1
|
Bialek J, Yankulov S, Kawan F, Fornara P, Theil G. Role of Nivolumab in the Modulation of PD-1 and PD-L1 Expression in Papillary and Clear Cell Renal Carcinoma (RCC). Biomedicines 2022; 10:biomedicines10123244. [PMID: 36552000 PMCID: PMC9776360 DOI: 10.3390/biomedicines10123244] [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: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
The expression and cellular mechanisms of programmed cell death-1 protein (PD-1) and its ligands (PD-L1 and PD-L2) in renal cancer cells are not well known. Here, we aimed to investigate the response of renal carcinoma subtypes to the immune checkpoint inhibitor nivolumab and its impact on related signaling pathways. All cell lines analyzed (clear cell (cc)RCC (Caki-1, RCC31) and papillary (p)RCC (ACHN, RCC30)) expressed PD-1 and both ccRCC cell lines, and RCC30 expressed PD-L1. Nivolumab treatment at increasing doses led to increased PD-1 levels in analyzed cells and resulted in aggressive behavior of pRCC but diminished this behavior in ccRCC. The analysis of PD-1/PD-L1-associated signaling pathways demonstrated increased AKT activity in Caki-1 and RCC30 cells but decreased activity in ACHN and RCC31 cells, while ribosomal protein S6 remained largely unchanged. Androgen receptors are related to RCC and were predominantly increased in RCC30 cells, which were the only cells that formed nivolumab-dependent spheroids. Finally, all cell lines exhibited a complex response to nivolumab treatment. Since the pRCC cells responded with increased tumorigenicity and PD-1/PD-L1 levels while ccRCC tumorigenicity was diminished, further studies are needed to improve nivolumab-based therapy for renal carcinoma subtypes, especially the identification of response-involved molecular pathways.
Collapse
|
2
|
Molecular Protein and Expression Profile in the Primary Tumors of Clear Cell Renal Carcinoma and Metastases. Cells 2020; 9:cells9071680. [PMID: 32668608 PMCID: PMC7408457 DOI: 10.3390/cells9071680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022] Open
Abstract
Metastasis involves the spread of cancer cells from the primary tumor to surrounding tissues and distant organs and is the primary cause of cancer morbidity and mortality. The aim of the study was the determination of change in molecular factors expression in primary kidney cancers (ccRCC) and metastatic sites. In total, 62 patients with RCC were enrolled in the study. The mRNA levels of molecular markers were studied by real-time PCR, and the content of the studied parameters was determined by Western blotting and ELISA. The features in the intracellular signal metabolites in the series of normal renal parenchyma, tumor tissue of localized, disseminated kidney cancer and metastatic tissue were studied. A decrease in some indicators in the tissue of the metastatic lesion was noted. Protein products of transcription factors HIF-1, CAIX, PTEN and activated AKT kinase, as well as expression of the VEGFR2 receptor and m-TOR protein kinase were revealed to be reduced in the metastatic sites. In addition, some indicators increased in metastasis: the protein levels of NF-κB p 50, NF-κB p 65, HIF-2, VEGF, VEGFR2, m-TOR and mRNA of HIF-1, CAIX, PTEN and PDK. There were indicators with multidirectional changes. HIF-1, CAIX, PTEN, VEGFR2 and m-TOR mRNA: VEGFR2, m-TOR, HIF-1, CAIX, PTEN and PDK had an opposite change in protein content and mRNA level. PTEN loss resulted in the downstream activation of AKT/mTOR signaling in secondary cancer lesions and determined the overall ccRCC patient's survival. The AKT/mTOR signaling cascade activation was found in the primary kidney tumors. The PTEN content and mRNA level were correlated with total AKT, GSK-3β, the 70S 6 kinases and AKT expression.
