in the colon cancer cells are still poorly understood. Here, we found that transcription factor MYC could occupy on the DPEP1 promoter and activate its activities, and DPEP1 was up-regulated by MYC proteins in mRNA and protein levels in a dose-dependent manner in colon cancer cells. The expression levels of DPEP1 were positively correlated with that of MYC in colorectal tumor tissues. Moreover, Laser confocal images and Co-immunoprecipitation (Co-IP) revealed that DPEP1 and MYC proteins could bind to each other in the colon cancer cells. In turn, DPEP1 could enhance the stability of MYC proteins by extending the half-life of MYC proteins in colon cancer cells. Thus, DPEP1 and MYC proteins might form a positive feedback loop to maintain their high expression levels in colon cancer cells. In function, the MTT, EdU, Clone Formation assays and xenograft tumors assays demonstrated that DPEP1 could boost the proliferation of colon cancer cells through the DPEP1/MYC positive feedback loop in vitro and in vivo. Theoretically, DPEP1 may serve as a colon cancer biomarker and a novel target of colorectal carcinogenesis therapy.
Collapse
Affiliation(s)
- Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Jianzhong Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Chunxia Yang
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Yue Wang
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Ying Shen
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Hua Zhang
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Zhixiang Ding
- Department of Clinical Laboratory, Changzhou Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Changzhou, 213003, China
| | - Cheng Zeng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China
| | - Yongzhong Hou
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Institute of Life Sciences of the Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China.
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu Province, 213017, China; Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 213017, China.
| |
Collapse
|
14
|
Gao X, Yang J. Identification of Genes Related to Clinicopathological Characteristics and Prognosis of Patients with Colorectal Cancer. DNA Cell Biol 2020; 39:690-699. [PMID: 32027181 DOI: 10.1089/dna.2019.5088] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to identify genes with clinical significance in colorectal cancer (CRC). Gene expression profiles of 585 CRC tissues and 61 normal colorectal tissues from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases were used to identify differentially expressed genes (DEGs) between CRC and normal colorectal tissues. DAVID and KOBAS tools were used to explore Gene Ontology (GO) and KEGG pathways enriched by DEGs, respectively. In addition, TCGA data sets were also used to identify prognostic factors and develop a prognostic prediction model for CRC. A total of 353 DEGs including 117 upregulated and 236 downregulated genes in CRC were identified based on GSE32323 data set. These DEGs were significantly enriched in the biological process related to the regulation of cell proliferation and 50 signaling pathways, such as "TGF-beta signaling pathway," "Wnt signaling pathway," and "Jak-STAT signaling pathway." GCG, ADH1B, SLC4A4, ZG16, and CLCA4 were the top five downregulated in CRC. FOXQ1, LGR5, CLDN1, KRT23, and DPEP1 were the top five upregulated in CRC. KRT23 expression could affect tumor stage and regional lymph node metastasis in CRC patients. FOXQ1 expression could affect tumor distant metastasis in CRC patients. Survival analysis indicated that SLC4A4 expression was associated with the prognosis of CRC patients. Prognostic prediction model developed based on age, tumor stage, and SLC4A4 expression exhibited an efficient performance in predicting 1-, 3-, and 5-year overall survival of CRC patients. In conclusion, the current study identified several genes and pathways related to CRC, which provided new insight in understanding molecular mechanism of tumorigenesis and development of CRC.
Collapse
Affiliation(s)
- Xueren Gao
- School of Pharmacy, Yancheng Teachers' University, Yancheng, China
| | - Jiaojiao Yang
- Department of Microbiology and Immunology, Shanxi Medical University, Tai yuan, China
| |
Collapse
|
15
|
Zhu Y, Huo X, Wang C, Meng Q, Liu Z, Sun H, Tan A, Ma X, Peng J, Liu K. Organic anion transporters also mediate the drug-drug interaction between imipenem and cilastatin. Asian J Pharm Sci 2020; 15:252-263. [PMID: 32373203 PMCID: PMC7193450 DOI: 10.1016/j.ajps.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/03/2018] [Accepted: 11/17/2018] [Indexed: 12/11/2022] Open
Abstract
This study aimed to clarify that organic anion transporters (OATs) mediate the drug-drug interaction (DDI) between imipenem and cilastatin. After co-administration with imipenem, the plasma concentrations and the plasma concentration-time curve (AUC) of cilastatin were significantly increased, while renal clearance and cumulative urinary excretion of cilastatin were decreased. At the same time, imipenem significantly inhibited the uptake of cilastatin in rat kidney slices and in human OAT1 (hOAT1)-HEK293 and human OAT3 (hOAT3)-HEK293 cells. Probenecid, p-aminohippurate, and benzylpenicillin inhibited the uptake of imipenem and cilastatin in rat kidney slices and in hOAT1- and hOAT3-HEK 293 cells, respectively. The uptakes of imipenem and cilastatin in hOAT1- and hOAT3-HEK 293 cells were significantly higher than that in mock-HEK-293 cells. Moreover, the Km values of cilastatin were increased in the presence of imipenem with unchanged Vmax , indicating that imipenem inhibited the uptake of cilastatin in a competitive manner. When imipenem and cilastatin were co-administered, the level of imipenem was higher compared with imipenem alone both in vivo and in vitro. But, cilastatin significantly inhibited the uptake of imipenem when dehydropeptidase-1 (DPEP1) was silenced by RNAi technology in hOAT1- and hOAT3-HEK 293 cells. In conclusion, imipenem and cilastatin are the substrates of OAT1 and OAT3. OAT1 and OAT3 mediate the DDI between imipenem and cilastatin. Meanwhile, cilastatin also reduces the hydrolysis of imipenem by inhibiting the uptake of imipenem mediated by OAT1 and OAT3 in the kidney as a complement.
