1
|
Yang X, Xia Y, Wang S, Sun C. Prognostic value of SPARC in hepatocellular carcinoma: A systematic review and meta-analysis. PLoS One 2022; 17:e0273317. [PMID: 35981080 PMCID: PMC9387809 DOI: 10.1371/journal.pone.0273317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/06/2022] [Indexed: 01/30/2023] Open
Abstract
Objective
Hepatocellular carcinoma (HCC) is characterized by a high degree of malignancy, rapid proliferation of tumor cells, and early liver metastasis. Resistance to multiple drugs independent of the high expression of secreted protein acidic and rich in cysteine (SPARC) is associated with a high risk of recurrence and mortality. However, the prognostic value of SPARC in patients with HCC remains unclear. Therefore, we performed a meta-analysis to evaluate the relationship between the expression of SPARC and the prognosis of patients with HCC.
Methods
We searched for relevant articles in the CNKI, PubMed, EMBASE, and Web of Science databases. The 95% confidence intervals (CIs) were calculated for combined overall survival (OS) and disease-free survival (DFS) to assess the prognostic value of expression of SPARC in patients with HCC.
Results
In six of the studies, SPARC expression status was significantly associated with OS (combined hazard ratio [HR], 1.38; 95% CI, 1.0–1.82; Z = 2.27, P = 0.02) but not with DFS (combined HR, 0.79; 95% CI, 0.16–4.00, Z = 0.28, P = 0.78). Therefore, it cannot be assumed that upregulated SPARC expression has an effect on DFS in patients with HCC.
Conclusion
Elevated SPARC expression is associated with a low survival rate but not with DFS in patients with HCC. Further studies are needed to confirm our conclusions.
Registration
INPLASY registration number: INPLASY202180115. https://inplasy.com/inplasy-2021-8-0115/.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Oncology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Yunhong Xia
- Department of Oncology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- * E-mail:
| | - Shuomin Wang
- Department of Oncology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Chen Sun
- Department of Oncology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
2
|
Carriere P, Calvo N, Novoa Díaz MB, Lopez-Moncada F, Herrera A, Torres MJ, Alonso E, Gandini NA, Gigola G, Contreras HR, Gentili C. Role of SPARC in the epithelial-mesenchymal transition induced by PTHrP in human colon cancer cells. Mol Cell Endocrinol 2021; 530:111253. [PMID: 33781836 DOI: 10.1016/j.mce.2021.111253] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Parathyroid hormone-related peptide (PTHrP) exerts its effects on cells derived from colorectal cancer (CRC) and tumor microenvironment and is involved in processes requiring the epithelial-mesenchymal transition (EMT). Here, we report that PTHrP modulates factors expression and morphological changes associated with EMT in HCT116 cells from CRC. PTHrP increased the protein expression of SPARC, a factor involved in EMT, in HCT116 cells but not in Caco-2 cells also from CRC but with less aggressiveness. PTHrP also increased SPARC expression and its subsequent release from endothelial HMEC-1 cells. The conditioned media of PTHrP-treated HMEC-1 cells induced early changes related to EMT in HCT116 cells. Moreover, SPARC treatment on HCT116 cells potentiated PTHrP modulation in E-cadherin expression and cell migration. In vivo PTHrP also increased SPARC expression and decreased E-cadherin expression. These results suggest a novel PTHrP action on CRC progression involving the microenvironment in the modulation of events associated with EMT.
Collapse
Affiliation(s)
- Pedro Carriere
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Natalia Calvo
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - María Belén Novoa Díaz
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Fernanda Lopez-Moncada
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - Alexander Herrera
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - María José Torres
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | | | | | - Graciela Gigola
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Hector R Contreras
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - Claudia Gentili
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina.
| |
Collapse
|
3
|
Abstract
Cancer is a complex disease with high incidence and mortality rates. The important role played by the tumor microenvironment in regulating oncogenesis, tumor growth, and metastasis is by now well accepted in the scientific community. SPARC is known to participate in tumor-stromal interactions and impact cancer growth in ambiguous ways, which either enhance or suppress cancer aggressiveness, in a context-dependent manner. p53 transcription factor, a well-established tumor suppressor, has been reported to promote tumor growth in certain situations, such as hypoxia, thus displaying a duality in its action. Although both proteins are being tested in clinical trials, the synergistic relation between them is yet to be explored in clinical practice. In this review, we address the controversial roles of SPARC and p53 as double agents in cancer, briefly summarizing the interaction found between these two molecules and its importance in cancer.