Collapse
|
3
|
Tumkur Sitaram R, Landström M, Roos G, Ljungberg B. Significance of PI3K signalling pathway in clear cell renal cell carcinoma in relation to VHL and HIF status. J Clin Pathol 2020; 74:216-222. [PMID: 32467322 DOI: 10.1136/jclinpath-2020-206693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/10/2020] [Indexed: 12/30/2022]
Abstract
Renal cell carcinoma (RCC) includes diverse tumour types characterised by various genetic abnormalities. The genetic changes, like mutations, deletions and epigenetic alterations, play a crucial role in the modification of signalling networks, tumour pathogenesis and prognosis. The most prevalent RCC type, clear cell RCC (ccRCC), is asymptomatic in the early stages and has a poorer prognosis compared with the papillary and the chromophobe types RCCs. Generally, ccRCC is refractory to chemotherapy and radiation therapy. Loss of von Hippel-Lindau (VHL) gene and upregulation of hypoxia-inducible factors (HIF), the signature of most sporadic ccRCC, promote multiple growth factors. Hence, VHL/HIF and a variety of pathways, including phosphatase and TEnsin homolog on chromosome 10/phosphatidylinositol-3-kinase (PI3K)/AKT, are closely connected and contribute to the ontogeny of ccRCC. In the recent decade, multiple targeting agents have been developed based on blocking major signalling pathways directly or indirectly involved in ccRCC tumour progression, metastasis, angiogenesis and survival. However, most of these drugs have limitations; either metastatic ccRCC develops resistance to these agents, or despite blocking receptors, tumour cells use alternate signalling pathways. This review compiles the state of knowledge about the PI3K/AKT signalling pathway confined to ccRCC and its cross-talks with VHL/HIF pathway.
Collapse
Affiliation(s)
- Raviprakash Tumkur Sitaram
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Maréne Landström
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Göran Roos
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Börje Ljungberg
- Department of Surgical and Preoperative Sciences, Urology and Andrology, Umeå Universitet, Umea, Västerbotten, Sweden
| |
Collapse
|
4
|
Rausch S, Schollenberger D, Hennenlotter J, Stühler V, Kruck S, Stenzl A, Bedke J. mTOR and mTOR phosphorylation status in primary and metastatic renal cell carcinoma tissue: differential expression and clinical relevance. J Cancer Res Clin Oncol 2019; 145:153-163. [PMID: 30368665 DOI: 10.1007/s00432-018-2775-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/19/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Impaired regulation of the Akt/mammalian target of rapamycin (mTOR) pathway has been implicated in mechanisms related to neoplastic transformation in renal cell cancer (RCC) through enhancement of cell proliferation and survival and mTOR activation has been reported to occur due to phosphorylation of mTOR. To further determine the relevance of mTOR expression and activation and to analyze their putative role as a biomarker for systemic treatment in metastatic RCC, we investigated the expression of mTOR and phospho(p)-mTOR in primary RCC and metastases and correlated levels with pathological variables and clinical outcome. METHODS Tissue microarrays (TMA) from paraffin-embedded tissue from 342 patients with primary clear cell renal cell carcinoma and 90 patients undergoing surgical resection for metastases were immunohistochemically stained for mTOR and p-mTOR and expression was quantified with immunoreactivity scores. Clinical patient characteristics and follow-up were recorded. Comparative evaluation of protein expression levels and association of expression with clinical variables and survival was performed. RESULTS mTOR staining revealed differential expression in benign, primary and RCC metastasis (average staining score: 1.64, 0.78, and 1.44, respectively). Average staining of p-mTOR was 0.99 in benign kidney tissue, 0.73 in primary RCC and 1.14 in RCC metastasis tissue. Elevated mTOR expression in primary RCC tissue was associated with the presence of tumor necrosis, while a high level of p-mTOR was significantly correlated with advanced T-stage, high Fuhrman grade, the presence of tumor necrosis and sarcomatoid features. An elevated ratio of p-mTOR/mTOR was significantly correlated with advanced stage and sarcomatoid histology. mTOR expression was not predictive of overall survival (OS), while high p-mTOR levels were associated with impaired OS (p = 0.0046) and cancer-specific survival (p = 0.0067). In univariate analysis, advanced stage (HR 3.78), high Fuhrman grade (HR 4.0), the presence of tumor necrosis (HR 1.99), and sarcomatoid features (HR 5.12) were significant predictors of OS. Moreover, elevated levels of p-mTOR (HR 1.67) and an elevated ratio of p-mTOR/mTOR ratio (HR 1.73) were significantly predictive of OS. In the multivariate regression model only the presence of locally advanced tumors (HR 2.44) was of independent prognostic value for OS, while there was a trend for impaired OS for patients with a high p-mTOR (HR 1.27, p = 0.21). CONCLUSIONS Phosphorylated mTOR is differentially expressed in localized RCC and metastasis. Elevated phosphorylation of mTOR is associated with aggressive pathologic features and unfavorable outcome. Whether these findings portend to relevance for mTOR inhibition treatment for metastatic RCC should be objective of further investigations.
