Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cells ; 13(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38891116

ABSTRACT

Polycystic kidney disease (PKD) is characterized by extensive cyst formation and progressive fibrosis. However, the molecular mechanisms whereby the loss/loss-of-function of Polycystin 1 or 2 (PC1/2) provokes fibrosis are largely unknown. The small GTPase RhoA has been recently implicated in cystogenesis, and we identified the RhoA/cytoskeleton/myocardin-related transcription factor (MRTF) pathway as an emerging mediator of epithelium-induced fibrogenesis. Therefore, we hypothesized that MRTF is activated by PC1/2 loss and plays a critical role in the fibrogenic reprogramming of the epithelium. The loss of PC1 or PC2, induced by siRNA in vitro, activated RhoA and caused cytoskeletal remodeling and robust nuclear MRTF translocation and overexpression. These phenomena were also manifested in PKD1 (RC/RC) and PKD2 (WS25/-) mice, with MRTF translocation and overexpression occurring predominantly in dilated tubules and the cyst-lining epithelium, respectively. In epithelial cells, a large cohort of PC1/PC2 downregulation-induced genes was MRTF-dependent, including cytoskeletal, integrin-related, and matricellular/fibrogenic proteins. Epithelial MRTF was necessary for the paracrine priming of the fibroblast-myofibroblast transition. Thus, MRTF acts as a prime inducer of epithelial fibrogenesis in PKD. We propose that RhoA is a common upstream inducer of both histological hallmarks of PKD: cystogenesis and fibrosis.


Subject(s)
Epithelial Cells , Polycystic Kidney Diseases , TRPP Cation Channels , rhoA GTP-Binding Protein , Animals , Humans , Mice , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Mice, Inbred C57BL , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/genetics , rhoA GTP-Binding Protein/metabolism , Trans-Activators/metabolism , TRPP Cation Channels/metabolism , TRPP Cation Channels/genetics
2.
J Clin Med ; 11(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35054064

ABSTRACT

With sunitinib treatment of metastatic renal cell carcinoma, most patients end up developing resistance over time. Recent clinical trials have shown that individualizing treatment protocols could delay resistance and result in better outcomes. We developed an in vivo xenograft tumor model and compared tumor growth rate, morphological, and transcriptomic differences between alternative and traditional treatment schedules. Our results show that the alternative treatment regime could delay/postpone cancer progression. Additionally, we identified distinct morphological changes in the tumor with alternative and traditional treatments, likely due to the significantly dysregulated signaling pathways between the protocols. Further investigation of the signaling pathways underlying these morphological changes may lead potential therapeutic targets to be used in a combined treatment with sunitinib, which offers promise in postponing/reversing the resistance of sunitinib.

3.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34204945

ABSTRACT

A lesser known but crucially important downstream effect of Rho family GTPases is the regulation of gene expression. This major role is mediated via the cytoskeleton, the organization of which dictates the nucleocytoplasmic shuttling of a set of transcription factors. Central among these is myocardin-related transcription factor (MRTF), which upon actin polymerization translocates to the nucleus and binds to its cognate partner, serum response factor (SRF). The MRTF/SRF complex then drives a large cohort of genes involved in cytoskeleton remodeling, contractility, extracellular matrix organization and many other processes. Accordingly, MRTF, activated by a variety of mechanical and chemical stimuli, affects a plethora of functions with physiological and pathological relevance. These include cell motility, development, metabolism and thus metastasis formation, inflammatory responses and-predominantly-organ fibrosis. The aim of this review is twofold: to provide an up-to-date summary about the basic biology and regulation of this versatile transcriptional coactivator; and to highlight its principal involvement in the pathobiology of kidney disease. Acting through both direct transcriptional and epigenetic mechanisms, MRTF plays a key (yet not fully appreciated) role in the induction of a profibrotic epithelial phenotype (PEP) as well as in fibroblast-myofibroblast transition, prime pathomechanisms in chronic kidney disease and renal fibrosis.


