1
|
Solidoro R, Centonze A, Miciaccia M, Baldelli OM, Armenise D, Ferorelli S, Perrone MG, Scilimati A. Fluorescent imaging probes for in vivo ovarian cancer targeted detection and surgery. Med Res Rev 2024; 44:1800-1866. [PMID: 38367227 DOI: 10.1002/med.22027] [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: 07/05/2023] [Revised: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/19/2024]
Abstract
Ovarian cancer is the most lethal gynecological cancer, with a survival rate of approximately 40% at five years from the diagno. The first-line treatment consists of cytoreductive surgery combined with chemotherapy (platinum- and taxane-based drugs). To date, the main prognostic factor is related to the complete surgical resection of tumor lesions, including occult micrometastases. The presence of minimal residual diseases not detected by visual inspection and palpation during surgery significantly increases the risk of disease relapse. Intraoperative fluorescence imaging systems have the potential to improve surgical outcomes. Fluorescent tracers administered to the patient may support surgeons for better real-time visualization of tumor lesions during cytoreductive procedures. In the last decade, consistent with the discovery of an increasing number of ovarian cancer-specific targets, a wide range of fluorescent agents were identified to be employed for intraoperatively detecting ovarian cancer. Here, we present a collection of fluorescent probes designed and developed for fluorescence-guided ovarian cancer surgery. Original articles published between 2011 and November 2022 focusing on fluorescent probes, currently under preclinical and clinical investigation, were searched in PubMed. The keywords used were targeted detection, ovarian cancer, fluorescent probe, near-infrared fluorescence, fluorescence-guided surgery, and intraoperative imaging. All identified papers were English-language full-text papers, and probes were classified based on the location of the biological target: intracellular, membrane, and extracellular.
Collapse
Affiliation(s)
- Roberta Solidoro
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Antonella Centonze
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Olga Maria Baldelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Domenico Armenise
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | | | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| |
Collapse
|
2
|
Guo X, Tai H, Li X, Liu P, Liu J, Yu S. SPARC is a Novel Prognostic Biomarker for Ovarian Cancer and Associated with Immune Signatures and Drug Response. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5103061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Background: The calcium-binding matricellular glycoprotein (SPARC, secreted protein, acidic and rich in cysteine) belongs to the extracellular-matrix-protein family, and its functions mainly focus on tissue injury, remodeling, and tumorigenesis. The role of SPARC in ovarian cancer remains controversial at present. Methods: We searched SPARC using The Cancer Genome Atlas/Genotype-Tissue Expression (TCGA/GTEx) and other databases to analyze the relationship between its expression level and survival, immunity signatures, and chemical drug response, in ovarian cancer. Additionally, we overexpressed SPARC with plasmids in ovarian cancer SKOV3 and ID8 cell lines, then measured the effects of SPARC on the proliferation, migration, invasiveness, clonality, and stemness of ovarian cancer cells by Cell Counting Kit-8 (CCK8), Transwell, wound healing assay, adhesion assay, plate cloning assay, and soft agar spheroid formation in vitro. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed the potential signaling pathway for SPARC. Results: The higher expression of SPARC in ovarian cancer is related to more advanced tumor stage, poorer clinical survival, and worse chemical drug response, whereas it is positively correlated with immune signatures. For ovarian cancer phenotypes, higher SPARC expression level promotes cell proliferation, migration, colony formation, and spheroid formation. The GO and KEGG enrichment highlighted the potential molecular mechanisms for SPARC with PI3K-AKT and MAPK signaling regulation. Conclusions: SPARC promotes ovarian cancer progression through proliferation, migration, invasiveness, clonality, and stemness. A high level of expression of SPARC in ovarian cancer patients can be used as a marker of poor prognosis and poor drug response.
Collapse
Affiliation(s)
- Xiaorong Guo
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, Heilongjiang, China
| | - Huilin Tai
- Department of Mathematics and Statistics, Mcgill University, Montreal, QC H3A 0G4, Canada
| | - Xiaoqing Li
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, Heilongjiang, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, 150086 Harbin, Heilongjiang, China
| | - Jin Liu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, Heilongjiang, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Ji Q, Zhu H, Qin Y, Zhang R, Wang L, Zhang E, Zhou X, Meng R. GP60 and SPARC as albumin receptors: key targeted sites for the delivery of antitumor drugs. Front Pharmacol 2024; 15:1329636. [PMID: 38323081 PMCID: PMC10844528 DOI: 10.3389/fphar.2024.1329636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Albumin is derived from human or animal blood, and its ability to bind to a large number of endogenous or exogenous biomolecules makes it an ideal drug carrier. As a result, albumin-based drug delivery systems are increasingly being studied. With these in mind, detailed studies of the transport mechanism of albumin-based drug carriers are particularly important. As albumin receptors, glycoprotein 60 (GP60) and secreted protein acidic and rich in cysteine (SPARC) play a crucial role in the delivery of albumin-based drug carriers. GP60 is expressed on vascular endothelial cells and enables albumin to cross the vascular endothelial cell layer, and SPARC is overexpressed in many types of tumor cells, while it is minimally expressed in normal tissue cells. Thus, this review supplements existing articles by detailing the research history and specific biological functions of GP60 or SPARC and research advances in the delivery of antitumor drugs using albumin as a carrier. Meanwhile, the deficiencies and future perspectives in the study of the interaction of albumin with GP60 and SPARC are also pointed out.
Collapse
Affiliation(s)
- Qingzhi Ji
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Huimin Zhu
- Sheyang County Comprehensive Inspection and Testing Center, Yancheng, China
| | - Yuting Qin
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Ruiya Zhang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Lei Wang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Erhao Zhang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Run Meng
- Department of Immunology, Medical School, Nantong University, Nantong, China
| |
Collapse
|
4
|
Rasool S, Ismaeel QAL, Arif SH. CYR61 promotes colorectal carcinoma progression via activating epithelial-mesenchymal transition. Am J Cancer Res 2023; 13:4872-4887. [PMID: 37970355 PMCID: PMC10636662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/26/2023] [Indexed: 11/17/2023] Open
Abstract
Colorectal carcinoma is the third most common type of cancer. Although the role of matricellular proteins and their association with tumor progression is well documented, limited data are available concerning their involvement in colorectal cancer. The current study investigated the expression pattern of matricellular proteins SPARC and CYR61 with epithelial-mesenchymal transition proteins in human CRC tissues and unleashed their association with colorectal cancer progression. The expression of these proteins was associated with advancement in tumor staging, nodal metastasis, and vascular invasion. Elevated CYR61 protein levels were also consistent with higher mesenchymal markers ZEB1 and Vimentin in collected biopsies and CRC cells. Moreover, expression of CYR61 promoted CRC cell migration, invasion, proliferation, and apoptosis. Our findings conclusively revealed the significant involvement of CYR61 in CRC progression through activating epithelial-mesenchymal transition. This discovery holds great promise for advancing therapeutic approaches in the treatment of CRC.
Collapse
Affiliation(s)
- Shelan Rasool
- Department of Anatomy, Biology and Histology, College of Medicine, University of DuhokDuhok 42001, Kurdistan Region of Iraq
| | - Qais AL Ismaeel
- Department of Anatomy, Biology and Histology, College of Medicine, University of DuhokDuhok 42001, Kurdistan Region of Iraq
| | - Sardar H Arif
- Department of Surgery, College of Medicine, University of DuhokDuhok 42001, Kurdistan Region of Iraq
| |
Collapse
|
5
|
Jiang S, Sun HF, Li S, Zhang N, Chen JS, Liu JX. SPARC: a potential target for functional nanomaterials and drugs. Front Mol Biosci 2023; 10:1235428. [PMID: 37577749 PMCID: PMC10419254 DOI: 10.3389/fmolb.2023.1235428] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), also termed osteonectin or BM-40, is a matricellular protein which regulates cell adhesion, extracellular matrix production, growth factor activity, and cell cycle. Although SPARC does not perform a structural function, it, however, modulates interactions between cells and the surrounding extracellular matrix due to its anti-proliferative and anti-adhesion properties. The overexpression of SPARC at sites, including injury, regeneration, obesity, cancer, and inflammation, reveals its application as a prospective target and therapeutic indicator in the treatment and assessment of disease. This article comprehensively summarizes the mechanism of SPARC overexpression in inflammation and tumors as well as the latest research progress of functional nanomaterials in the therapy of rheumatoid arthritis and tumors by manipulating SPARC as a new target. This article provides ideas for using functional nanomaterials to treat inflammatory diseases through the SPARC target. The purpose of this article is to provide a reference for ongoing disease research based on SPARC-targeted therapy.
Collapse
Affiliation(s)
- Shan Jiang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Hui-Feng Sun
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Shuang Li
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- College Pharmacy, Jiamusi University, Jiamusi, China
| | - Ning Zhang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Ji-Song Chen
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- School of Pharmaceutical Sciences, University of South China, Hengyang, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
6
|
Yang X, Tohda C. Diosgenin restores memory function via SPARC-driven axonal growth from the hippocampus to the PFC in Alzheimer's disease model mice. Mol Psychiatry 2023; 28:2398-2411. [PMID: 37085711 PMCID: PMC10611574 DOI: 10.1038/s41380-023-02052-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Central nervous system axons have minimal capacity to regenerate in adult brains, hindering memory recovery in Alzheimer's disease (AD). Although recent studies have shown that damaged axons sprouted in adult and AD mouse brains, long-distance axonal re-innervation to their targets has not been achieved. We selectively visualized axon-growing neurons in the neural circuit for memory formation, from the hippocampus to the prefrontal cortex, and showed that damaged axons successfully extended to their native projecting area in mouse models of AD (5XFAD) by administration of an axonal regenerative agent, diosgenin. In vivo transcriptome analysis detected the expression profile of axon-growing neurons directly isolated from the hippocampus of 5XFAD mice. Secreted protein acidic and rich in cysteine (SPARC) was the most expressed gene in axon-growing neurons. Neuron-specific overexpression of SPARC via adeno-associated virus serotype 9 delivery in the hippocampus recovered memory deficits and axonal projection to the prefrontal cortex in 5XFAD mice. DREADDs (Designer receptors exclusively activated by designer drugs) analyses revealed that SPARC overexpression-induced axonal growth in the 5XFAD mouse brain directly contributes to memory recovery. Elevated levels of SPARC on axonal membranes interact with extracellular rail-like collagen type I to promote axonal remodeling along their original tracings in primary cultured hippocampal neurons. These findings suggest that SPARC-driven axonal growth in the brain may be a promising therapeutic strategy for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
7
|
Tedja R, Alvero AB, Fox A, Cardenas C, Pitruzzello M, Chehade H, Bawa T, Adzibolosu N, Gogoi R, Mor G. Generation of Stable Epithelial-Mesenchymal Hybrid Cancer Cells with Tumorigenic Potential. Cancers (Basel) 2023; 15:cancers15030684. [PMID: 36765641 PMCID: PMC9913490 DOI: 10.3390/cancers15030684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Cancer progression, invasiveness, and metastatic potential have been associated with the activation of the cellular development program known as epithelial-to-mesenchymal transition (EMT). This process is known to yield not only mesenchymal cells, but instead an array of cells with different degrees of epithelial and mesenchymal phenotypes with high plasticity, usually referred to as E/M hybrid cells. The characteristics of E/M hybrid cells, their importance in tumor progression, and the key regulators in the tumor microenvironment that support this phenotype are still poorly understood. METHODS In this study, we established an in vitro model of EMT and characterized the different stages of differentiation, allowing us to identify the main genomic signature associated with the E/M hybrid state. RESULTS We report that once the cells enter the E/M hybrid state, they acquire stable anoikis resistance, invasive capacity, and tumorigenic potential. We identified the hepatocyte growth factor (HGF)/c-MET pathway as a major driver that pushes cells in the E/M hybrid state. CONCLUSIONS Herein, we provide a detailed characterization of the signaling pathway(s) promoting and the genes associated with the E/M hybrid state.