Collapse
|
4
|
Kurtul N, Taşdemir EA, Ünal D, İzmirli M, Eroglu C. SPARC: As a prognostic biomarker in rectal cancer patients treated with chemo-radiotherapy. Cancer Biomark 2018; 18:459-466. [PMID: 28009327 DOI: 10.3233/cbm-161733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The aim of this study is to search the prognostic value of SPARC expression in rectum cancer cases receiving postoperative radiotherapy. METHODS Forty three rectal cancer patients are recruited to this retrospective study. All patients received postoperative radiotherapy which the median dose was 5040 cGy and concomitant chemotherapy. Samples taken from their paraffin blocks were examined with immunohistochemical procedures. RESULTS When the association between SPARC expression and the clinicopathological feature was examined, there was a significant association between age and expression levels. Overall survival of patients with low expression was found to be 67 months whereas the overall survival of the patients with high expression was 32 months and the difference was statistically significant. Time to local recurrence of patients with low expression was found to be 74 months whereas time to local recurrence of the patients with high expression was 31 months. Progression free survival of the patients with low expression and high expression were 67 months and 32 months, respectively. In multivariate Cox regression analyses, high expression of SPARC was found to be associated with a statistically significant shorter overall survival and progression free survival. CONCLUSIONS High expression of SPARC is related to worse prognosis in rectal cancer patients.
Collapse
Affiliation(s)
- Neslihan Kurtul
- Department of Radiation Oncology, Faculty of Medicine, University of Sütçü İmam, Kahramanmaras, Turkey
| | | | - Dilek Ünal
- Department of Radiation Oncology, Kayseri Research Hospital, Kayseri, Turkey
| | - Mustafa İzmirli
- Department of Radiation Oncology, Faculty of Medicine, University of Sütçü İmam, Kahramanmaras, Turkey
| | - Celalettin Eroglu
- Department of Radiation Oncology, Faculty of Medicine, University of Erciyes, Kayseri, Turkey
| |
Collapse
|
5
|
Pieniazek M, Donizy P, Halon A, Leskiewicz M, Matkowski R. Prognostic significance of immunohistochemical epithelial–mesenchymal transition markers in skin melanoma patients. Biomark Med 2016; 10:975-85. [DOI: 10.2217/bmm-2016-0133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate secreted protein acidic and rich in cystein (SPARC) and neural cadherin (NCAD), which are associated with epithelial–mesenchymal transition in primary skin melanoma and nodal metastases and their prognostic impact in melanoma patients. Methods: Expression of proteins was assessed by immunochemistry in archival paraffin samples from 103 primary melanoma tumors and 16 nodal metastases. Results: Increased expression of SPARC and NCAD in primary skin melanoma was associated with decreased overall survival, adverse clinicopathological features and particularly with microsatellitosis (SPARC) and ulceration (NCAD). In univariate Cox regression analysis, both biomarkers were significantly associated with the risk of death; the multivariate Cox regression analysis identified no significance. Conclusion: The most important result of our study was that we confirmed the strict correlation between SPARC and NCAD expression and clinicopathological parameters related with melanoma progression, which is a specific clinical equivalent of the molecular mechanisms of epithelial–mesenchymal transition process and confirms its key role in the disease outcome.
Collapse
Affiliation(s)
- Malgorzata Pieniazek
- Department of Clinical Oncology, Tadeusz Koszarowski Regional Oncology Center, Opole, Katowicka 66a, Poland
| | - Piotr Donizy
- Department of Pathomorphology & Oncological Cytology, Wroclaw Medical University, Borowska 213, 50–556 Wroclaw, Poland
| | - Agnieszka Halon
- Department of Pathomorphology & Oncological Cytology, Wroclaw Medical University, Borowska 213, 50–556 Wroclaw, Poland
| | - Marek Leskiewicz
- Department of Statistics, Wroclaw University of Economics, Komandorska 118–120, 53–345 Wroclaw, Poland
| | - Rafal Matkowski
- Department of Oncology & Division of Surgical Oncology, Wroclaw Medical University, pl. Hirszfelda 12, 53–413 Wroclaw, Poland
- Lower Silesian Oncology Centre, pl. Hirszfelda 12, 53–413 Wroclaw, Poland
| |
Collapse
|
6
|
Salvatierra E, Alvarez MJ, Leishman CC, Rivas Baquero E, Lutzky VP, Chuluyan HE, Podhajcer OL. SPARC Controls Melanoma Cell Plasticity through Rac1. PLoS One 2015; 10:e0134714. [PMID: 26248315 PMCID: PMC4527691 DOI: 10.1371/journal.pone.0134714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/13/2015] [Indexed: 12/20/2022] Open
Abstract
Cell transition to a more aggressive mesenchymal-like phenotype is a hallmark of cancer progression that involves different steps and requires tightly regulated cell plasticity. SPARC (Secreted Protein Acidic and Rich in Cysteine) is a matricellular protein that promotes this transition in various malignant cell types, including melanoma cells. We found that suppression of SPARC expression in human melanoma cells compromised cell migration, adhesion, cytoskeleton structure, and cell size. These changes involved the Akt/mTOR pathway. Re-expression of SPARC or protein addition restored all the cell features. Suppression of SPARC expression was associated with increased Rac1-GTP levels and its membrane localization. Expression of the dominant negative mutant of Rac1 counteracted almost all the changes observed in SPARC-deficient cells. Overall, these data suggest that most of the SPARC-mediated effects occurred mainly through the blockade of Rac1 activity.