Collapse
Affiliation(s)
- Yanna Zhu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaokui Huo
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Zhihao Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Aiping Tan
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaodong Ma
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Jinyong Peng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
- College (Institute) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
16
|
Zhang J, Xu Y, Gale RP, Wu L, Zhang J, Feng Y, Qin Y, Jiang H, Jiang Q, Jiang B, Liu Y, Chen Y, Wang Y, Zhang X, Xu L, Huang X, Liu K, Ruan G. DPEP1 expression promotes proliferation and survival of leukaemia cells and correlates with relapse in adults with common B cell acute lymphoblastic leukaemia. Br J Haematol 2020; 190:67-78. [PMID: 32068254 DOI: 10.1111/bjh.16505] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/29/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Jia‐Min Zhang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Yan Xu
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Robert P. Gale
- Haematology Research Center Division of Experimental Medicine Department of Medicine Imperial College London London UK
| | - Li‐Xin Wu
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Jing Zhang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Yong‐Huai Feng
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Ya‐Zhen Qin
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Hao Jiang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Qian Jiang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Bin Jiang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Yan‐Rong Liu
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Yu‐Hong Chen
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Yu Wang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Xiao‐Hui Zhang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Lan‐Ping Xu
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Xiao‐Jun Huang
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
- Peking‐Tsinghua Center for Life Sciences Academy for Advanced Interdisciplinary StudiesPeking University Beijing China
| | - Kai‐Yan Liu
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| | - Guo‐Rui Ruan
- National Clinical Research Center for Hematologic Disease Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation Collaborative Innovation Center of Hematology Peking University People's Hospital Peking University Institute of Hematology Beijing China
| |
Collapse
|
17
|
Zhang F, Li X, Ni Y, Shan G, Gao Y. Preliminary study of the urinary proteome in Li and Han ethnic individuals from Hainan. SCIENCE CHINA. LIFE SCIENCES 2020; 63:125-137. [PMID: 31102176 DOI: 10.1007/s11427-018-9485-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023]
Abstract
Biomarkers indicate changes associated with disease. Blood is relatively stable due to the homeostatic mechanisms of the body; however, urine accumulates metabolites from changes in the body, making it a better source for early biomarker discovery. The Li ethnic group is a unique minority ethnic group that has only lived on Hainan Island for approximately 5,000 years. Studies have shown that various specific genetic variations are different between the Li and Han ethnic groups. However, whether the urinary proteome between these two ethnic groups is significantly different remains unknown. In this study, differential urinary proteins were identified in the Li and Han ethnic groups using liquid chromatography tandem mass spectrometry (LC-MS/MS). In total, 1,555 urinary proteins were identified. Twenty-five of the urinary proteins were statistically significantly different, 16 of which have been previously reported to be biomarkers of many diseases, and that these significantly different proteins were caused by ethnic differences rather than random differences. Ethnic group differences may be an influencing factor in urine proteome studies and should be considered when human urine samples are used for biomarker discovery.
Collapse
Affiliation(s)
- Fanshuang Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xundou Li
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yanying Ni
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Guangliang Shan
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, 100875, China.
| |
Collapse
|
18
|
Clinical significance and biological role of L1 cell adhesion molecule in gastric cancer. Br J Cancer 2019; 121:1058-1068. [PMID: 31754264 PMCID: PMC6964673 DOI: 10.1038/s41416-019-0646-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Background L1 cell adhesion molecule (L1CAM) is highly expressed in malignant tumours and might play a pivotal role in tumour progression. Methods We analysed by immunohistochemistry L1CAM protein expression in formalin-fixed, paraffin-embedded specimens from 309 GC patients. We performed propensity score matching (PSM) analysis to clarify the prognostic impact of L1CAM in GC patients. We evaluated L1CAM gene expression in fresh frozen specimens from another group of 131 GC patients to establish its clinical relevance. The effects of changes in L1CAM were investigated in vitro and in vivo. Results L1CAM was mainly expressed in tumour cells of GC tissues. Elevated L1CAM expression was an independent prognostic factor for overall and disease-free survival, and an independent risk factor for distant metastasis in GC patients. PSM analysis showed that high L1CAM expression was significantly associated with poor prognosis. L1CAM gene expression using fresh frozen specimens successfully validated all of these findings in an independent cohort. Inhibition of L1CAM suppressed cell proliferation, cycle progress, invasion, migration and anoikis resistance in GC cells. Furthermore, L1CAM inhibition suppressed the growth of peritoneal metastasis. Conclusion L1CAM may serve as a feasible biomarker for identification of patients who have a high risk of recurrence of GC.