Collapse
Affiliation(s)
- Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Correspondence: (R.T.); (G.M.)
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Carlos Cardenas
- Department of Obstetrics and Gynecology, Family HealthCare Network, Porterville, CA 93257, USA
| | - Mary Pitruzzello
- Department of Dermatology, Yale Medical School, New Haven, CT 06510, USA
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Tejeshwhar Bawa
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Nicholas Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Gil Mor
- Department of Obstetrics and Gynecology, Family HealthCare Network, Porterville, CA 93257, USA
- Correspondence: (R.T.); (G.M.)
| |
Collapse
|
8
|
Zhou XD, Qu YW, Wang L, Jia FH, Chen P, Wang YP, Liu HF. Identification of potential hub genes of gastric cancer. Medicine (Baltimore) 2022; 101:e30741. [PMID: 36254003 PMCID: PMC9575828 DOI: 10.1097/md.0000000000030741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a malignant tumor originated from gastric mucosa epithelium. It is the third leading cause of cancer mortality in China. The early symptoms are not obvious. When it is discovered, it has developed to the advanced stage, and the prognosis is poor. In order to screen for potential genes for GC development, this study obtained GSE118916 and GSE109476 from the gene expression omnibus (GEO) database for bioinformatics analysis. METHODS First, GEO2R was used to identify differentially expressed genes (DEG) and the functional annotation of DEGs was performed by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The Search Tool for the Retrieval of Interacting Genes (STRING) tool was used to construct protein-protein interaction (PPI) network and the most important modules and hub genes were mined. Real time quantitative polymerase chain reaction assay was performed to verify the expression level of hub genes. RESULTS A total of 139 DEGs were identified. The functional changes of DEGs are mainly concentrated in the cytoskeleton, extracellular matrix and collagen synthesis. Eleven genes were identified as core genes. Bioinformatics analysis shows that the core genes are mainly enriched in many processes related to cell adhesion and collagen. CONCLUSION In summary, the DEGs and hub genes found in this study may be potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Xu-Dong Zhou
- The Clinical College of the General Hospital of Chinese People's Armed Police Forces, Anhui Medical University, Hefei, P.R. China
| | - Ya-Wei Qu
- Department of Gastroenterology, Third Medical Center of PLA General Hospital, Beijing, P.R. China
| | - Li Wang
- Department of Gastroenterology, Huamei Hospital of China National University of Science and Technology, Ningbo, P.R. China
| | - Fu-Hua Jia
- Department of Gastroenterology, Huamei Hospital of China National University of Science and Technology, Ningbo, P.R. China
| | - Peng Chen
- Department of Ultrasound, Graduate School of Jinzhou Medical University, Jinzhou, P.R. China
| | - Yin-Pu Wang
- Department of Gastroenterology, Baoji Hospital Affiliated to Xi’an Jiaotong University, Baoji, P.R. China
| | - Hai-Feng Liu
- The Clinical College of the General Hospital of Chinese People's Armed Police Forces, Anhui Medical University, Hefei, P.R. China
- *Correspondence: Hai-Feng Liu, The Clinical College of the General Hospital of Chinese People's Armed Police Forces, Anhui Medical University, Hefei 230032, P.R. China (e-mail: )
| |
Collapse
|
9
|
MISIRLI D, BİNGÖL ÖZAKPINAR Ö, ŞEKERLER T, ARU B, YANIKKAYA DEMİREL G, TUNOĞLU S, OZSAVCİ D. Effects of SPARC and Possible Receptors on Colon Cancer Cell Line. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2022. [DOI: 10.33808/clinexphealthsci.1100770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Objective: The aim of this study was to observe the apoptotic/cytotoxic effects of exogenous SPARC on colon cancer cell line HT-29, then to investigate the function of stabilin-1 and integrin αvβ3, which are possible receptors for SPARC in colon cancer cells and to determine the quantitation of their receptor numbers.
Methods: Appropriate doses of exogenous SPARC and it’s inhibitor, cilengitide added to HT-29 cell line were determined by xCELLigence Real-Time Cell Analysis system, SPARC-mediated caspase 3 expressions were measured. Using the RT-PCR system, gene expression levels of SPARC, stabilin-1 and integrin αvβ3 receptors (silenced/nonsilenced with cilengitide) were detected then the numbers of receptors per cell were quantitated by flow cytometry.
Results: IC50 value of SPARC was determined as 4.57 μg/mL and IC50 value of cilengitide was determined as 50 nM. 5 μg/mL exogenous SPARC caused increased apoptosis in the HT-29 line. Significant increase in gene expression of integrin αvβ3 receptor was observed in the group incubated with 5 μg/mL SPARC, contrarily, the addition of cilengitide decreased gene expressions. The integrin αvβ3 receptor numbers
increased approximately 2-fold with SPARC compared to the control. No significant changes were observed in the gene expression and receptor numbers of stabilin-1.
Conclusion: Exogenous SPARC was shown to reduce proliferation and induce apoptosis in colon cancer cells. Integrin αvβ3 is thought to be the possible receptor mediating SPARC in colon cancer cells. Quantification of surface receptors per cell, which we think we have done first, can be considered as a marker in the follow-up of anticancer treatments.
Collapse
|
10
|
Susceptibility to disease (tropical theileriosis) is associated with differential expression of host genes that possess motifs recognised by a pathogen DNA binding protein. PLoS One 2022; 17:e0262051. [PMID: 35061738 PMCID: PMC8782480 DOI: 10.1371/journal.pone.0262051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/15/2021] [Indexed: 12/04/2022] Open
Abstract
Background Knowledge of factors that influence the outcome of infection are crucial for determining the risk of severe disease and requires the characterisation of pathogen-host interactions that have evolved to confer variable susceptibility to infection. Cattle infected by Theileria annulata show a wide range in disease severity. Native (Bos indicus) Sahiwal cattle are tolerant to infection, whereas exotic (Bos taurus) Holstein cattle are susceptible to acute disease. Methodology/Principal findings We used RNA-seq to assess whether Theileria infected cell lines from Sahiwal cattle display a different transcriptome profile compared to Holstein and screened for altered expression of parasite factors that could generate differences in host cell gene expression. Significant differences (<0.1 FDR) in the expression level of a large number (2211) of bovine genes were identified, with enrichment of genes associated with Type I IFN, cholesterol biosynthesis, oncogenesis and parasite infection. A screen for parasite factors found limited evidence for differential expression. However, the number and location of DNA motifs bound by the TashAT2 factor (TA20095) were found to differ between the genomes of B. indicus vs. B. taurus, and divergent motif patterns were identified in infection-associated genes differentially expressed between Sahiwal and Holstein infected cells. Conclusions/Significance We conclude that divergent pathogen-host molecular interactions that influence chromatin architecture of the infected cell are a major determinant in the generation of gene expression differences linked to disease susceptibility.
Collapse
|
11
|
Ye M, Lin Y, Pan S, Wang ZW, Zhu X. Applications of Multi-omics Approaches for Exploring the Molecular Mechanism of Ovarian Carcinogenesis. Front Oncol 2021; 11:745808. [PMID: 34631583 PMCID: PMC8497990 DOI: 10.3389/fonc.2021.745808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer ranks as the fifth most common cause of cancer-related death in females. The molecular mechanisms of ovarian carcinogenesis need to be explored in order to identify effective clinical therapies for ovarian cancer. Recently, multi-omics approaches have been applied to determine the mechanisms of ovarian oncogenesis at genomics (DNA), transcriptomics (RNA), proteomics (proteins), and metabolomics (metabolites) levels. Multi-omics approaches can identify some diagnostic and prognostic biomarkers and therapeutic targets for ovarian cancer, and these molecular signatures are beneficial for clarifying the development and progression of ovarian cancer. Moreover, the discovery of molecular signatures and targeted therapy strategies could noticeably improve the prognosis of ovarian cancer patients.
Collapse
Affiliation(s)
| | | | | | - Zhi-wei Wang
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Teuwen LA, De Rooij LPMH, Cuypers A, Rohlenova K, Dumas SJ, García-Caballero M, Meta E, Amersfoort J, Taverna F, Becker LM, Veiga N, Cantelmo AR, Geldhof V, Conchinha NV, Kalucka J, Treps L, Conradi LC, Khan S, Karakach TK, Soenen S, Vinckier S, Schoonjans L, Eelen G, Van Laere S, Dewerchin M, Dirix L, Mazzone M, Luo Y, Vermeulen P, Carmeliet P. Tumor vessel co-option probed by single-cell analysis. Cell Rep 2021; 35:109253. [PMID: 34133923 DOI: 10.1016/j.celrep.2021.109253] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor vessel co-option is poorly understood, yet it is a resistance mechanism against anti-angiogenic therapy (AAT). The heterogeneity of co-opted endothelial cells (ECs) and pericytes, co-opting cancer and myeloid cells in tumors growing via vessel co-option, has not been investigated at the single-cell level. Here, we use a murine AAT-resistant lung tumor model, in which VEGF-targeting induces vessel co-option for continued growth. Single-cell RNA sequencing (scRNA-seq) of 31,964 cells reveals, unexpectedly, a largely similar transcriptome of co-opted tumor ECs (TECs) and pericytes as their healthy counterparts. Notably, we identify cell types that might contribute to vessel co-option, i.e., an invasive cancer-cell subtype, possibly assisted by a matrix-remodeling macrophage population, and another M1-like macrophage subtype, possibly involved in keeping or rendering vascular cells quiescent.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Laura P M H De Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anne Cuypers
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Katerina Rohlenova
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Elda Meta
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Jacob Amersfoort
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Federico Taverna
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nuphar Veiga
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Vincent Geldhof
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nadine V Conchinha
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Shawez Khan
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Tobias K Karakach
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, CCB, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, Qingdao 266555, P.R. China; BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, BGI-Shenzhen, Shenzhen 518120, P.R. China.
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark.
| |
Collapse
|
13
|
Lu X, Ji C, Jiang L, Zhu Y, Zhou Y, Meng J, Gao J, Lu T, Ye J, Yan F. Tumour microenvironment-based molecular profiling reveals ideal candidates for high-grade serous ovarian cancer immunotherapy. Cell Prolif 2021; 54:e12979. [PMID: 33522069 PMCID: PMC7941229 DOI: 10.1111/cpr.12979] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/08/2020] [Accepted: 12/21/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Due to limited immunological profiles of high-grade serous ovarian cancer (HGSOC), we aimed to characterize its molecular features to determine whether a specific subset that can respond to immunotherapy exists. MATERIALS AND METHODS A training cohort of 418 HGSOC samples from TCGA was analysed by consensus non-negative matrix factorization. We correlated the expression patterns with the presence of immune cell infiltrates, immune regulatory molecules and other genomic or epigenetic features. Two independent cohorts containing 482 HGSOCs and in vitro experiments were used for validation. RESULTS We identified immune and non-immune groups where the former was enriched in signatures that reflect immune cells, infiltration and PD-1 signalling (all, P < 0.001), and presented with a lower chromosomal aberrations but increased neoantigens, tumour mutation burden, and microsatellite instability (all, P < 0.05); this group was further refined into two microenvironment-based subtypes characterized by either immunoactivation or carcinoma-associated fibroblasts (CAFs) and distinct prognosis. CAFs-immune subtype was enriched for factors that mediate immunosuppression and promote tumour progression, including highly expressed stromal signature, TGF-β signalling, epithelial-mesenchymal transition and tumour-associated M2-polarized macrophages (all, P < 0.001). Robustness of these immune-specific subtypes was verified in validation cohorts, and in vitro experiments indicated that activated-immune subtype may benefit from anti-PD1 antibody therapy (P < 0.05). CONCLUSION Our findings revealed two immune subtypes with different responses to immunotherapy and indicated that some HGSOCs may be susceptible to immunotherapies or combination therapies.