Collapse
Affiliation(s)
- Edgardo Salvatierra
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir-CONICET, Buenos Aires, C1405, Argentina
| | - Mariano J. Alvarez
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir-CONICET, Buenos Aires, C1405, Argentina
| | - Claudia C. Leishman
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir-CONICET, Buenos Aires, C1405, Argentina
| | - Elvia Rivas Baquero
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir-CONICET, Buenos Aires, C1405, Argentina
| | - Viviana P. Lutzky
- Laboratory of Immunomodulators, School of Medicine, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-University of Buenos Aires, Buenos Aires, Argentina
| | - H. Eduardo Chuluyan
- Laboratory of Immunomodulators, School of Medicine, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-University of Buenos Aires, Buenos Aires, Argentina
| | - Osvaldo L. Podhajcer
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir-CONICET, Buenos Aires, C1405, Argentina
- * E-mail:
| |
Collapse
|
7
|
Ribeiro N, Sousa SR, Brekken RA, Monteiro FJ. Role of SPARC in bone remodeling and cancer-related bone metastasis. J Cell Biochem 2014; 115:17-26. [PMID: 24038053 DOI: 10.1002/jcb.24649] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 08/13/2013] [Indexed: 12/23/2022]
Abstract
There is a growing socioeconomic recognition that clinical bone diseases such as bone infections, bone tumors and osteoporotic bone loss mainly associated with ageing, are major issues in today's society. SPARC (secreted protein, acidic and rich in cysteine), a matricellular glycoprotein, may be a promising therapeutic target for preventing or treating bone-related diseases. In fact, SPARC is associated with tissue remodeling, repair, development, cell turnover, bone mineralization and may also participate in growth and progression of tumors, namely cancer-related bone metastasis. Yet, the function of SPARC in such biological processes is poorly understood and controversial. The main objective of this work is to review the current knowledge related to the activity of SPARC in bone remodeling, tumorigenesis, and bone metastasis. Progress in understanding SPARC biology may provide novel strategies for bone regeneration and the development of anti-angiogenic, anti-proliferative, or counter-adhesive treatments specifically against bone metastasis.