Collapse
|
19
|
Cui X, Liu X, Han Q, Zhu J, Li J, Ren Z, Liu L, Luo Y, Wang Z, Zhang D, Fan Y, Zhang D, Dong G. DPEP1 is a direct target of miR-193a-5p and promotes hepatoblastoma progression by PI3K/Akt/mTOR pathway. Cell Death Dis 2019; 10:701. [PMID: 31541079 PMCID: PMC6754441 DOI: 10.1038/s41419-019-1943-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/26/2019] [Accepted: 08/30/2019] [Indexed: 02/08/2023]
Abstract
Hepatoblastoma (HB) is the most common hepatic neoplasm in childhood and the therapeutic outcomes remain undesirable due to its recurrence and metastasis. Increasing evidence shows that dipeptidase 1 (DPEP1) has pivotal function in tumorigenesis in multiple tumors. However, the expression pattern, biological function, and underlying mechanism of DPEP1 in HB have not been reported. Here we showed that DPEP1 was significantly upregulated and was associated with poor prognosis in HB patients. In vitro and in vivo assays indicated that silencing DPEP1 significantly suppressed HB cell proliferation, migration, and invasion, while DPEP1 overexpression exhibited the opposite effect. In addition, we identified that DPEP1 was a direct target of microRNA-193a-5p (miR-193a-5p). Functional experiments demonstrated that overexpression of miR-193a-5p significantly inhibited cell proliferation and invasion of HB cells, while the inhibitory effect could be reversed by DPEP1 overexpression. Moreover, miR-193a-5p was decreased in HB tumor tissues and associated with a poor clinical prognosis. Mechanistically, our results indicated that the miR-193a-5p/DPEP1 axis participated to the progression of HB via regulating the PI3K/Akt/mTOR (phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin) signaling. In conclusion, our findings suggest that the miR-193a-5p /DPEP1 axis might be a good prognostic predictor and therapeutic target in HB.
Collapse
Affiliation(s)
- Xichun Cui
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Xin Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Qicai Han
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Jianming Zhu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Jianhao Li
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Zhigang Ren
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Liwen Liu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yanbing Luo
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Zhifang Wang
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Dandan Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yingzhong Fan
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| | - Gang Dong
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
- Department of Ultrasonography, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| |
Collapse
|
20
|
Duijvis NW, Moerland PD, Kunne C, Slaman MMW, van Dooren FH, Vogels EW, de Jonge WJ, Meijer SL, Fluiter K, te Velde AA. Inhibition of miR-142-5P ameliorates disease in mouse models of experimental colitis. PLoS One 2017; 12:e0185097. [PMID: 29059189 PMCID: PMC5653202 DOI: 10.1371/journal.pone.0185097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are epigenetically involved in regulating gene expression. They may be of importance in the pathogenesis of inflammatory bowel disease (IBD). The aim of this study was to determine the role of miRNAs by their specific blocking in the CD4+CB45RBhi T-cell transfer model of chronic experimental colitis. METHODS Colitis caused by transfer of WT CD4+CD45RBhi T cells in severe combined immunodeficiency (SCID) mice shares many features with human IBD. Colonic miRNA expression levels were measured at three time points in colitic mice, where a time-dependent upregulation of multiple miRNAs was seen. To inhibit these miRNAs, specific locked-nucleic-acid-modified (LNA) oligonucleotides were administered in further experiments at the moment the mice demonstrated the first signs of colitis. As controls, PBS and a scrambled sequence of anti-miRNA were used. Genome-wide expression analyses were also performed in order to detect candidate target genes of miR-142-5p, of which inhibition resulted in most effective amelioration of colitis. RESULTS Anti-miR-142-5p reduced colitis and related wasting disease when administered in the T-cell transfer model, reflected in reduced weight loss and a lower disease activity index (DAI). In further validation experiments we also observed a higher survival rate and less colonic histological inflammation in the antagomir-treated mice. Moreover, by genome-wide expression analyses, we found downstream activation of the anti-inflammatory IL10RA pathway, including three genes also found in the top-20 candidate target genes of miR-142-5p. CONCLUSION In conclusion, CD4+CD45RBhi-transfer colitis induces miR-142-5p. Blocking miR-142-5p reduced colitis and prevented wasting disease, possibly by activation of the IL10RA pathway.
Collapse
Affiliation(s)
- Nicolette W. Duijvis
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
- * E-mail: (NWD); (AAV)
| | - Perry D. Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, the Netherlands
| | - Cindy Kunne
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Monique M. W. Slaman
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Faas H. van Dooren
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Esther W. Vogels
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Sybren L. Meijer
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Anje A. te Velde
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
- * E-mail: (NWD); (AAV)
| |
Collapse
|
21
|
Tu C, Mojica W, Straubinger RM, Li J, Shen S, Qu M, Nie L, Roberts R, An B, Qu J. Quantitative proteomic profiling of paired cancerous and normal colon epithelial cells isolated freshly from colorectal cancer patients. Proteomics Clin Appl 2017; 11:10.1002/prca.201600155. [PMID: 27943637 PMCID: PMC5418098 DOI: 10.1002/prca.201600155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 11/03/2016] [Accepted: 12/06/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE The heterogeneous structure in tumor tissues from colorectal cancer (CRC) patients excludes an informative comparison between tumors and adjacent normal tissues. Here, we develop and apply a strategy to compare paired cancerous (CEC) versus normal (NEC) epithelial cells enriched from patients and discover potential biomarkers and therapeutic targets for CRC. EXPERIMENTAL DESIGN CEC and NEC cells are respectively isolated from five different tumor and normal locations in the resected colon tissue from each patient (N = 12 patients) using an optimized epithelial cell adhesion molecule (EpCAM)-based enrichment approach. An ion current-based quantitative method is employed to perform comparative proteomic analysis for each patient. RESULTS A total of 458 altered proteins that are common among >75% of patients are observed and selected for further investigation. Besides known findings such as deregulation of mitochondrial function, tricarboxylic acid cycle, and RNA post-transcriptional modification, functional analysis further revealed RAN signaling pathway, small nucleolar ribonucleoproteins (snoRNPs), and infection by RNA viruses are altered in CEC cells. A selection of the altered proteins of interest is validated by immunohistochemistry analyses. CONCLUSION AND CLINICAL RELEVANCE The informative comparison between matched CEC and NEC enhances our understanding of molecular mechanisms of CRC development and provides biomarker candidates and new pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Chengjian Tu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| | - Wilfrido Mojica