Collapse
Affiliation(s)
- Steffen Rausch
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Daniel Schollenberger
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Joerg Hennenlotter
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Viktoria Stühler
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Stephan Kruck
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| |
Collapse
|
5
|
Spirina LV, Kondakova IV, Tarasenko NV, Slonimskaya EM, Usynin EA, Gorbunov AK, Yurmazov ZA, Chigevskaya SY. [Targeting of the AKT/m-TOR Pathway: Biomarkers of Resistance to Cancer Therapy--
AKT/m-TOR Pathway and Resistance to Cancer Therapy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 21:63-66. [PMID: 29357975 PMCID: PMC5972354 DOI: 10.3779/j.issn.1009-3419.2018.01.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Resistance to cancer therapy continues to be a major limitation for the successful treatment of cancer. There are many published studies on therapy resistance in breast and prostate cancers; however, there are currently no data on molecular markers associated with resistance. The conflicting data were reported regarding the AKT/m-TOR signaling pathway components as markers predicting resistance. The AKT/m-TOR signaling pathway is involved in the development of many human cancers; its activation is related to cell proliferation, angiogenesis, apoptosis, as well as to therapy resistance. Molecular alterations in the AKT/m-TOR signaling pathway provide a platform to identify universal markers associated with the development of resistance to cancer therapy.
Collapse
Affiliation(s)
- Liudmila V Spirina
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia.,Department of Biochemistry and Molecular Biology, Siberian State Medical University, Tomsk, Russia
| | - Irina V Kondakova
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| | - Natalia V Tarasenko
- Department of Medical Genetics, Siberian State Medical University, Tomsk, Russia.,Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| | - Elena M Slonimskaya
- Surgical Department, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia.,Department of Oncology, Siberian State Medical University, Tomsk, Russia
| | - Evgeny A Usynin
- Surgical Department, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| | - Alexey K Gorbunov
- Surgical Department, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| | - Zahar A Yurmazov
- Surgical Department, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| | - Svetlana Yu Chigevskaya
- Surgical Department, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Russia
| |
Collapse
|
6
|
Zou Y, Wang J, Leng X, Huang J, Xue W, Zhang J, Huang Y. The selective MEK1 inhibitor Selumetinib enhances the antitumor activity of everolimus against renal cell carcinoma in vitro and in vivo. Oncotarget 2017; 8:20825-20833. [PMID: 28212559 PMCID: PMC5400548 DOI: 10.18632/oncotarget.15346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/27/2017] [Indexed: 02/07/2023] Open
Abstract
Renal cell carcinoma (RCC) is a urologic malignant cancer and often diagnosed at an advanced stage, which results in high mortality. Targeted therapy may improve the quality of life and survival of patients who are not suitable for nephrectomy. Everolimus, an mTOR inhibitor, is currently used as sequential or second-line therapy for RCC refractory to Sunitinib or sorafenib. However, its efficiency is palliative. In this study, we evaluated whether the antitumor activity of everolimus against RCC is enhanced by Selumetinib, a selective MEK1 inhibitor. We discovered that everolimus in combination with Selumetinib synergistically inhibited the proliferation of Caki-1, 786-O and 769-P cells in vitro. Mechanistically, this combination decreased p-RPS6 and p-4E-BP1 dramatically, which causes G1 cell cycle arrest and prevents reactivation of AKT and ERK. In vivo, the antitumor efficacy and pharmacodynamic biomarkers of the combination therapy were recapitulated in Caki-1 xenograft model. In addition, this combination treatment potently inhibited angiogenesis in xenograft models by impairing VEGF secretion from tumor cells. Our findings provide a sound evidence that combination of everolimus and Selumetinib is a potential dual-targeted strategy for renal cell carcinoma.