Collapse
Affiliation(s)
- Xiaofan Lu
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Caoyu Ji
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liyun Jiang
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Texas, USA
| | - Yue Zhu
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yujie Zhou
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,Department of Urology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jun Gao
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tao Lu
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Junmei Ye
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
15
|
Wang L, Wang W, Xu Y, Wang Q. Low Levels of SPARC are Associated with Tumor Progression and Poor Prognosis in Human Endometrial Carcinoma. Onco Targets Ther 2020; 13:11549-11569. [PMID: 33204109 PMCID: PMC7667597 DOI: 10.2147/ott.s277795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background SPARC (secreted protein acidic and rich in cysteine), also known as osteonectin, BM-40, and 43 K protein, is a matricellular protein associated with various tumor progressions. The aim of this research was to investigate the prognostic value of SPARC in endometrial carcinoma (EC) and its function in cancer cell invasion and metastasis. Methods From both mRNA and protein levels, SPARC expression in normal endometrial tissue and EC tissue, normal endometrial cells and 4 EC cell lines (KLE, HEC-1A, HEC-1B, Ishikawa) were evaluated by immunohistochemistry (IHC) or immunocytochemistry (ICC), quantitative real-time PCR (qRT-PCR) and Western blotting. RNA interference mediated by lentivirus was performed to get the stable SPARC down-expressing cells. The functional analysis techniques in vitro and in vivo were used to detect the effects of SPARC knockdown on EC cell proliferation, apoptosis, invasion and metastasis. Results The expressions of SPARC in EC tissues and cells were much lower than those in normal endometrial cells and tissues; meanwhile, its low expression was closely related to the malignant clinicopathological characteristics of EC. SPARC knockdown could inhibit apoptosis, promote the process of EMT and improve the proliferation and invasion capacities of EC cells in vitro and in vivo. Conclusion The low expression of SPARC was detected in EC tissues and cells, which was positively correlated with the poor prognosis of EC patients. SPARC acted as a tumor suppressor gene that hindered EC progression, which proposed a new therapeutic strategy for EC treatment.
Collapse
Affiliation(s)
- Ling Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Wei Wang
- Department of Radiology, The First Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Yangchun Xu
- Department of Dermatology, The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Qiang Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| |
Collapse
|
16
|
Dasari S, Pandhiri T, Grassi T, Visscher DW, Multinu F, Agarwal K, Mariani A, Shridhar V, Mitra AK. Signals from the Metastatic Niche Regulate Early and Advanced Ovarian Cancer Metastasis through miR-4454 Downregulation. Mol Cancer Res 2020; 18:1202-1217. [PMID: 32350057 PMCID: PMC10788085 DOI: 10.1158/1541-7786.mcr-19-1162] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/16/2020] [Accepted: 04/24/2020] [Indexed: 11/16/2022]
Abstract
Treatment of ovarian cancer is limited by extensive metastasis and yet it remains poorly understood. We have studied the critical step of metastatic colonization in the context of the productive interactions with the metastatic microenvironment with a goal of identifying key regulators. By combining miRNA expression analysis using an organotypic 3D culture model of early ovarian cancer metastasis with that of matched primary and metastatic tumors from 42 patients with ovarian cancer, we identified miR-4454 as a key regulator of both early colonization and advanced metastasis in patients with ovarian cancer. miR-4454 was downregulated in the metastasizing ovarian cancer cells through paracrine signals from microenvironmental fibroblasts, which promoted migration, invasion, proliferation, and clonogenic growth in ovarian cancer cells as well as their ability to penetrate through the outer layers of the omentum. Stable overexpression of miR-4454 decreased metastasis in ovarian cancer xenografts. Its mechanism of action was through the upregulation of its targets, secreted protein acidic and cysteine rich (SPARC) and BCL2 associated athanogene 5 (BAG5), which activated focal adhesion kinase (FAK) signaling, promoted mutant p53 gain of function by its stabilization, and inhibited apoptosis. Because microenvironment-induced downregulation of miR-4454 is essential for early and advanced metastasis, targeting it could be a promising therapeutic approach. IMPLICATIONS: This study identifies a miRNA, miR-4454, which is downregulated by signals from the microenvironment and promotes early and advanced ovarian cancer metastasis through its effects on FAK activation, mutant p53 stabilization, and apoptosis inhibition.
Collapse
Affiliation(s)
- Subramanyam Dasari
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
| | - Taruni Pandhiri
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
| | - Tommaso Grassi
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Francesco Multinu
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Komal Agarwal
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
- Department of Obstetrics and Gynecology, St. Vincent Dunn Hospital, Bedford, Indiana
| | - Andrea Mariani
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Viji Shridhar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Anirban K Mitra
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| |
Collapse
|
17
|
Chignon A, Bon-Baret V, Boulanger MC, Li Z, Argaud D, Bossé Y, Thériault S, Arsenault BJ, Mathieu P. Single-cell expression and Mendelian randomization analyses identify blood genes associated with lifespan and chronic diseases. Commun Biol 2020; 3:206. [PMID: 32358504 PMCID: PMC7195437 DOI: 10.1038/s42003-020-0937-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/10/2020] [Indexed: 12/13/2022] Open
Abstract
The human lifespan is a heritable trait, which is intricately linked to the development of disorders. Here, we show that genetic associations for the parental lifespan are enriched in open chromatin of blood cells. By using blood expression quantitative trait loci (eQTL) derived from 31,684 samples, we identified for the lifespan 125 cis- and 559 trans-regulated expressed genes (eGenes) enriched in adaptive and innate responses. Analysis of blood single-cell expression data showed that eGenes were enriched in dendritic cells (DCs) and the modelling of cell ligand-receptor interactions predicted crosstalk between DCs and a cluster of monocytes with a signature of cytotoxicity. In two-sample Mendelian randomization (MR), we identified 16 blood cis-eGenes causally associated with the lifespan. In MR, the majority of cis-eGene-disorder association pairs had concordant effects with the lifespan. The present work underlined that the lifespan is linked with the immune response and identifies eGenes associated with the lifespan and disorders.
Collapse
Affiliation(s)
- Arnaud Chignon
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada
| | - Valentin Bon-Baret
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada
| | - Marie-Chloé Boulanger
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada
| | - Zhonglin Li
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada
| | - Deborah Argaud
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec, QC, Canada
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec, QC, Canada
| | | | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Quebec Heart and Lung Institute/Research Center, Laval University, Quebec, QC, Canada.
| |
Collapse
|
18
|
Cao Y, Wang X, Tang L, Li Y, Song X, Liu X, Li M, Chen F, Wan H. Engrailed-2 promotes a malignant phenotype of esophageal squamous cell carcinoma through upregulating the expression of pro-oncogenic genes. PeerJ 2020; 8:e8662. [PMID: 32117645 PMCID: PMC7036277 DOI: 10.7717/peerj.8662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/29/2020] [Indexed: 01/09/2023] Open
Abstract
Background A number of homeobox genes have been implicated in the development of various cancers. However, the role of engrailed 2 (EN2), a member of the homeobox gene superfamily, in esophageal squamous cell carcinoma (ESCC) remains unknown. Methods The expression of EN2 was examined using quantitative real-time PCR and immunohistochemistry. A stable cell line was established to express exogenous EN2 using a lentivirus system. The malignant phenotype was analyzed with proliferation, clonogenicity, wound-healing and invasion assays. The CRISPR/Cas9 system was adopted to deplete endogenous EN2. RNA profiling was performed using gene expression microarray. The ShRNA-mediated method was used to knock down the expression of SPARC. The structure-function relationship was determined using site-directed mutagenesis. Results EN2 is highly expressed in ESCC. The malignant phenotype of the ESCC cell line was amplified by an overexpression of EN2 but was attenuated by a disruption of EN2. RNA profiling analysis revealed that distinct sets of genes were modulated by the expression of EN2 in various ESCC cell lines and oncogenes were among these. EN2 greatly increased the expression of SPARC in Eca109. Site-directed mutagenesis revealed that the induction of SPARC was closely correlated with the protumor function of EN2. ShRNA-mediated knockdown of SPARC attenuated the malignant phenotype of EN2-infected cells. These data suggest that SPARC is crucial for mediating the protumor function of EN2. Discussion EN2 has an oncogenic function in ESCC that is mediated by upregulating the expression of pro-oncogenic genes downstream. EN2 may potentially act as a diagnostic marker or therapeutic target for ESCC treatment in the future.
Collapse
Affiliation(s)
- Yong Cao
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoyan Wang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Li Tang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yan Li
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xueqin Song
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Liu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mingying Li
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Feng Chen
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haisu Wan
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
19
|
Yang Y, Li G. Retracted: Icariin inhibits proliferation, migration, and invasion of medulloblastoma DAOY cells by regulation of SPARC. Phytother Res 2020; 34:591-600. [PMID: 32011040 DOI: 10.1002/ptr.6545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/12/2019] [Accepted: 10/22/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Yan Yang
- Department of NeurosurgeryJining No.1 People's Hospital Jining China
| | - Guifang Li
- Department of Occupational MedicineWeifang People's Hospital Weifang China
| |
Collapse
|
20
|
Secreted protein acidic and rich in cysteine and cancer: A homeostatic hormone? Cytokine 2020; 127:154996. [PMID: 31955132 DOI: 10.1016/j.cyto.2020.154996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
|
21
|
Differential DNA methylation in high-grade serous ovarian cancer (HGSOC) is associated with tumor behavior. Sci Rep 2019; 9:17996. [PMID: 31784612 PMCID: PMC6884482 DOI: 10.1038/s41598-019-54401-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022] Open
Abstract
The epigenome offers an additional facet of cancer that can help categorize patients into those at risk of disease, recurrence, or treatment failure. We conducted a retrospective, nested, case-control study of advanced and recurrent high-grade serous ovarian cancer (HGSOC) patients in which we assessed epigenome-wide association using Illumina methylationEPIC arrays to characterize DNA methylation status and RNAseq to evaluate gene expression. Comparing HGSOC tumors with normal fallopian tube tissues we observe global hypomethylation but with skewing towards hypermethylation when interrogating gene promoters. In total, 5,852 gene interrogating probes revealed significantly different methylation. Within HGSOC, 57 probes highlighting 17 genes displayed significant differential DNA methylation between primary and recurrent disease. Between optimal vs suboptimal surgical outcomes 99 probes displayed significantly different methylation but only 29 genes showed an inverse correlation between methylation status and gene expression. Overall, differentially methylated genes point to several pathways including RAS as well as hippo signaling in normal vs primary HGSOC; valine, leucine, and isoleucine degradation and endocytosis in primary vs recurrent HGSOC; and pathways containing immune driver genes in optimal vs suboptimal surgical outcomes. Thus, differential DNA methylation identified numerous genes that could serve as potential biomarkers and/or therapeutic targets in HGSOC.