- Department of Pathology, State University of New York at Buffalo, State University of New York, Buffalo, NY 14260 USA
| | - Robert M. Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| | - Jun Li
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| | - Miao Qu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- Beijing University of Chinese Medicine, Beijing, China, 100029
| | - Lei Nie
- School of pharmaceutical sciences, Shandong University, 44 Wenhua West Road, Jinan, China, 250012
| | - Rick Roberts
- Department of Structural Biology, State University of New York at Buffalo, State University of New York, Buffalo, NY 14260 USA
| | - Bo An
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260 USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203 USA
| |
Collapse
|
22
|
Profiles of microRNA networks in intestinal epithelial cells in a mouse model of colitis. Sci Rep 2015; 5:18174. [PMID: 26647826 PMCID: PMC4673535 DOI: 10.1038/srep18174] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) accompany a critical loss of the frontline barrier function that is achieved primarily by intestinal epithelial cells (IECs). Although the gene-regulation pathways underlying these host-defense roles of IECs presumably are deranged during IBD pathogenesis, the quantitative and qualitative alterations of posttranscriptional regulators such as microRNAs (miRNAs) within the cells largely remain to be defined. We aimed to uncover the regulatory miRNA–target gene relationships that arise differentially in inflamed small- compared with large-IECs. Whereas IBD significantly increased the expression of only a few miRNA candidates in small-IECs, numerous miRNAs were upregulated in inflamed large-IECs. These marked alterations might explain why the large, as compared with small, intestine is more sensitive to colitis and shows more severe pathology in this experimental model of IBD. Our in-depth assessment of the miRNA–mRNA expression profiles and the resulting networks prompts us to suggest that miRNAs such as miR-1224, miR-3473a, and miR-5128 represent biomarkers that appear in large-IECs upon IBD development and co-operatively repress the expression of key anti-inflammatory factors. The current study provides insight into gene-regulatory networks in IECs through which dynamic rearrangement of the involved miRNAs modulates the gene expression–regulation machinery between maintaining and disrupting gastrointestinal homeostasis.
Collapse
|
23
|
Raimondo F, Corbetta S, Savoia A, Chinello C, Cazzaniga M, Rocco F, Bosari S, Grasso M, Bovo G, Magni F, Pitto M. Comparative membrane proteomics: a technical advancement in the search of renal cell carcinoma biomarkers. MOLECULAR BIOSYSTEMS 2015; 11:1708-16. [DOI: 10.1039/c5mb00020c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Set-up of a specific protocol for membrane protein analysis, applied to label free, comparative proteomics of renal cell carcinoma microdomains.
Collapse
Affiliation(s)
| | | | - Andrea Savoia
- Department of Health Sciences
- Univ. of Milano-Bicocca
- Monza
- Italy
| | - Clizia Chinello
- Department of Health Sciences
- Univ. of Milano-Bicocca
- Monza
- Italy
| | - Marta Cazzaniga
- Department of Health Sciences
- Univ. of Milano-Bicocca
- Monza
- Italy
| | - Francesco Rocco
- Department of Specialistic Surgical Sciences
- Urology unit
- Ospedale Maggiore Policlinico Foundation
- IRCCS
- Milano
| | - Silvano Bosari
- Department of Medicine
- Surgery and Dental Sciences
- Pathology Unit
- Ospedale Maggiore Policlinico Foundation Milano
- IRCCS
| | - Marco Grasso
- Department of Surgical Pathology
- Cytology
- Medical Genetics and Nephropathology
- Azienda Ospedaliera San Gerardo
- Monza
| | - Giorgio Bovo
- Department of Surgical Pathology
- Cytology
- Medical Genetics and Nephropathology
- Azienda Ospedaliera San Gerardo
- Monza
| | - Fulvio Magni
- Department of Health Sciences
- Univ. of Milano-Bicocca
- Monza
- Italy
| | - Marina Pitto
- Department of Health Sciences
- Univ. of Milano-Bicocca
- Monza
- Italy
| |
Collapse
|
24
|
Sane S, Abdullah A, Boudreau DA, Autenried RK, Gupta BK, Wang X, Wang H, Schlenker EH, Zhang D, Telleria C, Huang L, Chauhan SC, Rezvani K. Ubiquitin-like (UBX)-domain-containing protein, UBXN2A, promotes cell death by interfering with the p53-Mortalin interactions in colon cancer cells. Cell Death Dis 2014; 5:e1118. [PMID: 24625977 PMCID: PMC3973214 DOI: 10.1038/cddis.2014.100] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 02/02/2014] [Accepted: 02/07/2014] [Indexed: 02/08/2023]
Abstract
Mortalin (mot-2) induces inactivation of the tumor suppressor p53's transcriptional and apoptotic functions by cytoplasmic sequestration of p53 in select cancers. The mot-2-dependent cytoprotective function enables cancer cells to support malignant transformation. Abrogating the p53-mot-2 interaction can control or slow down the growth of cancer cells. In this study, we report the discovery of a ubiquitin-like (UBX)-domain-containing protein, UBXN2A, which binds to mot-2 and consequently inhibits the binding between mot-2 and p53. Genetic analysis showed that UBXN2A binds to mot-2's substrate binding domain, and it partly overlaps p53's binding site indicating UBXN2A and p53 likely bind to mot-2 competitively. By binding to mot-2, UBXN2A releases p53 from cytosolic sequestration, rescuing the tumor suppressor functions of p53. Biochemical analysis and functional assays showed that the overexpression of UBXN2A and the functional consequences of unsequestered p53 trigger p53-dependent apoptosis. Cells expressing shRNA against UBXN2A showed the opposite effect of that seen with UBXN2A overexpression. The expression of UBXN2A and its apoptotic effects were not observed in normal colonic epithelial cells and p53-/- colon cancer cells. Finally, significant reduction in tumor volume in a xenograft mouse model in response to UBXN2A expression was verified in vivo. Our results introduce UBXN2A as a home defense response protein, which can reconstitute inactive p53-dependent apoptotic pathways. Inhibition of mot-2-p53 interaction by UBXN2A is an attractive therapeutic strategy in mot-2-elevated tumors.