Subject(s)
Kidney Diseases/genetics , Multiprotein Complexes/genetics , Serum Response Factor/genetics , Trans-Activators/genetics , Cell Movement/genetics , Cell Nucleus/genetics , Cytoskeleton/genetics , Gene Expression Regulation/genetics , Humans , Kidney Diseases/pathology , Promoter Regions, Genetic/genetics
4.
Am J Pathol ; 189(10): 2046-2060, 2019 10.
Article in English | MEDLINE | ID: mdl-31381885

ABSTRACT

Papillary renal cell carcinoma (PRCC) is the most common type of RCC in end-stage kidney disease (ESKD). Papillary adenoma (PA) is a small benign lesion morphologically similar to PRCC and is suggested to be its precursor. PA is also prevalent in ESKD. The evolution of PAs to PRCCs and their relationship to ESKD are poorly understood. A total of 140 PAs, normal kidneys, ESKDs, and PRCCs were analyzed. Previously described markers of renal tubular progenitor cells were analyzed using immunohistochemistry and quantified with digital analysis. Progenitor cells were significantly increased in ESKD (P < 0.0001) and PAs (P = 0.02) in comparison with the normal kidney. Pathway analysis using global miRNA and chromosomal copy number variations revealed a common developmental theme between PA and the PRCCs. Whole exome sequencing showed a KMT2C-specific pathogenic mutation among all PAs and PRCCs. KMT2C is a chromosome 7 epigenetic regulator implicated in development and oncogenesis. Collectively, results show possible connection of PRCCs to PA and the progenitor-like cell population, which are increased in response to renal tubular injury. In addition, each PRCC histologic subtype had its own set of mutational changes, indicating divergence from a common precursor. The study reports previously unknown biological aspects of PRCC development and could influence current surveillance criteria and early detection strategies of PRCC tumors.


Subject(s)
Adenoma/pathology , Biomarkers, Tumor/genetics , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Kidney Failure, Chronic/pathology , Kidney/pathology , Stem Cells/pathology , Adenoma/genetics , Carcinoma, Papillary/genetics , Carcinoma, Renal Cell/genetics , Case-Control Studies , Cells, Cultured , Chromosome Aberrations , Cohort Studies , DNA Copy Number Variations , Humans , Kidney/metabolism , Kidney Failure, Chronic/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Prognosis , Stem Cells/metabolism
5.
Eur Urol Focus ; 5(2): 250-261, 2019 03.
Article in English | MEDLINE | ID: mdl-28847686

ABSTRACT

CONTEXT: Regenerative medicine has recently presented a revolutionary solution to end-stage kidney disease. Reprogramming patients' own cells generates induced pluripotent stem cells that are subsequently differentiated to "kidney organoid," a structure that is anatomically and functionally similar to the kidney. This approach holds the promise of a transplantable, immunocompetent, and functional kidney that could be produced in vitro. However, caution must be taken due to the molecular-level similarities between induced pluripotent stem cells and renal cell carcinomas. As such, if cell reprogramming is not tightly controlled, it can lead to carcinogenic changes. OBJECTIVE: Based on recent next-generation sequencing results and other supporting data, we identified three major molecular attributes of renal cell carcinoma: metabolic alterations, epigenetic changes, and miRNA-based alterations. Strikingly, these variations are mirrored in induced pluripotent stem cells, which are the main cell source of renal regenerative medicine. Our objective was to discuss the shared metabolic, epigenetic and miRNA-regulated characteristics and to abridge their significance in renal regenerative medicine. EVIDENCE ACQUISITION: English-language literature was retrieved through PubMed. EVIDENCE SYNTHESIS: Authors collected the published evidence and evaluated the content based on independent literature findings. Articles were filtered to include only highly relevant, recent publications that presented reproducible results by authorities of the field. CONCLUSIONS: The kidney represents a unique metabolic environment that could be hijacked by induced pluripotent stem cells or by partially differentiated cells for oncogenic transformation. Future differentiation protocols must produce kidney organoids that are fully engaged in filtration function. PATIENT SUMMARY: A new technology can produce mini-kidneys or kidney organoids. This review discusses some of the challenges this technology has to face, including its high oncogenic potential. Understanding these similarities will lead to the safe creation of new functional kidney units in patients with kidney failure.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/transplantation , Kidney Neoplasms/genetics , Regenerative Medicine/methods , Carcinogenesis/pathology , Cell Differentiation/genetics , Clinical Trials as Topic , Epigenesis, Genetic , Epigenomics , Glucose/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Kidney Failure, Chronic/genetics , Kidney Neoplasms/pathology , MicroRNAs/genetics , Organoids/cytology , Organoids/metabolism
6.
FASEB J ; 33(1): 1347-1359, 2019 01.
Article in English | MEDLINE | ID: mdl-30148679