Collapse
|
22
|
Ceppi L, Bardhan NM, Na Y, Siegel A, Rajan N, Fruscio R, Del Carmen MG, Belcher AM, Birrer MJ. Real-Time Single-Walled Carbon Nanotube-Based Fluorescence Imaging Improves Survival after Debulking Surgery in an Ovarian Cancer Model. ACS NANO 2019; 13:5356-5365. [PMID: 31009198 DOI: 10.1021/acsnano.8b09829] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Improved cytoreductive surgery for advanced stage ovarian cancer (OC) represents a critical challenge in the treatment of the disease. Optimal debulking reaching no evidence of macroscopic disease is the primary surgical end point with a demonstrated survival advantage. Targeted molecule-based fluorescence imaging offers complete tumor resection down to the microscopic scale. We used a custom-built reflectance/fluorescence imaging system with an orthotopic OC mouse model to both quantify tumor detectability and evaluate the effect of fluorescence image-guided surgery on post-operative survival. The contrast agent is an intraperitoneal injectable nanomolecular probe, composed of single-walled carbon nanotubes, coupled to an M13 bacteriophage carrying a modified peptide binding to the SPARC protein, an extracellular protein overexpressed in OC. The imaging system is capable of detecting a second near-infrared window fluorescence (1000-1700 nm) and can display real-time video imagery to guide intraoperative tumor debulking. We observed high microscopic tumor detection with a pixel-limited resolution of 200 μm. Moreover, in a survival-surgery orthotopic OC mouse model, we demonstrated an increased survival benefit for animals treated with fluorescence image-guided surgical resection compared to standard surgery.
Collapse
Affiliation(s)
- Lorenzo Ceppi
- Center for Cancer Research, The Gillette Center for Gynecologic Oncology , Massachusetts General Hospital, Harvard Medical School , Boston , Massachusetts 02114 , United States
- Department of Medicine and Surgery , University of Milan-Bicocca , 20126 Milan , Italy
| | - Neelkanth M Bardhan
- Department of Materials Science and Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
- The David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , Massachusetts 02142 , United States
- Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - YoungJeong Na
- Center for Cancer Research, The Gillette Center for Gynecologic Oncology , Massachusetts General Hospital, Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Andrew Siegel
- Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Nandini Rajan
- Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Robert Fruscio
- Department of Medicine and Surgery , University of Milan-Bicocca , 20126 Milan , Italy
| | - Marcela G Del Carmen
- Division of Gynecologic Oncology, Vincent Obstetrics and Gynecology , Massachusetts General Hospital, Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Angela M Belcher
- Department of Materials Science and Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
- The David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , Massachusetts 02142 , United States
- Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Michael J Birrer
- Center for Cancer Research, The Gillette Center for Gynecologic Oncology , Massachusetts General Hospital, Harvard Medical School , Boston , Massachusetts 02114 , United States
- O'Neal Comprehensive Cancer Center, Division of Hematology-Oncology , University of Alabama at Birmingham , Birmingham , Alabama 35294 , United States
| |
Collapse
|
23
|
Tang L, Cao Y, Song X, Wang X, Li Y, Yu M, Li M, Liu X, Huang F, Chen F, Wan H. HOXC6 promotes migration, invasion and proliferation of esophageal squamous cell carcinoma cells via modulating expression of genes involved in malignant phenotypes. PeerJ 2019; 7:e6607. [PMID: 30886783 PMCID: PMC6421064 DOI: 10.7717/peerj.6607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/10/2019] [Indexed: 12/29/2022] Open
Abstract
Background HOXC6 is a member of the HOX gene family. The elevated expression of this gene occurs in prostate and breast cancers. However, the role of HOXC6 in esophageal squamous cell carcinoma (ESCC) remains largely uninvestigated. Methods The expression of HOXC6 was examined by immunohistochemistry, quantitative real-time PCR and immunoblotting assays. The lentivirus-mediated expression of HOXC6 was verified at mRNA and protein levels. Wound healing and Matrigel assays were performed to assess the effect of HOXC6 on the migration and invasion of cancer cells. The growth curving, CCK8, and colony formation assays were utilized to access the proliferation capacities. RNA-seq was performed to evaluate the downstream targets of HOXC6. Bioinformatic tool was used to analyze the gene expression. Results HOXC6 was highly expressed in ESCC tissues. HOXC6 overexpression promoted the migration, invasion, and proliferation of both Eca109 and TE10 cells. There were 2,155 up-regulated and 759 down-regulated genes in Eca109-HOXC6 cells and 95 up-regulated and 47 down-regulated genes in TE10-HOXC6 cells compared with the results of control. Interestingly, there were only 20 common genes, including 17 up-regulated and three down-regulated genes with similar changes upon HOXC6 transfection in both cell lines. HOXC6 activated several crucial genes implicated in the malignant phenotype of cancer cells. Discussion HOXC6 is highly expressed in ESCC and promotes malignant phenotype of ESCC cells. HOXC6 can be used as a new therapeutic target of ESCC.
Collapse
Affiliation(s)
- Li Tang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yong Cao
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xueqin Song
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoyan Wang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Li
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Minglan Yu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mingying Li
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xu Liu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fang Huang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Feng Chen
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Haisu Wan
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Non-catalytic region of tyrosine kinase adaptor protein 2 (NCK2) pathways as factor promoting aggressiveness in ovarian cancer. Int J Biol Markers 2018; 33:124-131. [PMID: 29218693 DOI: 10.5301/ijbm.5000264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND In this study we investigated the function of the non-catalytic region of tyrosine kinase adaptor protein 2 (NCK2) and its correlation with ITGB1 and ITGB4 integrins in driving ovarian cancer (OvCa) aggressiveness. We also evaluated whether NCK2 may influence prognosis in OvCa patients. METHODS Nanofluidic technology was used to analyze expression of NCK2 in 332 OvCa patients. To evaluate mRNA expression of NCK2, integrins and VEGFA in OvCa cell lines, qRT-PCR was performed. Stable NCK2 overexpression was obtained in OVCAR3. qRT-PCR and Western blot were performed to evaluate expression changes of VEGFA, vimentin, ITGB1, ITGB4, MMP2 and MMP9 under normoxia and hypoxia conditions. Coimmunoprecipitation (Co-IP) was performed in the A2780 cell line to study the interaction between NCK2 and proteins of interest. To investigate whether NCK2 can influence anchorage-independent growth, a soft agar assay was completed. Transwell invasion assay was performed on stable-transfected OVCAR-3 cell lines. RESULTS Nanofluidic data showed NCK2 can play an important role as a factor promoting tumor aggressiveness and survival in OvCa. This role was also linked to the behaviors of ITGB1 and ITGB4. Moreover, in cells overexpressing NCK2, the expression of vimentin, MMP2, MMP9, VEGFA and ITGB1, but not of ITGB4 was induced by hypoxia. Co-IP showed that NCK2 can directly bind ITGB1, but not VEGFA. NCK2 may be involved in mediating cell-extracellular matrix interactions in OvCa cells by influencing tumor aggressiveness. CONCLUSIONS This study provides evidence of a possible role of NCK2 as biomarker of OvCa progression.
Collapse
|
25
|
Totten SM, Adusumilli R, Kullolli M, Tanimoto C, Brooks JD, Mallick P, Pitteri SJ. Multi-lectin Affinity Chromatography and Quantitative Proteomic Analysis Reveal Differential Glycoform Levels between Prostate Cancer and Benign Prostatic Hyperplasia Sera. Sci Rep 2018; 8:6509. [PMID: 29695737 PMCID: PMC5916935 DOI: 10.1038/s41598-018-24270-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/29/2018] [Indexed: 02/06/2023] Open
Abstract
Currently prostate-specific antigen is used for prostate cancer (PCa) screening, however it lacks the necessary specificity for differentiating PCa from other diseases of the prostate such as benign prostatic hyperplasia (BPH), presenting a clinical need to distinguish these cases at the molecular level. Protein glycosylation plays an important role in a number of cellular processes involved in neoplastic progression and is aberrant in PCa. In this study, we systematically interrogate the alterations in the circulating levels of hundreds of serum proteins and their glycoforms in PCa and BPH samples using multi-lectin affinity chromatography and quantitative mass spectrometry-based proteomics. Specific lectins (AAL, PHA-L and PHA-E) were used to target and chromatographically separate core-fucosylated and highly-branched protein glycoforms for analysis, as differential expression of these glycan types have been previously associated with PCa. Global levels of CD5L, CFP, C8A, BST1, and C7 were significantly increased in the PCa samples. Notable glycoform-specific alterations between BPH and PCa were identified among proteins CD163, C4A, and ATRN in the PHA-L/E fraction and among C4BPB and AZGP1 glycoforms in the AAL fraction. Despite these modest differences, substantial similarities in glycoproteomic profiles were observed between PCa and BPH sera.
Collapse
Affiliation(s)
- Sarah M Totten
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Ravali Adusumilli
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Majlinda Kullolli
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Cheylene Tanimoto
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Parag Mallick
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Sharon J Pitteri
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA.
| |
Collapse
|
26
|
Li Z, Li AD, Xu L, Bai DW, Hou KZ, Zheng HC, Qu XJ, Liu YP. SPARC expression in gastric cancer predicts poor prognosis: Results from a clinical cohort, pooled analysis and GSEA assay. Oncotarget 2018; 7:70211-70222. [PMID: 28053291 PMCID: PMC5342547 DOI: 10.18632/oncotarget.12191] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
Background The prognostic role of Secreted Protein Acidic and Rich in Cysteine (SPARC) in gastric cancer (GC) remains controversial. We investigated the clinical significance, the survival relevance, and potential function of SPARC in GC with resected samples, online gene set GSE62254, and cell line SGC7901. Results High immunostaining of SPARC significantly correlated with tumor differentiation (P = 0.004), and independently predicted shorter overall survival (OS) (HR = 1.446, P = 0.022), based on the current IHC evaluation. The accuracy of the results was further validated with 1000 times bootstrapping and the time-dependent receiver-operating characteristics (ROC) curves. The meta-analysis (pooled HR = 1.60, 95% CI: 1.01−2.53) confirmed SPARC as the predictor for reduced OS in GC. Moreover, the association between enhanced SPARC expression and Adriamycin (Adr) sensitivity was revealed by GSEA, and then confirmed by comparative cellular experiments, such as the protein level analysis of SGC7901and SGC7901/Adr cell line. Materials and Methods Immunohistochemistry (IHC) method was used to detect SPARC expression in 137 GC cases. Meta-analysis was performed based on 5 studies published in English on PubMed up to March 2016. GSEA was performed using online data set GSE62254 and GC-related functional gene sets derived from molecular signatures database (MSigDB). Western Blot was carried out to compare protein-level differences between gastric carcinoma SGC7901 cell line and Adr resistant SGC7901/Adr cell line. MTT assay was done to confirm the induction of SPARC on Adr sensitivity Conclusions Increased SPARC expression in GC led to a worse clinical outcome of patients and might induce Adr sensitivity of GC cells.