Collapse
Affiliation(s)
- S Sane
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - A Abdullah
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - D A Boudreau
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - R K Autenried
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - B K Gupta
- Department of Pharmaceutical Sciences, Cancer Research Center, University of Tennessee Health Science Center, 19S Manassas Avenue, Memphis, TN, USA
| | - X Wang
- Departments of Physiology & Biophysics, University of California, Irvine, CA, USA
| | - H Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - E H Schlenker
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - D Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - C Telleria
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| | - L Huang
- Departments of Physiology & Biophysics, University of California, Irvine, CA, USA
| | - S C Chauhan
- Department of Pharmaceutical Sciences, Cancer Research Center, University of Tennessee Health Science Center, 19S Manassas Avenue, Memphis, TN, USA
| | - K Rezvani
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, USA
| |
Collapse
|
25
|
Monoclonal antibody binding-site diversity assessment with a cell-based clustering assay. J Immunol Methods 2014; 405:1-14. [DOI: 10.1016/j.jim.2013.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 12/12/2013] [Accepted: 12/19/2013] [Indexed: 12/11/2022]
|
26
|
Sponziello M, Lavarone E, Pegolo E, Di Loreto C, Puppin C, Russo MA, Bruno R, Filetti S, Durante C, Russo D, Di Cristofano A, Damante G. Molecular differences between human thyroid follicular adenoma and carcinoma revealed by analysis of a murine model of thyroid cancer. Endocrinology 2013; 154:3043-53. [PMID: 23751876 PMCID: PMC3749486 DOI: 10.1210/en.2013-1028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Mouse models can provide useful information to understand molecular mechanisms of human tumorigenesis. In this study, the conditional thyroid mutagenesis of Pten and Ras genes in the mouse, which induces very aggressive follicular carcinomas (FTCs), has been used to identify genes differentially expressed among human normal thyroid tissue (NT), follicular adenoma (FA), and FTC. Global gene expression of mouse FTC was compared with that of mouse normal thyroids: 911 genes were found deregulated ± 2-fold in FTC samples. Then the expression of 45 deregulated genes in mouse tumors was investigated by quantitative RT-PCR in a first cohort of human NT, FA, and FTC (discovery group). Five genes were found significantly down-regulated in FA and FTC compared with NT. However, 17 genes were found differentially expressed between FA and FTC: 5 and 12 genes were overexpressed and underexpressed in FTC vs FA, respectively. Finally, 7 gene products, selected from results obtained in the discovery group, were investigated in a second cohort of human tumors (validation group) by immunohistochemistry. Four proteins showed significant differences between FA and FTC (peroxisomal proliferator-activated receptor-γ, serum deprivation response protein, osteoglycin, and dipeptidase 1). Altogether our data indicate that the establishment of an enriched panel of molecular biomarkers using data coming from mouse thyroid tumors and validated in human specimens may help to set up a more valid platform to further improve diagnosis and prognosis of thyroid malignancies.
Collapse
Affiliation(s)
- Marialuisa Sponziello
- Dipartimento di Medicina Interna e Specialità Mediche, Università di Roma “Sapienza,” 00161 Roma, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Eisenach PA, Soeth E, Röder C, Klöppel G, Tepel J, Kalthoff H, Sipos B. Dipeptidase 1 (DPEP1) is a marker for the transition from low-grade to high-grade intraepithelial neoplasia and an adverse prognostic factor in colorectal cancer. Br J Cancer 2013; 109:694-703. [PMID: 23839495 PMCID: PMC3738143 DOI: 10.1038/bjc.2013.363] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/13/2013] [Accepted: 06/17/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Improvements in the understanding of its molecular mechanism and the characterisation of CRC-specific biomarkers facilitating early detection are considered to increase overall survival. METHODS A meta-analysis of microarray and Serial Analysis of Gene Expression (SAGE) has been performed to identify differentially regulated genes in CRC. Dipeptidase 1 (DPEP1/MDP/RDP) and Syntenin-2 (SDCBP2/SITAC18) were found to be differentially expressed in tumour tissue compared with normal mucosa. Expression of DPEP1 was assessed in a validation set of 87 normal mucosa samples, 20 hyperplastic polyps, 46 CR adenomas with low- and high-grade intraepithelial neoplasia (IEN) and 217 well-documented CRCs by immunohistochemistry and partially by immunoblotting and real-time PCR. RESULTS Expression of DPEP1 was specifically increased in human CRC tissue samples compared with normal mucosa (P<0.0001, Mann-Whitney U-test), showing a striking upregulation in high-grade compared with low-grade IEN. Furthermore, high DPEP1 expression was found to strongly correlate with histological stage (P<0.0001, chi-square test) as well as localisation (P<0.0001, chi-square test) and has been recognised as an independent adverse prognostic factor, showing significant prognostic values with an ROC (receiver operating characteristic)-AUC of 0.9230. CONCLUSION Dipeptidase 1 has been identified as an excellent marker of high-grade IEN and CRC, and may thus be applied for screening of early neoplastic lesions and for prognostic stratification.