Collapse
Affiliation(s)
- Nilza Ribeiro
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal; Departamento de Engenharia Metalúrgica e de Materiais, Faculdade de Engenharia (FEUP), Universidade do Porto, Rua Roberto Frias, s/n, 4200-465, Porto, Portugal
| | | | | | | |
Collapse
|
8
|
Nagaraju GP, Dontula R, El-Rayes BF, Lakka SS. Molecular mechanisms underlying the divergent roles of SPARC in human carcinogenesis. Carcinogenesis 2014; 35:967-73. [PMID: 24675529 DOI: 10.1093/carcin/bgu072] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Communication between the cell and its surrounding environment, consisting of proteinaceous (non-living material) and extracellular matrix (ECM), is important for biophysiological and chemical signaling. This signaling results in a range of cellular activities, including cell division, adhesion, differentiation, invasion, migration and angiogenesis. The ECM non-structural secretory glycoprotein called secreted protein, acidic and rich in cysteine (SPARC), plays a significant role in altering cancer cell activity and the tumor's microenvironment (TME). However, the role of SPARC in cancer research has been the subject of controversy. This review mainly focuses on recent advances in understanding the contradictory nature of SPARC in relation to ECM assembly, cancer cell proliferation, adhesion, migration, apoptosis and tumor growth.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA and
| | | | | | | |
Collapse
|
9
|
Kim JY, Jeong D, Ahn TS, Kim HJ, Park DS, Park SY, Bae SB, Lee S, Lee SS, Lee MS, Cho HD, Baek MJ. Expression of Secreted Protein Acidic and Rich in Cysteine in the Stroma of a Colorectal Carcinoma is Associated With Patient Prognosis. Ann Coloproctol 2013; 29:93-9. [PMID: 23862126 PMCID: PMC3710779 DOI: 10.3393/ac.2013.29.3.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/17/2013] [Indexed: 01/17/2023] Open
Abstract
Purpose Secreted protein acidic and rich in cysteine (SPARC), also known as osteonectin or basement-membrane-40 (BM-40), is a member of a family of matricellular proteins, whose functions are to modulate cell-matrix interactions, growth and angiogenesis in colorectal cancer. In this study, the expression of SPARC was evaluated and its correlations with clinicopathological parameters were investigated. Methods The researchers analyzed the expression patterns of SPARC by using immunohistochemistry in 332 cases of colorectal cancer of tissue microarray. The clinicopathological characteristics were defined by using the TNM criteria of the Union for International Cancer Control. Clinicopathological factors such as age, sex, histologic type of the tumor, pathologic tumor stage, TNM stage, and lymphovascular invasion were evaluated according to the SPARC expression. Results The hazard ratios expressing SPARC in tumor cells, in the stroma, and in both tumor cells and the stroma were 2.10 (P = 0.036), 3.27 (P = 0.003) and 2.12 (P = 0.038), respectively. Patient survival was decreased in patient expressing SPARC in the stroma, and this result showed statistical significance (P = 0.016). Conclusion These findings suggest that SPARC expression in a tumor and in the stroma correlates with disease progression and may be used as a prognostic marker for colorectal cancer.
Collapse
Affiliation(s)
- Jeong Yeon Kim
- Department of Pathology, Soonchunhyang University College of Medicine, Cheonan, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Scatena R, Bottoni P, Giardina B. Circulating tumour cells and cancer stem cells: a role for proteomics in defining the interrelationships between function, phenotype and differentiation with potential clinical applications. Biochim Biophys Acta Rev Cancer 2012; 1835:129-43. [PMID: 23228700 DOI: 10.1016/j.bbcan.2012.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/29/2012] [Accepted: 12/01/2012] [Indexed: 01/22/2023]
Abstract
Research on the discovery and implementation of valid cancer biomarkers is one of the most challenging fields in oncology and oncoproteomics in particular. Moreover, it is generally accepted that an evaluation of cancer biomarkers from the blood could significantly enable biomarker assessments by providing a relatively non-invasive source of representative tumour material. In this regard, circulating tumour cells (CTCs) isolated from the blood of metastatic cancer patients have significant promise. It has been demonstrated that localised and metastatic cancers may give rise to CTCs, which are detectable in the bloodstream. Despite technical difficulties, recent studies have highlighted the prognostic significance of the presence and number of CTCs in the blood. Future studies are necessary not only to detect CTCs but also to characterise them. Furthermore, another pathogenically significant type of cancer cells, known as cancer stem cells (CSCs) or more recently termed circulating tumour stem cells (CTSCs), appears to have a significant role as a subpopulation of CTCs. This review discusses the potential application of proteomic methodologies to improve the isolation and characterisation of CTCs and to distinguish between CTCs with a poor clinical significance and those with important biological and clinical implications.
Collapse
|
11
|
Fresno C, Llera AS, Girotti MR, Valacco MP, López JA, Podhajcer OL, Balzarini MG, Prada F, Fernández EA. The multi-reference contrast method: Facilitating set enrichment analysis. Comput Biol Med 2012; 42:188-94. [DOI: 10.1016/j.compbiomed.2011.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 11/13/2011] [Accepted: 11/18/2011] [Indexed: 11/26/2022]
|
12
|
SPARC Promotes Cathepsin B-Mediated Melanoma Invasiveness through a Collagen I/α2β1 Integrin Axis. J Invest Dermatol 2011; 131:2438-47. [DOI: 10.1038/jid.2011.239] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
13
|
Rucevic M, Hixson D, Josic D. Mammalian plasma membrane proteins as potential biomarkers and drug targets. Electrophoresis 2011; 32:1549-64. [PMID: 21706493 DOI: 10.1002/elps.201100212] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining the plasma membrane proteome is crucial to understand the role of plasma membrane in fundamental biological processes. Change in membrane proteins is one of the first events that take place under pathological conditions, making plasma membrane proteins a likely source of potential disease biomarkers with prognostic or diagnostic potential. Membrane proteins are also potential targets for monoclonal antibodies and other drugs that block receptors or inhibit enzymes essential to the disease progress. Despite several advanced methods recently developed for the analysis of hydrophobic proteins and proteins with posttranslational modifications, integral membrane proteins are still under-represented in plasma membrane proteome. Recent advances in proteomic investigation of plasma membrane proteins, defining their roles as diagnostic and prognostic disease biomarkers and as target molecules in disease treatment, are presented.