Collapse
Affiliation(s)
- Zhi Li
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - Ao-Di Li
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - Lu Xu
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - De-Wei Bai
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Department of Cell Biological Treatment Ward, Dalian Centre Hospital, Dalian, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - Ke-Zuo Hou
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - Hua-Chuan Zheng
- Life Science Institute of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xiu-Juan Qu
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| | - Yun-Peng Liu
- Department of Medical Oncology, The First Hospital, China Medical University, Shenyang, Liaoning Province, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province
| |
Collapse
|
27
|
Carrasquinha E, Veríssimo A, Lopes MB, Vinga S. Identification of influential observations in high-dimensional cancer survival data through the rank product test. BioData Min 2018; 11:1. [PMID: 29456628 PMCID: PMC5813402 DOI: 10.1186/s13040-018-0162-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/12/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Survival analysis is a statistical technique widely used in many fields of science, in particular in the medical area, and which studies the time until an event of interest occurs. Outlier detection in this context has gained great importance due to the fact that the identification of long or short-term survivors may lead to the detection of new prognostic factors. However, the results obtained using different outlier detection methods and residuals are seldom the same and are strongly dependent of the specific Cox proportional hazards model selected. In particular, when the inherent data have a high number of covariates, dimensionality reduction becomes a key challenge, usually addressed through regularized optimization, e.g. using Lasso, Ridge or Elastic Net regression. In the case of transcriptomics studies, this is an ubiquitous problem, since each observation has a very high number of associated covariates (genes). RESULTS In order to solve this issue, we propose to use the Rank Product test, a non-parametric technique, as a method to identify discrepant observations independently of the selection method and deviance considered. An example based on the The Cancer Genome Atlas (TCGA) ovarian cancer dataset is presented, where the covariates are patients' gene expressions. Three sub-models were considered, and, for each one, different outliers were obtained. Additionally, a resampling strategy was conducted to demonstrate the methods' consistency and robustness. The Rank Product worked as a consensus method to identify observations that can be influential under survival models, thus potential outliers in the high-dimensional space. CONCLUSIONS The proposed technique allows us to combine the different results obtained by each sub-model and find which observations are systematically ranked as putative outliers to be explored further from a clinical point of view.
Collapse
Affiliation(s)
- Eunice Carrasquinha
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Rovisco Pais, 1, Lisbon, Portugal
| | - André Veríssimo
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Rovisco Pais, 1, Lisbon, Portugal
| | - Marta B. Lopes
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Rovisco Pais, 1, Lisbon, Portugal
| | - Susana Vinga
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Rovisco Pais, 1, Lisbon, Portugal
| |
Collapse
|
28
|
Yan J, Zhang J, Zhang X, Li X, Li L, Li Z, Chen R, Zhang L, Wu J, Wang X, Sun Z, Fu X, Chang Y, Nan F, Yu H, Wu X, Feng X, Li W, Zhang M. SPARC is down-regulated by DNA methylation and functions as a tumor suppressor in T-cell lymphoma. Exp Cell Res 2017; 364:125-132. [PMID: 29277504 DOI: 10.1016/j.yexcr.2017.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
The aim of this study was to assess the functional role of SPARC in T-cell non-Hodgkin's lymphoma (T-NHL), as well as the underlying molecular mechanisms. Here, we first identified SPARC expression in T-NHL tissues and cell lines through western blot and real-time PCR (RT-PCR). Overall survival of T-NHL patients with different levels of SPARC was assessed by Kaplan-Meier survival curves. Then cell proliferation, apoptosis, migration and invasion of T-NHL cells with either knockdown or overexpression of SPARC were determined by MTT, flow cytometry, transwell migration and invasion assay, respectively. Finally, the molecular mechanism by which SPARC modulated T-NHL cell progression was assessed. We confirmed that SPARC was significantly down-regulated in T-NHL tissues and cell lines. T-NHL patients with high levels of SPARC demonstrated a favorable clinical outcome. SPARC significantly suppressed cell proliferation, migration and invasion, and EMT process, but facilitated cell apoptosis in T-NHL cells. Further, we found that loss of SPARC expression in T-NHL tissues and cell lines, both in mRNA and protein levels, was associated with the aberrant DNA methylation in SPRAC gene, and the disrupted SPARC expression could be rescued after treatment with the demethylating agent 5-Aza-2'-deoxycitydine (5-Aza-Cdr). Additionally, 5-Aza-Cdr reversed SPARC hypermethylation to restore its biological role as a tumor suppressor in T-NHL cells, including inhibiting cell proliferation, invasion and migration, while promoting cell apoptosis. Our data provided evidence that DNA methylation in SPARC gene may play a role in the progression of T-NHL.
Collapse
Affiliation(s)
- Jiaqin Yan
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Junhui Zhang
- Department of Otorhinolaryngology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xin Li
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Renyin Chen
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Lei Zhang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Jingjing Wu
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Yu Chang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Feifei Nan
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Hui Yu
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xiaolong Wu
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xiaoyan Feng
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
29
|
Martín M, Chacón JI, Antón A, Plazaola A, García-Martínez E, Seguí MA, Sánchez-Rovira P, Palacios J, Calvo L, Esteban C, Espinosa E, Barnadas A, Batista N, Guerrero A, Muñoz M, Romio E, Rodríguez-Martín C, Caballero R, Casas MI, Rojo F, Carrasco E, Antolín S. Neoadjuvant Therapy with Weekly Nanoparticle Albumin-Bound Paclitaxel for Luminal Early Breast Cancer Patients: Results from the NABRAX Study (GEICAM/2011-02), a Multicenter, Non-Randomized, Phase II Trial, with a Companion Biomarker Analysis. Oncologist 2017; 22:1301-1308. [PMID: 28701571 PMCID: PMC5679821 DOI: 10.1634/theoncologist.2017-0052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/09/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Nanoparticle albumin-bound paclitaxel (nab-Paclitaxel) is an alternative to standard taxanes for breast cancer (BC) treatment. We evaluated nab-Paclitaxel efficacy as neoadjuvant treatment for early estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) disease. MATERIALS AND METHODS Women with ER+, HER2-, stage II-III BC were treated preoperatively with four cycles of weekly nab-Paclitaxel (150 mg/m2), 3 weeks on and 1 week off. We hypothesized that poor pathological response rate (residual cancer burden [RCB] III; Symmans criteria) would be ≤16%. RESULTS Eighty-one patients with a median age of 47 years were treated; 64.2% were premenopausal, and 69% of tumors were stage II. Residual cancer burden III rate was 28.4% (95% confidence interval [CI]: 18.6%-38.2%), RCB 0+I (good response) rate was 24.7% (95% CI: 15.3%-34.1%) and RCB 0 (complete response) rate was 7.4% (95% CI: 1.7%-13.1%). Objective response rate by magnetic resonance imaging was 76.5% and rate of conversion to breast conserving surgery was 40.0%. The most frequent grade 3 and 4 toxicity was neutropenia (12.3% and 3.7% of patients, respectively), without any febrile neutropenia. Sensory neuropathy grade 2 and 3 were seen in 25.9% and 2.5% of patients, respectively. Tumor secreted protein, acidic, cysteine-rich (SPARC) overexpression was significantly associated with RCB 0 (odds ratio: 0.079; 95% CI: 0.009-0.689; p = .0216). CONCLUSION Despite failing to confirm an RCB III rate ≤16% in nab-Paclitaxel-treated patients, the RCB 0+I rate indicates a significant drug antitumor activity with low rates of grade 3-4 toxicity. Our exploratory biomarker analysis suggests a potential predictive role of complete response for SPARC. Confirmatory analyses are warranted, adapting dose and schedule to decrease peripheral neurotoxicity. (Trial registration: European Clinical Trials Database study number: 2011-004476-10; ClinicalTrials.gov: NCT01565499). IMPLICATIONS FOR PRACTICE The pathological response rate (residual cancer burden [RCB]; Symmans criteria) of nanoparticle albumin-bound paclitaxel administered as neoadjuvant treatment for early estrogen receptor-positive, human epidermal growth factor receptor 2-negative disease was evaluated. Whereas poor response (RCB III) was 24.7%, similar to that for docetaxel, good response (RCB 0+I) reached 23.0%, far superior to the 13% for docetaxel, while keeping toxicity low. Exploratory biomarker analysis suggests secreted protein, acidic, cysteine-rich overexpression in tumor cells as a potential predictor of complete response (RCB 0). Findings point to an encouraging single-agent neoadjuvant treatment with low toxicity, which warrants future research and development.
Collapse
Affiliation(s)
- Miguel Martín
- Oncology Department, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Spain, on behalf of GEICAM (Spanish Breast Cancer Group), Spain
| | - José I Chacón
- Oncology Department, Hospital Universitario Virgen de la Salud, Toledo, Spain
| | - Antonio Antón
- Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | - Elena García-Martínez
- Oncology Department, Hospital General Universitario Morales Messeguer, Murcia, Spain
| | - Miguel A Seguí
- Oncology Department, Corporación Sanitaria Parc Taulí, Barcelona, Spain
| | | | - José Palacios
- Pathology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Spain, on behalf of GEICAM (Spanish Breast Cancer Group), Spain
| | - Lourdes Calvo
- Oncology Department, Complejo Hospitalario Universitario de la Coruña, La Coruña, Spain
| | - Carmen Esteban
- Oncology Department, Hospital Universitario Virgen de la Salud, Toledo, Spain
| | - Enrique Espinosa
- Oncology Department, Hospital Universitario la Paz, Madrid, Spain
| | - Agusti Barnadas
- Medical Oncology Department, Hospital de Santa Creu i Sant Pau, Institut de Recerca IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Spain, on behalf of GEICAM (Spanish Breast Cancer Group), Spain
| | - Norberto Batista
- Oncology Department, Hospital Universitario de Canarias, Universidad de La Laguna, Tenerife, Spain
| | - Angel Guerrero
- Oncology Department, Instituto Valenciano de Oncología, Valencia, Spain
| | - Montserrat Muñoz
- Oncology Department, Hospital Clinic i Provincial, Barcelona, Spain
| | - Estefania Romio
- Pathology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | | | - Federico Rojo
- Fundación Jiménez Díaz, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Spain, on behalf of GEICAM (Spanish Breast Cancer Group), Spain
| | | | - Silvia Antolín
- Oncology Department, Complejo Hospitalario Universitario de la Coruña, La Coruña, Spain
| |
Collapse
|
30
|
Peng F, Zhong Y, Liu Y, Zhang Y, Xie Y, Lu Y, Zhang X, Li D. SPARC suppresses lymph node metastasis by regulating the expression of VEGFs in ovarian carcinoma. Int J Oncol 2017; 51:1920-1928. [PMID: 29075785 DOI: 10.3892/ijo.2017.4168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/12/2017] [Indexed: 11/06/2022] Open
Abstract
Lymph node metastasis is one of the most valuable determinants for the prognosis of ovarian cancer. However, the molecular mechanisms underlying lymphangiogenesis in ovarian cancer is still poorly understood. Secreted protein acidic and rich in cysteine (SPARC), a Ca2+-binding matricellular glycoprotein that modulates cell adhesion, migration and differentiation, is thought to play a decisive role in tumor metastasis. Vascular endothelial growth factor (VEGF)-C and VEGF-D contributes to tumor-associated lymphatic vessel growth, enhancing the metastatic spread of tumor cells to lymph nodes. The aim of the present study was to investigate the relationship among SPARC, VEGFs and lymph node metastasis in ovarian cancer. We found that SKOV3 cells expressed high-level SPARC, much more than SKOV3-PM4 cells (a subline with high directional lymphatic metastatic potentials established from the metastatic lymph node generated by human ovarian carcinoma cell line SKOV3 in nude mice) did at both mRNA and protein levels. A SPARC-overexpressed SKOV3-PM4 cell line was constructed and it was found that upregulation of SPARC expression suppressed the growth, migration and invasion of SKOV3-PM4 cells as well as markedly reduced the expression of VEGF-D at both mRNA and protein level by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assay. In 47 of ovarian malignant tissues, the expression of SPARC, VEGF-C and VEGF-D were determined by immunohistochemistry. Lymphatic microvessel density (LVD) and microvessel density (MVD) were evaluated by immunostaining with CD34 and D2-40 antibodies, respectively. We found that SPARC expression was significantly lower in tissues with lymph node metastasis as compared to tissues without lymph node metastasis. SPARC expression was inversely associated with the degree of malignancy and it had a negative correlation with VEGF-C expression, VEGF-D expression, LVD and MVD which were actually higher for advanced tumors than for non-advanced tumors. These results suggest SPARC might function as a tumor suppressor inhibiting angiogenesis and lymphangiogenesis in ovarian cancer by reducing the expression of VEGF-C and VEGF-D.