Collapse
Affiliation(s)
- P A Eisenach
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Background: Brain-derived neutrophic factor (BDNF) is a member of the neutrophin family that is known to activate the high-affinity tropomyosin-related receptor kinase B (TrkB). This study aimed to clarify the clinical and biological significance of the BDNF/TrkB pathway in gastric cancer. Methods: We analysed BDNF and TrkB expression in gastric cancer samples by real-time reverse transcription PCR and immunohistochemistry. To investigate the biological role of BDNF/TrkB axis, recombinant human BDNF (rhBDNF) and the Trk antagonist K252a were used for in vitro and in vivo analysis. Results: The BDNF expression at the invasive front of primary tumours was significantly elevated compared with that in the tumour core and adjacent normal mucosa. Increased BDNF expression at the invasive front was significantly correlated with factors reflecting disease progression, and poor prognosis. Increased co-expression of the BDNF/TrkB axis was significantly correlated with poor prognosis. Gastric cancer cells expressed BDNF, and administration of rhBDNF promoted proliferation, migration, invasion, and inhibition of anoikis. These effects were generally inhibited by K252a. In an in vivo assay, BDNF(+)/TrkB(+) gastric cancer cells injected into nude mice established peritoneal dissemination, whereas K252a inhibited tumour growth. Conclusion: The BDNF/TrkB pathway might be deeply involved in gastric cancer disease progression.
Collapse
|
29
|
Smad interacting protein 1 (SIP1) is associated with peritoneal carcinomatosis in intestinal type gastric cancer. Clin Exp Metastasis 2012; 30:417-29. [PMID: 23143680 DOI: 10.1007/s10585-012-9547-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 10/26/2012] [Indexed: 12/13/2022]
Abstract
Smad interacting protein 1 (SIP1) is an epithelial-mesenchymal transition (EMT)-inducible gene that plays a key role in tumor progression in various cancers. This study seeks to clarify the clinical and biological significance of SIP1 expression, especially in intestinal type gastric cancer. We analyzed the mRNA levels of SIP1 and other EMT regulators by real-time reverse transcription PCR in gastric tissue samples of 134 gastric cancer patients, and in five gastric cancer cell lines. SIP1 gene knockdown by siRNA transfection was performed to evaluate SIP1 function in gastric cancer cells. Expression of the SIP1 gene was significantly higher in cancerous tissue than in adjacent normal mucosa. Although the mRNA expression of the other EMT regulators tested (Snail, Slug, and Twist) was not correlated with clinicopathological factors, increased SIP1 expression was an independent prognostic factor and an independent risk factor for peritoneal dissemination. In addition, SIP1 expression was significantly positive and correlated with vimentin expression. For intestinal type gastric cancer in particular, elevated SIP1 expression was significantly correlated with peritoneal dissemination and poor prognosis (p < 0.05). In vitro, cell proliferation, migration, invasion, and resistance to anoikis were significantly inhibited in SIP1 siRNA-transfected MKN7 cells compared to control siRNA. SIP1 appears to play an important role in progression to peritoneal carcinomatosis and may be a therapeutic target for patients with intestinal type gastric cancer.
Collapse
|
30
|
Choi DS, Choi DY, Hong BS, Jang SC, Kim DK, Lee J, Kim YK, Kim KP, Gho YS. Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells. J Extracell Vesicles 2012; 1:18704. [PMID: 24009881 PMCID: PMC3760640 DOI: 10.3402/jev.v1i0.18704] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/15/2012] [Accepted: 08/15/2012] [Indexed: 12/18/2022] Open
Abstract
Cancer cells actively release extracellular vesicles (EVs), including exosomes and microvesicles, into surrounding tissues. These EVs play pleiotropic roles in cancer progression and metastasis, including invasion, angiogenesis, and immune modulation. However, the proteomic differences between primary and metastatic cancer cell-derived EVs remain unclear. Here, we conducted comparative proteomic analysis between EVs derived from human primary colorectal cancer cells (SW480) and their metastatic derivatives (SW620). Using label-free quantitation, we identified 803 and 787 proteins in SW480 EVs and SW620 EVs, respectively. Based on comparison between the estimated abundance of EV proteins, we identified 368 SW480 EV-enriched and 359 SW620 EV-enriched proteins. SW480 EV-enriched proteins played a role in cell adhesion, but SW620 EV-enriched proteins were associated with cancer progression and functioned as diagnostic indicators of metastatic cancer; they were overexpressed in metastatic colorectal cancer and played roles in multidrug resistance. As the first proteomic analysis comparing primary and metastatic cancer-derived EVs, this study increases our understanding of the pathological function of EVs in the metastatic process and provides useful biomarkers for cancer metastasis.