Collapse
Affiliation(s)
- Marijana Rucevic
- COBRE Center for Cancer Research Development, Rhode Island Hospital, Providence, RI, USA
| | | | | |
Collapse
|
14
|
Atorrasagasti C, Aquino JB, Hofman L, Alaniz L, Malvicini M, Garcia M, Benedetti L, Friedman SL, Podhajcer O, Mazzolini G. SPARC downregulation attenuates the profibrogenic response of hepatic stellate cells induced by TGF-β1 and PDGF. Am J Physiol Gastrointest Liver Physiol 2011; 300:G739-48. [PMID: 21311029 PMCID: PMC3094149 DOI: 10.1152/ajpgi.00316.2010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver fibrosis is an active process that involves changes in cell-cell and cell-extracellular matrix (ECM) interaction. Secreted protein, acidic and rich in cysteine (SPARC) is an ECM protein with many biological functions that is overexpressed in cirrhotic livers and upregulated in activated hepatic stellate cells (aHSCs). We have recently shown that SPARC downregulation ameliorates liver fibrosis in vivo. To uncover the cellular mechanisms involved, we have specifically knocked down SPARC in two aHSC lines [the CFSC-2G (rat) and the LX-2 (human)] and in primary cultured rat aHSCs. Transient downregulation of SPARC in hepatic stellate cells (HSCs) did not affect their proliferation and had only minor effects on apoptosis. However, SPARC knockdown increased HSC adhesion to fibronectin and significantly decreased their migration toward PDFG-BB and TGF-β(1). Interestingly, TGF-β(1) secretion by HSCs was reduced following SPARC small interfering RNA (siRNA) treatment, and preincubation with TGF-β(1) restored the migratory capacity of SPARC siRNA-treated cells through mechanisms partially independent from TGF-β(1)-mediated induction of SPARC expression; thus SPARC knockdown seems to exert its effects on HSCs partially through modulation of TGF-β(1) expression levels. Importantly, collagen-I mRNA expression was reduced in SPARC siRNA-transfected HSCs. Consistent with previous results, SPARC knockdown in aHSCs was associated with altered F-actin expression patterns and deregulation of key ECM and cell adhesion molecules, i.e., downregulation of N-cadherin and upregulation of E-cadherin. Our data together suggest that the upregulation of SPARC previously reported for aHSCs partially mediates profibrogenic activities of TGF-β(1) and PDGF-BB and identify SPARC as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
| | - Jorge B. Aquino
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University; ,2CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas); and
| | - Leonardo Hofman
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University;
| | - Laura Alaniz
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University; ,2CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas); and
| | - Mariana Malvicini
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University;
| | - Mariana Garcia
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University; ,2CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas); and
| | - Lorena Benedetti
- 3Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina; and
| | - Scott L. Friedman
- 4Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York
| | - Osvaldo Podhajcer
- 2CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas); and ,3Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina; and
| | - Guillermo Mazzolini
- 1Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University; ,2CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas); and
| |
Collapse
|
15
|
Hardesty WM, Kelley MC, Mi D, Low RL, Caprioli RM. Protein signatures for survival and recurrence in metastatic melanoma. J Proteomics 2011; 74:1002-14. [PMID: 21549228 DOI: 10.1016/j.jprot.2011.04.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 04/11/2011] [Accepted: 04/15/2011] [Indexed: 12/11/2022]
Abstract
Patients with melanoma metastatic to regional lymph nodes exhibit a range in tumor progression, survival, and treatment. Current approaches to stratify patients with this stage of disease predominantly involve clinical and histological methods. Molecular classification thus far has focused almost exclusively on genetic mutations. In this study, proteomic data from 69 melanoma lymph node metastases and 17 disease free lymph nodes acquired by histology-directed MALDI imaging mass spectrometry were used to classify tumor from control lymph node and to molecularly sub-classify patients with stage III disease. From these data, 12 survival associated protein signals and 3 recurrence associated signals in the acquired mass spectra were combined to generate a multiplex molecular signature to group patients into either poor or favorable groups for recurrence and survival. Proteins represented in the signature include cytochrome c, s100 A6, histone H4, and cleaved forms of thymosin β-4, thymosin β-10, and ubiquitin. In total over 40 protein signals from the tissue were identified.