Collapse
Affiliation(s)
- Fenghui Peng
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yanping Zhong
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yunfeng Liu
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yueming Zhang
- Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yihong Xie
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yingxin Lu
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xinyin Zhang
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Danrong Li
- Affliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
31
|
Chen Y, Zhang Y, Tan Y, Liu Z. Clinical significance of SPARC in esophageal squamous cell carcinoma. Biochem Biophys Res Commun 2017; 492:184-191. [PMID: 28818666 DOI: 10.1016/j.bbrc.2017.08.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 08/12/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Secreted protein acidic and rich in cysteine (SPARC) is matricellular protein that modulates interactions between cells and the extracellular matrix. The role of SPARC in carcinogenesis is controversialin that SPARC can be a tumor suppressor, but overexpression of SPARC is associated with poorer prognosis. METHODS We collected 145 esophageal squamous cell carcinoma and adjacent normal tissues in Shantou, a high incidence region for esophageal cancer. The mRNA and protein expression levels of SPARC in cancer tissue and in adjacent normal mucosa were measured by qRT-PCR, western blot and immunohistochemistry (IHC). RESULTS The mRNA and protein levels of SPARCwere5.78-fold higher in cancer tissues compared with the case-matched normal epithelium. High expression levels of SPARC in ESCC parenchyma, as detected by IHC, were related to lymph node metastasis and poor prognosis (p = 0.049 and p = 0.04). CONCLUSION High expression of SPARC in the parenchyma may be a potential predictor of prognosis, suggesting SPARC could serve as a therapeutic target in ESCC.
Collapse
Affiliation(s)
- Yelong Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong 515041, China; Department of Pathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Ying Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Yang Tan
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong 515041, China.
| |
Collapse
|
32
|
Chioran A, Duncan S, Catalano A, Brown TJ, Ringuette MJ. Collagen IV trafficking: The inside-out and beyond story. Dev Biol 2017; 431:124-133. [PMID: 28982537 DOI: 10.1016/j.ydbio.2017.09.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
Collagen IV networks endow basement membranes (BMs) with remarkable tensile strength and function as morphoregulatory substrata for diverse tissue-specific developmental events. A complex repertoire of intracellular and extracellular molecular interactions are required for collagen IV secretion and supramolecular assembly into BMs. These include intracellular chaperones such as Heat shock protein 47 (Hsp47) and the chaperone-binding trafficking protein Transport and Golgi organization protein 1 (Tango1). Mutations in these proteins lead to compromised collagen IV protomer stability and secretion, leading to defective BM assembly and function. In addition to intracellular chaperones, a role for extracellular chaperones orchestrating the transport, supramolecular assembly, and architecture of collagen IV in BM is emerging. We present evidence derived from evolutionarily distant model organisms that supports an extracellular collagen IV chaperone-like activity for the matricellular protein SPARC (Secreted Protein, Acidic, Rich in Cysteine). Loss of SPARC disrupts BM homeostasis and compromises tissue biomechanics and physiological function. Thus, the combined contributions of intracellular and extracellular collagen IV-associated chaperones and chaperone-like proteins are critical to ensure proper secretion and stereotypic assembly of collagen IV networks in BMs.
Collapse
Affiliation(s)
- Alexa Chioran
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | - Sebastian Duncan
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | | | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, ON, Canada
| | - Maurice J Ringuette
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5.
| |
Collapse
|
33
|
Hung JY, Yen MC, Jian SF, Wu CY, Chang WA, Liu KT, Hsu YL, Chong IW, Kuo PL. Secreted protein acidic and rich in cysteine (SPARC) induces cell migration and epithelial mesenchymal transition through WNK1/snail in non-small cell lung cancer. Oncotarget 2017; 8:63691-63702. [PMID: 28969021 PMCID: PMC5609953 DOI: 10.18632/oncotarget.19475] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix is a component of physiological microenvironment and a regulator of cellular processes such as migration and proliferation. Secreted Protein Acidic and Rich in Cysteine (SPARC/osteonectin) is an extracellular matrix-associated glycoprotein involved in the regulation of cell proliferation and cell migration in several types of cancers. However, the role of SPARC in lung cancer is paradoxical and details of the regulatory mechanism are not well-known. In this study, we investigated novel SPARC-mediated signaling pathways. Treatment of SPARC increased cell proliferation, migration, and mesenchymal phenotype in two non-small cell lung cancer cell lines, CL1-5 and H1299. We found that these phenotypes were not regulated by focal adhesion kinase and Src kinase, but were mediated by with no lysine (K) kinase 1 (WNK1). Suppression of WNK1 expression decreased the expression of SPARC-induced N-cadherin and smooth muscle actin. Moreover, Snail, an important transcription factor for regulating epithelial-mesenchymal transition, is also involved in SPARC/WNK1 pathway. In a murine tumor model, SPARC treatment significantly induced phosphorylation of Akt and WNK1 in lung tumor nodules when compared to control mice. In conclusion, these data suggest that WNK1 is a novel molecule in SPARC-mediated mesenchymal signaling pathway in non-small cell lung cancer.
Collapse
Affiliation(s)
- Jen-Yu Hung
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Fang Jian
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Ying Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-An Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuan-Ting Liu
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Inn-Wen Chong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
34
|
Impact of Secreted Protein Acidic and Rich in Cysteine (SPARC) Expression on Prognosis After Surgical Resection for Biliary Carcinoma. J Gastrointest Surg 2017; 21:990-999. [PMID: 28342122 DOI: 10.1007/s11605-017-3407-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/16/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that influences chemotherapy effectiveness and prognosis. The aim of this study was to investigate whether SPARC expression correlates with the postoperative survival of patients treated with surgical resection for biliary carcinoma. METHODS SPARC expression in resected biliary carcinoma specimens was investigated immunohistochemically in 175 patients. The relationship between SPARC expression and prognosis after surgery was evaluated using univariate and multivariate analyses. RESULTS High SPARC expression in peritumoral stroma was found in 61 (35%) patients. In all patients, stromal SPARC expression was significantly associated with overall survival (OS) (P = 0.006). Multivariate analysis revealed that high stromal SPARC expression was an independent risk factor for poor OS (HR 1.81, P = 0.006). Moreover, high stromal SPARC expression was independently associated with poor prognosis in a subset of 118 patients treated with gemcitabine-based adjuvant chemotherapy (HR 2.04, P = 0.010) but not in the 57 patients who did not receive adjuvant chemotherapy (P = 0.21). CONCLUSIONS Stromal SPARC expression correlated with the prognosis of patients with resectable biliary carcinoma, and its significance was enhanced in patients treated with adjuvant gemcitabine-based chemotherapy.
Collapse
|
35
|
Irles P, Ramos S, Piulachs MD. SPARC preserves follicular epithelium integrity in insect ovaries. Dev Biol 2017; 422:105-114. [DOI: 10.1016/j.ydbio.2017.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
|
36
|
Komiya K, Nakamura T, Nakashima C, Takahashi K, Umeguchi H, Watanabe N, Sato A, Takeda Y, Kimura S, Sueoka-Aragane N. SPARC is a possible predictive marker for albumin-bound paclitaxel in non-small-cell lung cancer. Onco Targets Ther 2016; 9:6663-6668. [PMID: 27822069 PMCID: PMC5089830 DOI: 10.2147/ott.s114492] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Objectives Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) produced good tumor response in cases with lung squamous cell carcinoma, one of the most difficult cancers to treat. Secreted protein acidic and rich in cysteine (SPARC) binds to albumin, suggesting that SPARC plays an important role in tumor uptake of nab-paclitaxel. There is as yet no predictive marker for cytotoxic agents against non-small-cell lung cancer (NSCLC), and hence we believed that SPARC expression might be associated with tumor response to nab-paclitaxel. Patients and methods We studied stromal SPARC reactivity and its association with clinicopathological characteristics in 200 cases of NSCLC using a custom tissue microarray fabricated in our laboratory by immunohistochemical staining. We also investigated the relationship between stromal SPARC reactivity and tumor response to nab-paclitaxel using biopsy or surgical specimens obtained from advanced or recurrent lung cancer patients. Results High SPARC stromal reactivity (>50% of optical fields examined) was detected in 16.5% of cases and intermediate SPARC reactivity (10%–50%) in 56% of cases. High expression in cancer cells was rare (five cases). Stromal SPARC level was correlated with smoking index, squamous cell carcinoma, and vessel invasion. Furthermore, patients with high stromal SPARC reactivity in biopsy specimens such as transbronchial lung biopsy or surgical specimens tended to respond better to nab-paclitaxel. Conclusion Stromal SPARC was detected by immunohistochemical staining in ∼70% of NSCLC cases, and good tumor response to nab-paclitaxel was correlated with high stromal SPARC reactivity. SPARC may be a useful predictive marker for selecting patients likely to respond favorably to nab-paclitaxel treatment.
Collapse
Affiliation(s)
- Kazutoshi Komiya
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Tomomi Nakamura
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Chiho Nakashima
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Koichiro Takahashi
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Hitomi Umeguchi
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University; Japanese Red Cross Karatsu Hospital
| | - Naomi Watanabe
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Akemi Sato
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Yuji Takeda
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University; Department of Thoracic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinya Kimura
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| | - Naoko Sueoka-Aragane
- Department of Internal Medicine, Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University
| |
Collapse
|
37
|
Tumbarello DA, Andrews MR, Brenton JD. SPARC Regulates Transforming Growth Factor Beta Induced (TGFBI) Extracellular Matrix Deposition and Paclitaxel Response in Ovarian Cancer Cells. PLoS One 2016; 11:e0162698. [PMID: 27622658 PMCID: PMC5021370 DOI: 10.1371/journal.pone.0162698] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/26/2016] [Indexed: 12/23/2022] Open
Abstract
TGFBI has been shown to sensitize ovarian cancer cells to the cytotoxic effects of paclitaxel via an integrin receptor-mediated mechanism that modulates microtubule stability. Herein, we determine that TGFBI localizes within organized fibrillar structures in mesothelial-derived ECM. We determined that suppression of SPARC expression by shRNA decreased the deposition of TGFBI in mesothelial-derived ECM, without affecting its overall protein expression or secretion. Conversely, overexpression of SPARC increased TGFBI deposition. A SPARC-YFP fusion construct expressed by the Met5a cell line co-localized with TGFBI in the cell-derived ECM. Interestingly, in vitro produced SPARC was capable of precipitating TGFBI from cell lysates dependent on an intact SPARC carboxy-terminus with in vitro binding assays verifying a direct interaction. The last 37 amino acids of SPARC were shown to be required for the TGFBI interaction while expression of a SPARC-YFP construct lacking this region (aa 1-256) did not interact and co-localize with TGFBI in the ECM. Furthermore, ovarian cancer cells have a reduced motility and decreased response to the chemotherapeutic agent paclitaxel when plated on ECM derived from mesothelial cells lacking SPARC compared to control mesothelial-derived ECM. In conclusion, SPARC regulates the fibrillar ECM deposition of TGFBI through a novel interaction, subsequently influencing cancer cell behavior.