Collapse
Affiliation(s)
- Dong-Sic Choi
- Department of Life Science and Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhang G, Schetter A, He P, Funamizu N, Gaedcke J, Ghadimi BM, Ried T, Hassan R, Yfantis HG, Lee DH, Lacy C, Maitra A, Hanna N, Alexander HR, Hussain SP. DPEP1 inhibits tumor cell invasiveness, enhances chemosensitivity and predicts clinical outcome in pancreatic ductal adenocarcinoma. PLoS One 2012; 7:e31507. [PMID: 22363658 PMCID: PMC3282755 DOI: 10.1371/journal.pone.0031507] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 01/12/2012] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide. To identify biologically relevant genes with prognostic and therapeutic significance in PDAC, we first performed the microarray gene-expression profiling in 45 matching pairs of tumor and adjacent non-tumor tissues from resected PDAC cases. We identified 36 genes that were associated with patient outcome and also differentially expressed in tumors as compared with adjacent non-tumor tissues in microarray analysis. Further evaluation in an independent validation cohort (N = 27) confirmed that DPEP1 (dipeptidase 1) expression was decreased (T:N ratio ∼0.1, P<0.01) in tumors as compared with non-tumor tissues. DPEP1 gene expression was negatively correlated with histological grade (Spearman correlation coefficient = -0.35, P = 0.004). Lower expression of DPEP1 in tumors was associated with poor survival (Kaplan Meier log rank) in both test cohort (P = 0.035) and validation cohort (P = 0.016). DPEP1 expression was independently associated with cancer-specific mortality when adjusted for tumor stage and resection margin status in both univariate (hazard ratio = 0.43, 95%CI = 0.24-0.76, P = 0.004) and multivariate analyses (hazard ratio = 0.51, 95%CI = 0.27-0.94, P = 0.032). We further demonstrated that overexpression of DPEP1 suppressed tumor cells invasiveness and increased sensitivity to chemotherapeutic agent Gemcitabine. Our data also showed that growth factor EGF treatment decreased DPEP1 expression and MEK1/2 inhibitor AZD6244 increased DPEP1 expression in vitro, indicating a potential mechanism for DPEP1 gene regulation. Therefore, we provide evidence that DPEP1 plays a role in pancreatic cancer aggressiveness and predicts outcome in patients with resected PDAC. In view of these findings, we propose that DPEP1 may be a candidate target in PDAC for designing improved treatments.
Collapse
Affiliation(s)
- Geng Zhang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Aaron Schetter
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Peijun He
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Naotake Funamizu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Jochen Gaedcke
- Department of General and Visceral Surgery, University Medicine, Göttingen, Germany
| | - B. Michael Ghadimi
- Department of General and Visceral Surgery, University Medicine, Göttingen, Germany
| | - Thomas Ried
- Genetics Branch, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Raffit Hassan
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Harris G. Yfantis
- Pathology and Laboratory Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Dong H. Lee
- Pathology and Laboratory Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Curtis Lacy
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Anirban Maitra
- The Sol Goldman Pancreatic Cancer Research Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nader Hanna
- Division of Surgical Oncology, The Department of Surgery and the Marlene, School of Medicine, Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland, United States of America
| | - H. Richard Alexander
- Division of Surgical Oncology, The Department of Surgery and the Marlene, School of Medicine, Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland, United States of America
| | - S. Perwez Hussain
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| |
Collapse
|
32
|
Guebel DV, Schmitz U, Wolkenhauer O, Vera J. Analysis of cell adhesion during early stages of colon cancer based on an extended multi-valued logic approach. MOLECULAR BIOSYSTEMS 2012; 8:1230-42. [PMID: 22298312 DOI: 10.1039/c2mb05277f] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell adhesion in the normal colon is typically associated with differentiated cells, whereas in cancerous colon it is associated with advanced tumors. For advanced tumors growing evidence supports the existence of stem-like cells that have originated from transdifferentiation. Because stem cells can also be transformed in their own niche, at the base of the Lieberkühn's crypts, we conjectured that cell adhesion can also be critical in early tumorigenesis. To assess this hypothesis we built an annotated, multi-valued logic model addressing cell adhesion of normal and tumorigenic stem cells in the human colon. The model accounts for (i) events involving intercellular adhesion structures, (ii) interactions involving cytoskeleton-related structures, (iii) compartmental distribution of α/β/γ/δ-catenins, and (iv) variations in critical cell adhesion regulators (e.g., ILK, FAK, IQGAP, SNAIL, Caveolin). We developed a method that can deal with graded multiple inhibitions, something which is not possible with conventional logical approaches. The model comprises 315 species (including 26 genes), interconnected by 269 reactions. Simulations of the model covered six scenarios, which considered two types of colonic cells (stem vs. differentiated cells), under three conditions (normal, stressed and tumor). Each condition results from the combination of 92 inputs. We compared our multi-valued logic approach with the conventional Boolean approach for one specific example and validated the predictions against published data. Our analysis suggests that stem cells in their niche synthesize high levels of cytoplasmatic E-cadherin and CdhEP(Ser684,686,692), even under normal-mitogenic stimulus or tumorigenic conditions. Under these conditions, E-cadherin would be incorporated into the plasmatic membrane, but only as a non-adhesive CdhE_β-catenin_IQGAP complex. Under stress conditions, however, this complex could be displaced, yielding adhesive CdhE_β-catenin((cis/trans)) complexes. In the three scenarios tested with stem cells, desmosomes or tight junctions were not assembled. Other model predictions include expected levels of the nuclear complex β-catenin_TCF4 and the anti-apoptotic protein Survivin for both normal and tumorigenic colonic stem cells.
Collapse
Affiliation(s)
- Daniel V Guebel
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany.