ABSTRACT

Sunitinib is the standard-of-care, first-line treatment for advanced renal cell carcinoma (RCC). Characteristics of treatment-resistant RCC have been described; however, complex tumor adaptation mechanisms obstruct the identification of significant operators in resistance. We hypothesized that resistance is a late manifestation of early, treatment-induced histomolecular alterations; therefore, studying early drug response may identify drivers of resistance. We describe an epithelioid RCC growth pattern in RCC xenografts, which emerges in sunitinib-sensitive tumors and is augmented during resistance. This growth modality is molecularly and morphologically related to the RCC spheroids that advance during in vitro treatment. Based on time-lapse microscopy, mRNA and microRNA screening, and tumor behavior-related characteristics, we propose that the spheroid and adherent RCC growth patterns differentially respond to sunitinib. Gene expression analysis indicated that sunitinib promoted spheroid formation, which provided a selective survival advantage under treatment. Functional studies confirm that E-cadherin is a key contributor to the survival of RCC cells under sunitinib treatment. In summary, we suggest that sunitinib-resistant RCC cells exist in treatment-sensitive tumors and are histologically identifiable.-Lichner, Z., Saleeb, R., Butz, H., Ding, Q., Nofech-Mozes, R., Riad, S., Farag, M., Varkouhi, A. K., dos Santos, C. C., Kapus, A., Yousef, G. M. Sunitinib induces early histomolecular changes in a subset of renal cancer cells that contribute to resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/pathology , Drug Resistance, Neoplasm/drug effects , Kidney Neoplasms/pathology , Sunitinib/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Spheroids, Cellular , Stem Cells/metabolism , Xenograft Model Antitumor Assays
7.
Mol Oncol ; 12(10): 1673-1688, 2018 10.
Article in English | MEDLINE | ID: mdl-29896907

ABSTRACT

Papillary renal cell carcinoma (PRCC) is the most common nonclear cell RCCs and is known to comprise two histological subtypes. PRCC2 is more aggressive and is molecularly distinct from the other subtypes. Despite this, PRCCs are treated together as one entity, and they show poor response to the current therapies that do not target pathways implicated in their pathogenesis. We have previously detected ABCC2 (an ABC transporter), VEGF, and mTOR pathways to be enriched in PRCC2. In this study, we assess the therapeutic potential of targeting these pathways in PRCC2. Twenty RCC cell lines from the Cancer Cell Encyclopedia were compared to the Cancer Genome Atlas PRCC cohort (290), to identify representative PRCC2 cell lines. Cell lines were further validated in xenograft models. Selected cell lines were treated in vitro and in vivo (mice models) under five different conditions, untreated, anti-VEGF (sunitinib), ABCC2 blocker (MK571), mTOR inhibitor (everolimus) and sunitinib + MK571. Sunitinib +ABCC2 blocker group showed a significant response to therapy compared to the other treatment groups both in vitro (P ≤ 0.0001) and in vivo (P = 0.0132). ABCC2 blockage resulted in higher sunitinib uptake, both in vitro (P = 0.0016) and in vivo (P = 0.0031). Everolimus group demonstrated the second best response in vivo. The double-treatment group showed the highest apoptotic rate and lowest proliferation rate. There is an urgent need for individualized therapies of RCC subtypes that take into account their specific biology. Our results demonstrate that combined targeted therapy with sunitinib and ABCC2 blocker in PRCC2 has therapeutic potential. The results are likewise potentially significant for other ABCC2 high tumors. However, the results are preliminary and clinical trials are needed to confirm these effects in PRCC2 patients.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Molecular Targeted Therapy , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Biological Transport, Active , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Disease Models, Animal , Humans , Immunophenotyping , Kidney Neoplasms/pathology , Mice, SCID , Multidrug Resistance-Associated Protein 2 , Reproducibility of Results , Sunitinib/pharmacology , Sunitinib/therapeutic use
8.
Oncotarget ; 9(4): 4661-4674, 2018 Jan 12.
Article in English | MEDLINE | ID: mdl-29435133