Collapse
Affiliation(s)
- Yun Zou
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianfeng Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuejiao Leng
- Department of Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiwei Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Kim SH, Park WS, Park EY, Park B, Joo J, Joung JY, Seo HK, Lee KH, Chung J. The prognostic value of BAP1, PBRM1, pS6, PTEN, TGase2, PD-L1, CA9, PSMA, and Ki-67 tissue markers in localized renal cell carcinoma: A retrospective study of tissue microarrays using immunohistochemistry. PLoS One 2017; 12:e0179610. [PMID: 28654655 PMCID: PMC5487017 DOI: 10.1371/journal.pone.0179610] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To assess the prognostic roles of BAP1, PBRM1, pS6, PTEN, TGase2, PD-L1, CA9, PSMA, and Ki-67 tissue biomarkers in localized renal cell carcinoma (RCC). METHODS Patients who underwent a nephrectomy during 1992-2015 and had a primary specimen of their kidney tumor were included. The nine tissue biomarkers were immunohistochemically stained on tissue microarrays of RCC, and the semi-quantitative H-score, including intensity score, was used to grade the sample. The Cox proportional hazards model was used to evaluate tissue markers significant for overall survival (OS), cancer-specific survival (CSS), and recurrence-free survival (RFS) after adjusting for significant clinicopathological parameters. RESULTS Samples from 351 RCC patients were included. The mean age of the patients was 53.9 years; the rates of pathologic T1-2/≥T3 stage, Fuhrman 1+2/3+4 grade, recurrence, and death were 269/65(80.5/19.5%), 222/107 (67.5/32.5%), 6.6%, and 10.5%, respectively. Median OS, CSS, and RFS were 220.6, 220.6, and 147.1 months, respectively. The multivariable analysis showed that pathologic T stage and Fuhrman nuclear grade were significantly associated with OS and CSS. Pathologic T stage and tumor size were associated with RFS. After adjusting for these significant prognostic clinicopathological factors, Ki-67 was significantly associated with OS (hazard ratio [HR], 2.7), CSS (HR, 3.82), and RFS (HR, 4.85) and pS6 was associated with CSS (HR, 8.63) and RFS (HR, 8.51) in the multivariable model (p<0.05). CONCLUSION pS6 and Ki-67 are significant prognostic factors of RCC; however, BAP1, PBRM1, TGase 2, PD-L1, CA9, PTEN loss, and PSMA markers did not show this association.
Collapse
Affiliation(s)
- Sung Han Kim
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Weon Seo Park
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
- Department of Pathology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Eun Young Park
- Biometrics Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Boram Park
- Biometrics Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jungnam Joo
- Biometrics Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jae Young Joung
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Ho Kyung Seo
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Kang Hyun Lee
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| |
Collapse
|
8
|
Goh WWB. Fuzzy-FishNET: a highly reproducible protein complex-based approach for feature selection in comparative proteomics. BMC Med Genomics 2016; 9:67. [PMID: 28117654 PMCID: PMC5260792 DOI: 10.1186/s12920-016-0228-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background The hypergeometric enrichment analysis approach typically fares poorly in feature-selection stability due to its upstream reliance on the t-test to generate differential protein lists before testing for enrichment on a protein complex, subnetwork or gene group. Methods Swapping the t-test in favour of a fuzzy rank-based weight system similar to that used in network-based methods like Quantitative Proteomics Signature Profiling (QPSP), Fuzzy SubNets (FSNET) and paired FSNET (PFSNET) produces dramatic improvements. Results This approach, Fuzzy-FishNET, exhibits high precision-recall over three sets of simulated data (with simulated protein complexes) while excelling in feature-selection reproducibility on real data (based on evaluation with real protein complexes). Overlap comparisons with PFSNET shows Fuzzy-FishNET selects the most significant complexes, which are also strongly class-discriminative. Cross-validation further demonstrates Fuzzy-FishNET selects class-relevant protein complexes. Conclusions Based on evaluation with simulated and real datasets, Fuzzy-FishNET is a significant upgrade of the traditional hypergeometric enrichment approach and a powerful new entrant amongst comparative proteomics analysis methods. Electronic supplementary material The online version of this article (doi:10.1186/s12920-016-0228-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wilson Wen Bin Goh
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People's Republic of China.