| | | | | | | |
Collapse
|
33
|
Raimondo F, Morosi L, Chinello C, Perego R, Bianchi C, Albo G, Ferrero S, Rocco F, Magni F, Pitto M. Protein profiling of microdomains purified from renal cell carcinoma and normal kidney tissue samples. ACTA ACUST UNITED AC 2012; 8:1007-16. [DOI: 10.1039/c2mb05372a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Kawamura M, Toiyama Y, Tanaka K, Saigusa S, Okugawa Y, Hiro J, Uchida K, Mohri Y, Inoue Y, Kusunoki M. CXCL5, a promoter of cell proliferation, migration and invasion, is a novel serum prognostic marker in patients with colorectal cancer. Eur J Cancer 2011; 48:2244-51. [PMID: 22197219 DOI: 10.1016/j.ejca.2011.11.032] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 11/29/2011] [Accepted: 11/29/2011] [Indexed: 01/12/2023]
Abstract
PURPOSE Serum CXCL5 levels in patients with colorectal cancer (CRC) were assessed to evaluate correlation with clinicopathologic features and prognosis. The effects of CXCL5 on CRC cells were also investigated in vitro. METHODS Based on cytokine array analysis, CXCL5 was identified as a novel prognostic serum marker. Serum levels of CXCL5 were assessed in 250 CRC patients and 33 normal volunteers by enzyme-linked immunosorbent assay (ELISA), and their relation to clinicopathologic findings and survival investigated. CXCL5 levels in CRC cell lines were also measured by ELISA, and CXCL5 and CXCR2 expression was evaluated by immunohistochemistry. To investigate the biological role of the CXCL5/CXCR2 axis, recombinant human CXCL5 and CXCR2 neutralisation antibodies were used for proliferation, migration and invasion assays. RESULTS Preoperative serum CXCL5 was significantly elevated in patients with CRC compared with healthy volunteers (p=0.013). High serum CXCL5 was significantly associated with female sex (p=0.0098) and liver metastasis (p=0.0040). Univariate analysis correlated elevated CXCL5 with poor overall survival (p=0.0002). Multivariate analysis showed that elevated CXCL5 was a significant and independent prognostic factor of survival in all CRC patients (p=0.038). CRC cells secreted CXCL5, and administration of recombinant human CXCL5 promoted proliferation, migration and partial invasion. These effects were generally inhibited by CXCR2 neutralisation antibody. CONCLUSIONS Preoperative serum CXCL5 could serve as a novel predictive marker for prognosis determination of CRC patients. CXCL5/CXCR2 axis might be associated with colorectal cancer progression.
Collapse
Affiliation(s)
- Mikio Kawamura
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Matsushita K, Toiyama Y, Tanaka K, Saigusa S, Hiro J, Uchida K, Inoue Y, Kusunoki M. Soluble CXCL16 in preoperative serum is a novel prognostic marker and predicts recurrence of liver metastases in colorectal cancer patients. Ann Surg Oncol 2011; 19 Suppl 3:S518-27. [PMID: 21845497 DOI: 10.1245/s10434-011-1993-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Indexed: 12/14/2022]
Abstract
PURPOSE This study was designed to identify novel and reliable serum prognostic markers in patients with colorectal cancer (CRC). METHODS Based on cytokine array analysis, we identified soluble CXCL16 as a novel prognostic serum marker. Serum levels of CXCL16 were assessed in 314 CRC patients and 20 normal volunteers by enzyme-linked immunosorbent assay, and their relationships with clinicopathologic findings, including survival, were investigated. Proliferation, invasion, and wound healing assays were used to investigate the biological role of soluble CXCL16 in CRC cells, by exposure of HT-29 cells to recombinant CXCL16. RESULTS The median serum CXCL16 concentration in CRC patients was significantly higher than that in normal volunteers. In addition, serum CXCL16 levels increased significantly in accordance with the progression of UICC stage classification. Elevated serum CXCL16 level was significantly associated with poor survival and was an independent prognostic marker in CRC patients. Furthermore, in stage I-III CRC patients who underwent curative intent surgery, elevated serum CXCL16 levels were significantly associated with metachronous liver recurrence and poor survival. Recombinant soluble CXCL16 promoted the epithelial-mesenchymal transition (EMT) phenotype characterized by impaired E-cadherin production and induction of vimentin in vitro. In addition, recombinant soluble CXCL16 promoted cell growth, migration, and invasion in a CRC cell line. CONCLUSIONS In this study, we identified CXCL16 as a novel prognostic marker. Preoperative high serum levels of CXCL16 were associated with metachronous liver recurrence and poor prognosis in CRC patients. Soluble CXCL16 may play an important role in liver metastases through the induction of EMT.
Collapse
Affiliation(s)
- Kohei Matsushita
- Division of Reparative Medicine, Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Wiśniewski JR, Ostasiewicz P, Mann M. High recovery FASP applied to the proteomic analysis of microdissected formalin fixed paraffin embedded cancer tissues retrieves known colon cancer markers. J Proteome Res 2011; 10:3040-9. [PMID: 21526778 DOI: 10.1021/pr200019m] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteomic analysis of samples isolated by laser capture microdissection from clinical specimens requires sample preparation and fractionation methods suitable for small amounts of protein. Here we describe a streamlined filter-aided sample preparation (FASP) workflow that allows efficient analysis of lysates from low numbers of cells. Addition of carrier substances such as polyethylene glycol or dextran to the processed samples improves the peptide yields in the low to submicrogram range. In a single LC-MS/MS run, analyses of 500, 1000, and 3000 cells allowed identification of 905, 1536, and 2055 proteins, respectively. Incorporation of an additional SAX fractionation step at somewhat higher amounts enabled the analysis of formalin fixed and paraffin embedded human tissues prepared by LCM to a depth of 3600-4400 proteins per single experiment. We applied this workflow to compare archival neoplastic and matched normal colonic mucosa cancer specimens for three patients. Label-free quantification of more than 6000 proteins verified this technology through the differential expression of 30 known colon cancer markers. These included Carcino-Embryonic Antigen (CEA), the most widely used colon cancer marker, complement decay accelerating factor (DAF, CD55) and Metastasis-associated in colon cancer protein 1 (MACC1). Concordant with literature knowledge, mucin 1 was overexpressed and mucin 2 underexpressed in all three patients. These results show that FASP is suitable for the low level analysis of microdissected tissue and that it has the potential for exploration of clinical samples for biomarker and drug target discovery.
Collapse
Affiliation(s)
- Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, D-82152 Martinsried, Germany.
| | | | | |
Collapse
|