Collapse
Affiliation(s)
- David A. Tumbarello
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, United Kingdom
| | - Melissa R. Andrews
- University of St Andrews, School of Medicine, MBSB, North Haugh, St Andrews, United Kingdom
| | - James D. Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, United Kingdom
| |
Collapse
|
38
|
Pires DP, Cleto S, Sillankorva S, Azeredo J, Lu TK. Genetically Engineered Phages: a Review of Advances over the Last Decade. Microbiol Mol Biol Rev 2016; 80:523-43. [PMID: 27250768 PMCID: PMC4981678 DOI: 10.1128/mmbr.00069-15] [Citation(s) in RCA: 265] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Soon after their discovery in the early 20th century, bacteriophages were recognized to have great potential as antimicrobial agents, a potential that has yet to be fully realized. The nascent field of phage therapy was adversely affected by inadequately controlled trials and the discovery of antibiotics. Although the study of phages as anti-infective agents slowed, phages played an important role in the development of molecular biology. In recent years, the increase in multidrug-resistant bacteria has renewed interest in the use of phages as antimicrobial agents. With the wide array of possibilities offered by genetic engineering, these bacterial viruses are being modified to precisely control and detect bacteria and to serve as new sources of antibacterials. In applications that go beyond their antimicrobial activity, phages are also being developed as vehicles for drug delivery and vaccines, as well as for the assembly of new materials. This review highlights advances in techniques used to engineer phages for all of these purposes and discusses existing challenges and opportunities for future work.
Collapse
Affiliation(s)
- Diana P Pires
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Sara Cleto
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Sanna Sillankorva
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Joana Azeredo
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Timothy K Lu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
39
|
Boyineni J, Tanpure S, Gnanamony M, Antony R, Fernández KS, Lin J, Pinson D, Gondi CS. SPARC overexpression combined with radiation retards angiogenesis by suppressing VEGF-A via miR‑410 in human neuroblastoma cells. Int J Oncol 2016; 49:1394-406. [PMID: 27498840 PMCID: PMC5021251 DOI: 10.3892/ijo.2016.3646] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/06/2016] [Indexed: 12/11/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children and despite aggressive therapy survival rates remain low. One of the contributing factors for low survival rates is aggressive tumor angiogenesis, which is known to increase due to radiation, one of the standard therapies for neuroblastoma. Therefore, targeting tumor angiogenesis can be a viable add-on therapy for the treatment of neuroblastomas. In the present study, we demonstrate that overexpression of secreted protein acidic and rich in cysteine (SPARC) suppresses radiation induced angiogenesis in SK-N-BE(2) and NB1691 neuroblastoma cells. We observed that overexpression of SPARC in SK-N-BE(2) and NB1691 cells reduced radiation induced angiogenesis in an in vivo mouse dorsal skin model and an ex vivo chicken CAM (chorioallantoic-membrane) model and also reduced tumor size in subcutaneous mouse tumor models of NB. We also observed that SPARC overexpression reduces VEGF-A expression, in SK-N-BE(2) and NB1691 NB cells via miR-410, a VEGF-A targeting microRNA. SPARC overexpression alone or in combination with miR-410 and radiation was shown to be effective at reducing angiogenesis. Moreover, addition of miR-410 inhibitors reversed SPARC mediated inhibition of VEGF-A in NB1691 cells but not in SK-N-BE(2) NB cells. In conclusion, the present study demonstrates that the over-expression of SPARC in combination with radiation reduced tumor angiogenesis by downregulating VEGF-A via miR-410.
Collapse
Affiliation(s)
- Jerusha Boyineni
- Department of Internal Medicine, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Smita Tanpure
- Department of Internal Medicine, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Manu Gnanamony
- Department of Internal Medicine, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Reuben Antony
- Department of Pediatrics, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Karen S Fernández
- Department of Pediatrics, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Julian Lin
- Department of Neurosurgery, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - David Pinson
- Department of Pathology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Christopher S Gondi
- Department of Internal Medicine, University of Illinois College of Medicine, Peoria, IL 61605, USA
| |
Collapse
|
40
|
Jayaraman S, Doucet M, Lau WM, Kominsky SL. CITED2 Modulates Breast Cancer Metastatic Ability through Effects on IKKα. Mol Cancer Res 2016; 14:730-9. [PMID: 27216153 PMCID: PMC4987170 DOI: 10.1158/1541-7786.mcr-16-0081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/14/2016] [Indexed: 12/31/2022]
Abstract
UNLABELLED Previously, we identified the transcriptional coactivator CITED2 as a potential facilitator of bone metastasis using a murine mammary cancer model. Extending these studies to human breast cancer, it was observed that CITED2 mRNA expression was significantly elevated in patient specimens of metastatic breast cancer relative to primary tumors, with highest levels in metastasis to bone relative to non-bone sites. To further evaluate CITED2 functions in breast cancer metastasis, CITED2 expression was stably reduced in the human breast cancer cell lines MDA-MB-231 and MDA-MB-468, which are metastatic in animal models. While CITED2 knockdown had no effect on cell proliferation, cell migration and invasion were significantly reduced, as was the establishment of metastasis following intracardiac administration in athymic nude mice. To explore the mechanism behind these effects, gene expression following CITED2 knockdown in MDA-MB-231 cells by cDNA microarray was performed. As confirmed at the mRNA and protein levels in both MDA-MB-231 and MDA-MB-468 cells, expression of the NF-κB regulator IKKα was significantly reduced, along with several NF-κB targets with known roles in metastasis (OPN, MMP9, uPA, SPARC, IL11, and IL1β). Furthermore, ChIP assay revealed recruitment of CITED2 to the promoter of IKKα, indicating a direct role in regulating its expression. Consistent with reduced IKKα expression, CITED2 knockdown inhibited both canonical and noncanonical NF-κB signaling. Finally, restoration of IKKα expression following CITED2 knockdown in MDA-MB-231 and MDA-MB-468 cells rescued their invasive ability. Collectively, these data demonstrate that CITED2 modulates metastatic ability in human breast cancer cells, at least in part, through the regulation of IKKα. IMPLICATIONS The current study highlights the role of CITED2 in facilitating breast cancer metastasis, partly via regulation of IKKα. Mol Cancer Res; 14(8); 730-9. ©2016 AACR.
Collapse
Affiliation(s)
- Swaathi Jayaraman
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michele Doucet
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wen Min Lau
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Scott L Kominsky
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
41
|
Hellewell AL, Adams JC. Insider trading: Extracellular matrix proteins and their non-canonical intracellular roles. Bioessays 2015; 38:77-88. [PMID: 26735930 DOI: 10.1002/bies.201500103] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In metazoans, the extracellular matrix (ECM) provides a dynamic, heterogeneous microenvironment that has important supportive and instructive roles. Although the primary site of action of ECM proteins is extracellular, evidence is emerging for non-canonical intracellular roles. Examples include osteopontin, thrombospondins, IGF-binding protein 3 and biglycan, and relate to roles in transcription, cell-stress responses, autophagy and cancer. These findings pose conceptual problems on how proteins signalled for secretion can be routed to the cytosol or nucleus, or can function in environments with diverse redox, pH and ionic conditions. We review evidence for intracellular locations and functions of ECM proteins, and current knowledge of the mechanisms by which they may enter intracellular compartments. We evaluate the experimental methods that are appropriate to obtain rigorous evidence for intracellular localisation and function. Better insight into this under-researched topic is needed to decipher the complete spectrum of physiological and pathological roles of ECM proteins.
Collapse
|
42
|
Secreted protein acidic and rich in cysteine enhances the chemosensitivity of pancreatic cancer cells to gemcitabine. Tumour Biol 2015; 37:2267-73. [DOI: 10.1007/s13277-015-4044-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/02/2015] [Indexed: 02/07/2023] Open
|
43
|
Niskakoski A, Kaur S, Staff S, Renkonen-Sinisalo L, Lassus H, Järvinen HJ, Mecklin JP, Bützow R, Peltomäki P. Epigenetic analysis of sporadic and Lynch-associated ovarian cancers reveals histology-specific patterns of DNA methylation. Epigenetics 2015; 9:1577-87. [PMID: 25625843 PMCID: PMC4622692 DOI: 10.4161/15592294.2014.983374] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Diagnosis and treatment of epithelial ovarian cancer is challenging due to the poor understanding of the pathogenesis of the disease. Our aim was to investigate epigenetic mechanisms in ovarian tumorigenesis and, especially, whether tumors with different histological subtypes or hereditary background (Lynch syndrome) exhibit differential susceptibility to epigenetic inactivation of growth regulatory genes. Gene candidates for epigenetic regulation were identified from the literature and by expression profiling of ovarian and endometrial cancer cell lines treated with demethylating agents. Thirteen genes were chosen for methylation-specific multiplex ligation-dependent probe amplification assays on 104 (85 sporadic and 19 Lynch syndrome-associated) ovarian carcinomas. Increased methylation (i.e., hypermethylation) of variable degree was characteristic of ovarian carcinomas relative to the corresponding normal tissues, and hypermethylation was consistently more prominent in non-serous than serous tumors for individual genes and gene sets investigated. Lynch syndrome-associated clear cell carcinomas showed the highest frequencies of hypermethylation. Among endometrioid ovarian carcinomas, lower levels of promoter methylation of RSK4, SPARC, and HOXA9 were significantly associated with higher tumor grade; thus, the methylation patterns showed a shift to the direction of high-grade serous tumors. In conclusion, we provide evidence of a frequent epigenetic inactivation of RSK4, SPARC, PROM1, HOXA10, HOXA9, WT1-AS, SFRP2, SFRP5, OPCML, and MIR34B in the development of non-serous ovarian carcinomas of Lynch and sporadic origin, as compared to serous tumors. Our findings shed light on the role of epigenetic mechanisms in ovarian tumorigenesis and identify potential targets for translational applications.
Collapse
Affiliation(s)
- Anni Niskakoski
- a Department of Medical Genetics; Biomedicum Helsinki ; University of Helsinki ; Helsinki , Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
LI AODI, QU XIUJUAN, LI ZHI, QU JINGLEI, SONG NA, MA YANJU, LIU YUNPENG. Secreted protein acidic and rich in cysteine antagonizes bufalin-induced apoptosis in gastric cancer cells. Mol Med Rep 2015; 12:2926-32. [DOI: 10.3892/mmr.2015.3676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 08/14/2014] [Indexed: 11/06/2022] Open
|
45
|
Chudecka-Głaz AM, Cymbaluk-Płoska AA, Menkiszak JL, Pius-Sadowska E, Machaliński BB, Sompolska-Rzechuła A, Rzepka-Górska IA. Assessment of selected cytokines, proteins, and growth factors in the peritoneal fluid of patients with ovarian cancer and benign gynecological conditions. Onco Targets Ther 2015; 8:471-85. [PMID: 25750541 PMCID: PMC4348053 DOI: 10.2147/ott.s73438] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objectives The ovarian tumor microenvironment, ie, the peritoneal fluid, is an intriguing research subject. The goal of this study was to assess the behavior of selected cytokines and growth factors within the peritoneal fluid in pathologies associated with ascites and to assess the relationship between the levels of these substances and select prognostic factors of ovarian cancer. Methods A total of 74 patients were enrolled in the study, including 36 patients with ovarian cancer and 38 patients with benign gynecological conditions. Peritoneal fluid collected during surgical procedures was used to assess the levels of interleukin (IL)-6, IL-8, stem cell factor (SCF), dickkopf-1, growth differentiation factor-15 (GDF-15), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), osteoprotegerin (OPG), osteopontin, osteonectin, and human epididymis protein 4. The median levels of these factors were compared between the two groups, and the levels of selected factors were assessed in the ovarian cancer group with regard to the clinical stage of cancer, tumor differentiation, presence of peritoneal spread and positive peritoneal fluid cytology results. The diagnostic value of the analyzed proteins within the peritoneal fluid was also assessed. Results Differences were observed between the patients with ovarian cancer and the patients with benign gynecological conditions associated with ascites with regard to the levels of IL-6, IL-8, GDF-15, SCF, osteopontin, osteonectin, and OPG. There were no differences in dickkopf-1, TRAIL, and human epididymis protein 4 levels between the two study groups. Cancer stage affected only the mean SCF and OPG levels, with lower SCF values and higher OPG values in advanced cancers compared to less-advanced cancers. Tumor differentiation was associated with significantly lower SCF values in the group of poorly differentiated tumors. A significant reduction in SCF values and a significant increase in OPG and IL-6 values were also observed within cancer cell-positive peritoneal fluid. Peritoneal spread was associated with higher levels of TRAIL, osteonectin, and IL-6 in ovarian cancer patients. Conclusion On the basis of the conducted studies, it appears that of the studied factors, GDF-15, SCF, and OPG deserve special attention in the context of future research on the tumor microenvironment. With regard to diagnostics, attention should be given primarily to GDF-15, IL-6, and osteonectin.