Collapse
|
16
|
The interplay of the EIIA(Ntr) component of the nitrogen-related phosphotransferase system (PTS(Ntr)) of Pseudomonas putida with pyruvate dehydrogenase. Biochim Biophys Acta Gen Subj 2011; 1810:995-1005. [PMID: 21236318 DOI: 10.1016/j.bbagen.2011.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 11/20/2022]
Abstract
BACKGROUND Pseudomonas putida KT2440 is endowed with a variant of the phosphoenolpyruvate-carbohydrate phosphotransferase system (PTS(Ntr)), which is not related to sugar transport but believed to rule the metabolic balance of carbon vs. nitrogen. The metabolic targets of such a system are largely unknown. METHODS Dielectric breakdown of P. putida cells grown in rich medium revealed the presence of forms of the EIIA(Ntr) (PtsN) component of PTS(Ntr), which were strongly associated to other cytoplasmic proteins. To investigate such intracellular partners of EIIA(Ntr), a soluble protein extract of bacteria bearing an E epitope tagged version of PtsN was immunoprecipitated with a monoclonal anti-E antibody and the pulled-down proteins identified by mass spectrometry. RESULTS The E1 subunit of the pyruvate dehydrogenase (PDH) complex, the product of the aceE gene, was identified as a major interaction partner of EIIA(Ntr). To examine the effect of EIIA(Ntr) on PDH, the enzyme activity was measured in extracts of isogenic ptsN(+)/ptsN(-)P. putida strains and the role of phosphorylation was determined. Expression of PtsN and AceE proteins fused to different fluorescent moieties and confocal laser microscopy indicated a significant co-localization of the two proteins in the bacterial cytoplasm. CONCLUSION EIIA(Ntr) down-regulates PDH activity. Both genetic and biochemical evidence revealed that the non-phosphorylated form of PtsN is the protein species that inhibits PDH. GENERAL SIGNIFICANCE EIIA(Ntr) takes part in the node of C metabolism that checks the flux of carbon from carbohydrates into the Krebs cycle by means of direct protein-protein interactions with AceE. This type of control might connect metabolism to many other cellular functions. This article is part of a Special Issue entitled: Systems Biology of Microorganisms.
Collapse
|
17
|
Makridakis M, Roubelakis MG, Bitsika V, Dimuccio V, Samiotaki M, Kossida S, Panayotou G, Coleman J, Candiano G, Anagnou NP, Vlahou A. Analysis of Secreted Proteins for the Study of Bladder Cancer Cell Aggressiveness. J Proteome Res 2010; 9:3243-59. [DOI: 10.1021/pr100189d] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Manousos Makridakis
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Maria G. Roubelakis
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Vasiliki Bitsika
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Veronica Dimuccio
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Martina Samiotaki
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Sophia Kossida
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - George Panayotou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Jonathan Coleman
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Giovanni Candiano
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Nikolaos P. Anagnou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory of Cell and Gene Therapy, Biomedical Research Foundation, Academy of Athens, Greece, Laboratory on Physiopathology of Uremia, G. Gaslini Children’s Hospital, Genoa, Italy, Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, and Laboratory of Biology, University of Athens School of Medicine
| |
Collapse
|
18
|
Zhou Y, Hofstetter WL, He Y, Hu W, Pataer A, Wang L, Wang J, Zhou Y, Yu L, Fang B, Swisher SG. KLF4 inhibition of lung cancer cell invasion by suppression of SPARC expression. Cancer Biol Ther 2010; 9:507-13. [PMID: 20215880 DOI: 10.4161/cbt.9.7.11106] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Krüppel-Like Factor 4 (KLF4) functions as a tumor suppressor in some cancers, but its molecular mechanism is not clear. Our recent study also showed that the expression of KLF4 is dramatically reduced in primary lung cancer tissues. To investigate the possible role of KLF4 in lung cancer, we stably transfected KLF4 into cells from lung cancer cell lines H322 and A549 to determine the cells' invasion ability. Our results showed that ectopic expression of KLF4 extensively suppressed lung cancer cell invasion in Matrigel. This effect was independent of KLF4-mediated p21 up-regulation because ectopic expression of p21 had minimal effect on cell invasion. Our analysis of the expression of 12 genes associated with cell invasion in parental, vector-transfected, and KLF4-transfected cells showed that ectopic expression of KLF4 resulted in extensively repressed expression of secreted protein acidic and rich in cysteine (SPARC), an extracellular matrix protein that plays a role in tumor development and metastasis. Knockdown of SPARC expression in H322 and A549 cells led to suppression of cell invasion, comparable to that observed in KLF4-transfected cells. Moreover, retrovirus-mediated restoration of SPARC expression in KLF4-transfected cells abrogated KLF4-induced anti-invasion activity. Together, our results indicate that KLF4 inhibits lung cancer cell invasion by suppressing SPARC gene expression.