| |
Collapse
|
9
|
Bin Goh WW, Guo T, Aebersold R, Wong L. Quantitative proteomics signature profiling based on network contextualization. Biol Direct 2015; 10:71. [PMID: 26666224 PMCID: PMC4678536 DOI: 10.1186/s13062-015-0098-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/30/2015] [Indexed: 12/02/2022] Open
Abstract
Background We present a network-based method, namely quantitative proteomic signature profiling (qPSP) that improves the biological content of proteomic data by converting protein expressions into hit-rates in protein complexes. Results We demonstrate, using two clinical proteomics datasets, that qPSP produces robust discrimination between phenotype classes (e.g. normal vs. disease) and uncovers phenotype-relevant protein complexes. Regardless of acquisition paradigm, comparisons of qPSP against conventional methods (e.g. t-test or hypergeometric test) demonstrate that it produces more stable and consistent predictions, even at small sample size. We show that qPSP is theoretically robust to noise, and that this robustness to noise is also observable in practice. Comparative analysis of hit-rates and protein expressions in significant complexes reveals that hit-rates are a useful means of summarizing differential behavior in a complex-specific manner. Conclusions Given qPSP’s ability to discriminate phenotype classes even at small sample sizes, high robustness to noise, and better summary statistics, it can be deployed towards analysis of highly heterogeneous clinical proteomics data. Reviewers This article was reviewed by Frank Eisenhaber and Sebastian Maurer-Stroh. Open peer review Reviewed by Frank Eisenhaber and Sebastian Maurer-Stroh. Electronic supplementary material The online version of this article (doi:10.1186/s13062-015-0098-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wilson Wen Bin Goh
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin City, 300072, China. .,Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Boston, USA. .,Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland. .,School of Computing, National University of Singapore, Singapore, Singapore.
| | - Tiannan Guo
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland. .,Faculty of Science, University of Zurich, Zurich, Switzerland.
| | - Limsoon Wong
- School of Computing, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
van Dijk M, Visser A, Buabeng KML, Poutsma A, van der Schors RC, Oudejans CBM. Mutations within the LINC-HELLP non-coding RNA differentially bind ribosomal and RNA splicing complexes and negatively affect trophoblast differentiation. Hum Mol Genet 2015; 24:5475-85. [PMID: 26173455 DOI: 10.1093/hmg/ddv274] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/09/2015] [Indexed: 11/14/2022] Open
Abstract
LINC-HELLP, showing chromosomal linkage with the pregnancy-specific HELLP syndrome in Dutch families, reduces differentiation from a proliferative to an invasive phenotype of first-trimester extravillous trophoblasts. Here we show that mutations in LINC-HELLP identified in HELLP families negatively affect this trophoblast differentiation either by inducing proliferation rate or by causing cell cycle exit as shown by a decrease in both proliferation and invasion. As LincRNAs predominantly function through interactions with proteins, we identified the directly interacting proteins using chromatin isolation by RNA purification followed by protein mass spectrometry. We found 22 proteins predominantly clustering in two functional networks, i.e. RNA splicing and the ribosome. YBX1, PCBP1, PCBP2, RPS6 and RPL7 were validated, and binding to these proteins was influenced by the HELLP mutations carried. Finally, we show that the LINC-HELLP transcript levels are significantly upregulated in plasma of women in their first trimester of pregnancy compared with non-pregnant women, whereas this upregulation seems absent in a pilot set of patients later developing pregnancy complications, indicative of its functional significance in vivo.
Collapse
Affiliation(s)
- Marie van Dijk
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands and
| | - Allerdien Visser
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands and
| | - Kwadwo M L Buabeng
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands and
| | - Ankie Poutsma
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands and
| | - Roel C van der Schors
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Cees B M Oudejans
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands and
| |
Collapse
|
11
|
Graves A, Hessamodini H, Wong G, Lim WH. Metastatic renal cell carcinoma: update on epidemiology, genetics, and therapeutic modalities. Immunotargets Ther 2013; 2:73-90. [PMID: 27471690 PMCID: PMC4928369 DOI: 10.2147/itt.s31426] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The treatment of advanced renal cell carcinoma (RCC) remains a major therapeutic challenge for clinicians. Despite advances in the understanding of the immunobiology of RCC and the availability of several novel targeted agents, there has been little improvement in the survival of patients with metastatic RCC. This review will focus on the recent understanding of risk factors and treatment options and outcomes of metastatic RCC, in particular, targeted therapeutic agents that inhibit vascular endothelial growth factor and mammalian target of rapamycin pathways. Prospective studies are required to determine whether sequential targeted therapy will further improve progression-free survival in RCC. Ongoing research to develop novel agents with better tolerability and enhanced efficacy in the treatment of metastatic RCC is required.
Collapse
Affiliation(s)
- Angela Graves
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Hannah Hessamodini
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Germaine Wong
- Centre for Kidney Research, University of Sydney, Sydney, NSW, Australia
| | - Wai H Lim
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia
| |
Collapse
|