Collapse
Affiliation(s)
- Anita Monika Chudecka-Głaz
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| | - Aneta Alicja Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| | - Janusz Leszek Menkiszak
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | | | | | - Izabella Anna Rzepka-Górska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
46
|
Annexin A2 plays a critical role in epithelial ovarian cancer. Arch Gynecol Obstet 2014; 292:175-82. [PMID: 25547062 DOI: 10.1007/s00404-014-3598-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE This study aimed at investigating the potential role and prognostic significance of Annexin A2 in human epithelial ovarian cancer (EOC). METHODS Western blot was used to evaluate the expression of Annexin A2 in nine fresh EOC tissues, and immunohistochemical analysis was performed on formalin-fixed paraffin-embedded sections of 119 cases of ovarian cancers. Then, we used Fisher exact test to analyze the correlation between Annexin A2 and clinicopathological parameters. Starvation refeeding was used to detect the alteration of Annexin A2 in HO8910 cell cycle. RESULTS Annexin A2 was overexpressed in carcinoma tissues compared with normal tissue, and the expression levels gradually increased from G1 to G3. Moreover, the staining of tissue microarray was consistent with the result we got from western blot, increasing from G1 to G3 gradually, and it was related to the Figo stage (P = 0.005), histologic grade (P = 0.002), ascite (P < 0.001), malignant tumor cells (P < 0.001), residual tumor size (P = 0.044), Ki-67 (P = 0.003). Kaplan-Meier analysis revealed that high Annexin A2 expression was significantly associated with poor prognoses of the patients (P < 0.001). Multivariate analysis demonstrated that Annexin A2 was an independent prognostic indicator for overall survival. Starvation refeeding indicated that Annexin A2 was related to EOC cell proliferation. CONCLUSIONS We could hypothesize that Annexin A2 acted a critical role in EOC cell proliferation, and may be used as a potential and novel therapeutic target for EOC. These data suggest that Annexin A2 may promote the progression of EOC and be a therapeutic target for EOC therapy.
Collapse
|
47
|
Mateo F, Meca-Cortés O, Celià-Terrassa T, Fernández Y, Abasolo I, Sánchez-Cid L, Bermudo R, Sagasta A, Rodríguez-Carunchio L, Pons M, Cánovas V, Marín-Aguilera M, Mengual L, Alcaraz A, Schwartz S, Mellado B, Aguilera KY, Brekken R, Fernández PL, Paciucci R, Thomson TM. SPARC mediates metastatic cooperation between CSC and non-CSC prostate cancer cell subpopulations. Mol Cancer 2014; 13:237. [PMID: 25331979 PMCID: PMC4210604 DOI: 10.1186/1476-4598-13-237] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 10/08/2014] [Indexed: 12/29/2022] Open
Abstract
Background Tumor cell subpopulations can either compete with each other for nutrients and physical space within the tumor niche, or co-operate for enhanced survival, or replicative or metastatic capacities. Recently, we have described co-operative interactions between two clonal subpopulations derived from the PC-3 prostate cancer cell line, in which the invasiveness of a cancer stem cell (CSC)-enriched subpopulation (PC-3M, or M) is enhanced by a non-CSC subpopulation (PC-3S, or S), resulting in their accelerated metastatic dissemination. Methods M and S secretomes were compared by SILAC (Stable Isotope Labeling by Aminoacids in Cell Culture). Invasive potential in vitro of M cells was analyzed by Transwell-Matrigel assays. M cells were co-injected with S cells in the dorsal prostate of immunodeficient mice and monitored by bioluminescence for tumor growth and metastatic dissemination. SPARC levels were determined by immunohistochemistry and real-time RT-PCR in tumors and by ELISA in plasma from patients with metastatic or non-metastatic prostate cancer. Results Comparative secretome analysis yielded 213 proteins differentially secreted between M and S cells. Of these, the protein most abundantly secreted in S relative to M cells was SPARC. Immunodepletion of SPARC inhibited the enhanced invasiveness of M induced by S conditioned medium. Knock down of SPARC in S cells abrogated the capacity of its conditioned medium to enhance the in vitro invasiveness of M cells and compromised their potential to boost the metastatic behavior of M cells in vivo. In most primary human prostate cancer samples, SPARC was expressed in the epithelial tumoral compartment of metastatic cases. Conclusions The matricellular protein SPARC, secreted by a prostate cancer clonal tumor cell subpopulation displaying non-CSC properties, is a critical mediator of paracrine effects exerted on a distinct tumor cell subpopulation enriched in CSC. This paracrine interaction results in an enhanced metastatic behavior of the CSC-enriched tumor subpopulation. SPARC is expressed in the neoplastic cells of primary prostate cancer samples from metastatic cases, and could thus constitute a tumor progression biomarker and a therapeutic target in advanced prostate cancer. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-237) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Timothy M Thomson
- Department of Cell Biology, Molecular Biology Institute of Barcelona, National Research Council (CSIC), c, Baldiri Reixac 15-21, Barcelona 08028, Spain.
| |
Collapse
|
48
|
Deep, noninvasive imaging and surgical guidance of submillimeter tumors using targeted M13-stabilized single-walled carbon nanotubes. Proc Natl Acad Sci U S A 2014; 111:13948-53. [PMID: 25214538 DOI: 10.1073/pnas.1400821111] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Highly sensitive detection of small, deep tumors for early diagnosis and surgical interventions remains a challenge for conventional imaging modalities. Second-window near-infrared light (NIR2, 950-1,400 nm) is promising for in vivo fluorescence imaging due to deep tissue penetration and low tissue autofluorescence. With their intrinsic fluorescence in the NIR2 regime and lack of photobleaching, single-walled carbon nanotubes (SWNTs) are potentially attractive contrast agents to detect tumors. Here, targeted M13 virus-stabilized SWNTs are used to visualize deep, disseminated tumors in vivo. This targeted nanoprobe, which uses M13 to stably display both tumor-targeting peptides and an SWNT imaging probe, demonstrates excellent tumor-to-background uptake and exhibits higher signal-to-noise performance compared with visible and near-infrared (NIR1) dyes for delineating tumor nodules. Detection and excision of tumors by a gynecological surgeon improved with SWNT image guidance and led to the identification of submillimeter tumors. Collectively, these findings demonstrate the promise of targeted SWNT nanoprobes for noninvasive disease monitoring and guided surgery.
Collapse
|
49
|
Feki-Tounsi M, Hamza-Chaffai A. Cadmium as a possible cause of bladder cancer: a review of accumulated evidence. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:10561-73. [PMID: 24894749 DOI: 10.1007/s11356-014-2970-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 04/23/2014] [Indexed: 05/16/2023]
Abstract
Bladder cancer is a significant disease, the rates of which have increased over the few last years. However, its etiology remains as yet undefined. Cadmium, a widespread environmental carcinogen that has received considerable interest, presents evidence as a possible cause of bladder cancer. A literature review was conducted from the years 1984-2013 to study the accumulated evidence for cadmium as a possible cause of bladder cancer, including routes of cadmium exposure, accumulation, toxicity, carcinogenicity, and evidence from epidemiological and experimental studies. Special reference is devoted to cadmium nephrotoxicity, which illustrates how cadmium exerts its effects on the transitional epithelium of the urinary tract. Mechanisms of carcinogenesis are discussed. The effects of cadmium on gene expression in urothelial cells exposed to cadmium are also addressed. Despite different methodologies, several epidemiologic and nephrotoxicity studies of cadmium indicate that occupational exposure to cadmium is associated with increased risk of bladder cancer and provide additional evidence that cadmium is a potential toxic element in urothelial cells. In vitro studies provide further evidence that cadmium is involved in urothelial carcinogenesis. Animal studies encounter several problems such as morphology differences between species. Among the complex mechanisms of cadmium carcinogenesis, gene expression deregulation is the subject of recent studies on bladder cadmium-induced carcinogenesis. Further research, however, will be required to promise a better understanding of the mechanisms underlying cadmium carcinogenesis and to establish the precise role of cadmium in this important malignancy.
Collapse
Affiliation(s)
- Molka Feki-Tounsi
- Unit of Marine and Environmental Toxicology, IPEIS, Sfax University, PB 805, 3018, Sfax, Tunisia,
| | | |
Collapse
|
50
|
Nakashima S, Kobayashi S, Sakai D, Tomokuni A, Tomimaru Y, Hama N, Wada H, Kawamoto K, Marubashi S, Eguchi H, Matsuura N, Doki Y, Mori M, Nagano H. Prognostic impact of tumoral and/or peri-tumoral stromal SPARC expressions after surgery in patients with biliary tract cancer. J Surg Oncol 2014; 110:1016-22. [PMID: 25155283 DOI: 10.1002/jso.23767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 07/29/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVE SPARC (secreted protein acidic and rich in cysteine) is a matricellular glycoprotein that modulates interactions between tumoral cells and the peri-tumoralstroma. SPARC induces proliferation and invasion in vitro, and is a poor prognostic factor in several gastrointestinal cancers. Herein, we evaluated the prognostic value of tumoral and stromal SPARC expression in patients with biliary tract cancer (BTC) after surgery. METHODS We examined immunohistochemical patterns of SPARC expression in 110 resected BTC specimens and evaluated the prognostic value using prospectively collected data. RESULTS SPARC was expressed in tumoral cells in 46 samples (42%) and inperi-tumoralstromain 65 samples (59%). Tumoral SPARC expression was not related to major patient characteristics. Stromal SPARC expression was related to lymph node metastasis, stage, margin status, and tumor location. Overall survival at 5 years after surgery was 34.2%. Stromal SPARC (P < 0.001) and tumoral SPARC (P = 0.048) were associated with poor prognosis. Multivariate analysis revealed invasion into lymphatic system, residual tumor, and stromal SPARC as independent prognostic factors. The hazard ratio for patients with positive stromal SPARC was 3.20 (P < 0.001). CONCLUSION SPARC expression inperi-tumoralstroma predicts a poor prognosis for patients with BTC after surgery.
Collapse
Affiliation(s)
- Shinsuke Nakashima
- Department of Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|