Collapse
Affiliation(s)
- Yanbin Zhou
- Department of Pulmonary Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Llera AS, Girotti MR, Benedetti LG, Podhajcer OL. Matricellular proteins and inflammatory cells: A task force to promote or defeat cancer? Cytokine Growth Factor Rev 2010; 21:67-76. [DOI: 10.1016/j.cytogfr.2009.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
20
|
Chlenski A, Cohn SL. Modulation of matrix remodeling by SPARC in neoplastic progression. Semin Cell Dev Biol 2009; 21:55-65. [PMID: 19958839 DOI: 10.1016/j.semcdb.2009.11.018] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 11/20/2009] [Indexed: 01/10/2023]
Abstract
SPARC is a matricellular glycoprotein that mediates interactions between cells and their microenvironment. It is produced at sites of tissue remodeling, where it regulates matrix deposition and turnover, cell adhesion, and signaling by extracellular factors, exerting profound effects on tissue architecture and cell physiology. During extensive matrix remodeling in neoplastic progression, SPARC is expressed in cancer-associated stroma and in malignant cells of some types, affecting tumor development, invasion, metastases, angiogenesis and inflammation. SPARC-induced changes in the tumor microenvironment can suppress or promote progression of different cancers depending on the tissue and cell type. Understanding the mechanism of matrix remodeling and its regulation by SPARC is essential for the development of new treatment strategies for highly aggressive cancers.
Collapse
Affiliation(s)
- Alexandre Chlenski
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, United States.
| | | |
Collapse
|
21
|
Arnold SA, Brekken RA. SPARC: a matricellular regulator of tumorigenesis. J Cell Commun Signal 2009; 3:255-73. [PMID: 19809893 PMCID: PMC2778590 DOI: 10.1007/s12079-009-0072-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 09/14/2009] [Indexed: 12/11/2022] Open
Abstract
Although many clinical studies have found a correlation of SPARC expression with malignant progression and patient survival, the mechanisms for SPARC function in tumorigenesis and metastasis remain elusive. The activity of SPARC is context- and cell-type-dependent, which is highlighted by the fact that SPARC has shown seemingly contradictory effects on tumor progression in both clinical correlative studies and in animal models. The capacity of SPARC to dictate tumorigenic phenotype has been attributed to its effects on the bioavailability and signaling of integrins and growth factors/chemokines. These molecular pathways contribute to many physiological events affecting malignant progression, including extracellular matrix remodeling, angiogenesis, immune modulation and metastasis. Given that SPARC is credited with such varied activities, this review presents a comprehensive account of the divergent effects of SPARC in human cancers and mouse models, as well as a description of the potential mechanisms by which SPARC mediates these effects. We aim to provide insight into how a matricellular protein such as SPARC might generate paradoxical, yet relevant, tumor outcomes in order to unify an apparently incongruent collection of scientific literature.