ABSTRACT

Upon sunitinib treatment of metastatic renal cell carcinoma patients eventually acquire resistance. Our aim was to investigate microRNAs behind sunitinib resistance. We developed an in vivo xenograft and an in vitro model and compared morphological, immunhistochemical, transcriptomical and miRNome data changes during sunitinib response and resistance by performing next-generation mRNA and miRNA sequencing. Complex bioinformatics (pathway, BioFunction and network) analysis were performed. Results were validated by in vitro functional assays. Our morphological, immunhistochemical, transcriptomical and miRNome data all pointed out that during sunitinib resistance tumor cells changed to migratory phenotype. We identified the downregulated miR-1 and miR-663a targeting FRAS1 (Fraser Extracellular Matrix Complex Subunit 1) and MDGA1 (MAM Domain Containing Glycosylphosphatidylinositol Anchor 1) in resistant tumors. We proved firstly miR-1-FRAS1 and miR-663a-MDGA1 interactions. We found that MDGA1 knockdown decreased renal cancer cell migration and proliferation similarly to restoration of levels of miR-1 and miR-663. Our results support the central role of cell migration as an adaptive mechanism to secure tumor survival behind sunitinib resistance. MDGA1, FRAS1 or the targeting miRNAs can be potential adjuvant therapeutic targets, through inhibition of cancer cell migration, thus eliminating the development of resistance and metastasis.

9.
Oncotarget ; 7(42): 68057-68071, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27486976

ABSTRACT

Extracellular miRNAs are increasingly studied as markers for specific diseases. They are released in biological fluids in a remarkably stable form, and may play a role in intercellular communication. They are thought to be protected against degradation by either encapsulation within microparticles, or by binding to proteins (mostly AGO2). The particulate forms may be internalized by endocytosis or membrane fusion, but the protein-bound forms require a receptor mechanism for their uptake. A major question is whether there are natural cell-membrane receptors that capture and internalize protein-bound functional miRNAs. We examined neuropilin-1 (NRP1), in view of its properties as a receptor for many ligands, including growth factors such as vascular endothelial growth factor (VEGF), and efficiency at mediating ligand internalization. It is expressed by endothelial cells, many other normal cell types, and cancer cells. Here, we report that NRP1 binds miRNAs with high affinity, and promotes their entry into the cell. Furthermore, the internalized miRNAs remain functional, as they specifically regulate proliferation and migration of cancer cells, as well as tube formation by human endothelial cells. Anti-NRP1 antibodies or NRP1 siRNA knockdown block miRNA effects, further confirming NRP1-mediated uptake. VEGF does not compete with miRNAs for binding to NRP1. In addition, NRP1 binds extracellular AGO2 (carrying miRNA or not), and internalizes AGO2/miRNA complexes. Because miRNA bound to AGO2 appears to the most abundant form in body fluids, this may have important physiological and pathological effects.


Subject(s)
Argonaute Proteins/metabolism , Endocytosis , MicroRNAs/metabolism , Neuropilin-1/metabolism , Argonaute Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Neuropilin-1/genetics , Protein Binding , RNA Interference , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
Article in English | MEDLINE | ID: mdl-26543564

ABSTRACT

BACKGROUND: Translational research is an evolving discipline that is intended to bridge the gaps between basic science research, clinical research, and implementation in clinical practice. It is a fluid, multidirectional process that requires strong interdisciplinary collaboration to produce research that is relevant to end-users. PURPOSE OF THIS REVIEW: This review summarizes current perspectives on translational research and outlines its relevance and importance to kidney transplantation research. SOURCES OF INFORMATION: Sources of information used for this review include published reports, articles, and research funding websites. FINDINGS: Tissue typing is used as an in-depth example of how translational research has been applied in the field of kidney transplant medicine, and how it has resulted in successful implementation of diagnostic and management options for sensitized individuals undergoing kidney transplantation. The value of actively involving kidney transplant stakeholders (patients, caregivers, and clinicians) in setting research priorities and determining relevant outcomes for future investigation is also discussed. LIMITATIONS: This is a narrative review of the literature which has been partly influenced by the perspectives and experiences of its authors. IMPLICATIONS: Translational and patient-oriented research practices should be incorporated into future research endeavours in the field of kidney transplantation in order to create beneficial change in clinical practice and improve patient outcomes. WHAT WAS KNOWN BEFORE: Translational research which engages patients in the investigative process can enhance the likelihood that medical discoveries will have a meaningful impact at the bedside. WHAT THIS ADDS: This article applies current perspectives on translational research and patient engagement to the field of kidney transplantation, illustrating how these approaches have led to significant advancements in the field. It provides further justification for deliberate, targeted efforts to cross-collaborate and incorporate the patient voice into kidney transplant research.