Collapse
Affiliation(s)
- Shanna A Arnold
- Hamon Center for Therapeutic Oncology Research, Division of Surgical Oncology and Departments of Surgery and Pharmacology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-8593 USA
| | | |
Collapse
|
22
|
Bottoni P, Giardina B, Scatena R. Proteomic profiling of heat shock proteins: An emerging molecular approach with direct pathophysiological and clinical implications. Proteomics Clin Appl 2009; 3:636-53. [DOI: 10.1002/prca.200800195] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Podhajcer OL, Benedetti LG, Girotti MR, Prada F, Salvatierra E, Llera AS. The role of the matricellular protein SPARC in the dynamic interaction between the tumor and the host. Cancer Metastasis Rev 2008; 27:691-705. [PMID: 18542844 DOI: 10.1007/s10555-008-9146-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Tumor growth is essentially the result of an evolving cross-talk between malignant and surrounding stromal cells (fibroblasts, endothelial cells and inflammatory cells). This heterogeneous mass of extracellular matrix and intermingled cells interact through cell-cell and cell-matrix contacts. Malignant cells also secrete soluble proteins that reach neighbor stromal cells, forcing them to provide the soil on which they will grow and metastasize. Different studies including expression array analysis identified the matricellular protein SPARC as a marker of poor prognosis in different cancer types. Further evidence demonstrated that high SPARC levels are often associated with the most aggressive and highly metastatic tumors. Here we describe the most recent evidence that links SPARC with human cancer progression, the controversy regarding its role in certain human cancers and the physiological processes in which SPARC is involved: epithelial-mesenchymal transition, immune surveillance and angiogenesis. Its relevance as a potential target in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Osvaldo L Podhajcer
- Laboratory of Molecular and Cellular Therapy, Fundacion Instituto Leloir, University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
24
|
Fernández EA, Girotti MR, López del Olmo JA, Llera AS, Podhajcer OL, Cantet RJC, Balzarini M. Improving 2D-DIGE protein expression analysis by two-stage linear mixed models: assessing experimental effects in a melanoma cell study. ACTA ACUST UNITED AC 2008; 24:2706-12. [PMID: 18818217 DOI: 10.1093/bioinformatics/btn508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
MOTIVATION Difference in-gel electrophoresis (DIGE)-based protein expression analysis allows assessing the relative expression of proteins in two biological samples differently labeled (Cy5, Cy3 CyDyes). In the same gel, a reference sample is also used (Cy2 CyDye) for spot matching during image analysis and volume normalization. The standard statistical techniques to identify differentially expressed (DE) proteins are the calculation of fold-changes and the comparison of treatment means by the t-test. The analyses rarely accounts for other experimental effects, such as CyDye and gel effects, which could be important sources of noise while detecting treatment effects. RESULTS We propose to identify DIGE DE proteins using a two-stage linear mixed model. The proposal consists of splitting the overall model for the measured intensity into two interconnected models. First, we fit a normalization model that accounts for the general experimental effects, such as gel and CyDye effects as well as for the features of the associated random term distributions. Second, we fit a model that uses the residuals from the first step to account for differences between treatments in protein-by-protein basis. The modeling strategy was evaluated using data from a melanoma cell study. We found that a heteroskedastic model in the first stage, which also account for CyDye and gel effects, best normalized the data, while allowing for an efficient estimation of the treatment effects. The Cy2 reference channel was used as a covariate in the normalization model to avoid skewness of the residual distribution. Its inclusion improved the detection of DE proteins in the second stage.
Collapse
Affiliation(s)
- Elmer A Fernández
- School of Engineering, Intelligent Data Analysis Group, Catholic University of Córdoba, Argentina.
| | | | | | | | | | | | | |
Collapse
|
25
|
Podhajcer OL, Benedetti L, Girotti MR, Prada F, Salvatierra E, Llera AS. The role of the matricellular protein SPARC in the dynamic interaction between the tumor and the host. Cancer Metastasis Rev 2008; 27:523-37. [PMID: 18459035 DOI: 10.1007/s10555-008-9135-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Tumor growth is essentially the result of an evolving cross-talk between malignant and surrounding stromal cells (fibroblasts, endothelial cells and inflammatory cells). This heterogeneous mass of extracellular matrix and intermingled cells interact through cell-cell and cell-matrix contacts. Malignant cells also secrete soluble proteins that reach neighbor stromal cells, forcing them to provide the soil on which they will grow and metastasize. Different studies including expression array analysis identified the matricellular protein SPARC as a marker of poor prognosis in different cancer types. Further evidence demonstrated that high SPARC levels are often associated with the most aggressive and highly metastatic tumors. Here we describe the most recent evidence that links SPARC with human cancer progression, the controversy regarding its role in certain human cancers and the physiological processes in which SPARC is involved: epithelial-mesenchymal transition, immune surveillance and angiogenesis. Its relevance as a potential target in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Osvaldo L Podhajcer
- Laboratory of Molecular and Cellular Therapy, Fundacion Instituto Leloir, University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|