CONTEXTE: La recherche translationnelle est une discipline évolutive qui a pour but de faire le pont entre la recherche fondamentale, la recherche clinique et la mise en œuvre de pratiques cliniques dans le domaine des transplantations rénales. Il s'agit d'un processus multidirectionnel et fluide qui demande la collaboration étroite de toutes les disciplines impliquées afin que la recherche qui en résulte soit pertinente et touche directement les usagers. OBJECTIFS DE LA REVUE: Cette revue fait la synthèse des éléments actuels de la recherche translationnelle, et décrit sa pertinence et son importance dans le domaine de la recherche sur la transplantation rénale. SOURCES: La réalisation de cette revue a été possible suite à la consultation de recueils et d'articles publiés ainsi que de sites web dédiés au financement de la recherche. CONSTATATIONS: L'étude de la compatibilité immunologique est utilisée à titre d'exemple pour démontrer en détail la façon dont la recherche translationnelle a été appliquée dans le domaine des greffes du rein jusqu'à maintenant, et comment elle a permis la mise en œuvre de solutions efficaces pour le diagnostic et l'organisation des soins aux patients subissant une greffe de rein. On a également discuté de l'importance d'impliquer toutes les parties prenantes d'une procédure de transplantation rénale, soit les patients eux-mêmes et le personnel soignant et le personnel clinique, afin d'établir les priorités de recherche et de définir les résultats pertinents en vue d'études ultérieures. LIMITES DE L'ÉTUDE: Il s'agit d'une revue non systématique de la littérature influencée en partie par la perspective et les connaissances des auteurs sur le sujet. CONSÉQUENCES: Il apparait important d'intégrer les pratiques courantes en recherche translationnelle de même qu'en recherche axée sur le patient lors de futures études sur les greffes de reins. Ceci afin d'instaurer un changement bénéfique dans la pratique clinique et par conséquent, d'améliorer les résultats chez les patients. DONNÉES CONNUES: Une approche de recherche translationnelle favorisant l'implication des patients dans le processus d'analyse peut augmenter les chances de voir les découvertes médicales avoir des répercussions directes et plus significatives pour le patient. CE QUE CETTE ÉTUDE AJOUTE: Cette revue expose les différents points de vue sur la recherche translationnelle et la collaboration des patients au processus, dans le domaine de la transplantation rénale. Elle illustre également la façon dont ces approches ont mené à des progrès marqués dans le domaine et plaide pour une collaboration volontaire et ciblée entre les différents intervenants ainsi que pour une plus grande implication des patients dans la recherche.

11.
Oncotarget ; 6(36): 39088-97, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26384299

ABSTRACT

Loss of the tumor suppressor gene AT-rich interactive domain-containing protein 1A (ARID1A) has been demonstrated in several cancers, but its prognostic role is unknown. We aimed to investigate the risk associated with loss of ARID1A (ARID1A-) for all-cause mortality, cancer-specific mortality and recurrence of disease in subjects with cancer. PubMed and SCOPUS search from database inception until 01/31/2015 without language restriction was conducted, contacting authors for unpublished data. Eligible were prospective studies reporting data on prognostic parameters in subjects with cancer, comparing participants with presence of ARID1A (ARID1A+) vs. ARID1A-, assessed either via immunohistochemistry (loss of expression) or with genetic testing (presence of mutation). Data were summarized using risk ratios (RR) for number of deaths/recurrences and hazard ratios (HR) for time-dependent risk related to ARID1A- adjusted for potential confounders. Of 136 hits, 25 studies with 5,651 participants (28 cohorts; ARID1A-: n = 1,701; ARID1A+: n = 3,950), with a mean follow-up period of 4.7 ± 1.8 years, were meta-analyzed. Compared to ARID1A+, ARID1A- significantly increased cancer-specific mortality (studies = 3; RR = 1.55, 95% confidence interval (CI) = 1.19-2.00, I(2) = 31%). Using HRs adjusted for potential confounders, ARID1A- was associated with a greater risk of cancer-specific mortality (studies = 2; HR = 2.55, 95%CI = 1.19-5.45, I(2) = 19%) and cancer recurrence (studies = 10; HR = 1.93, 95%CI = 1.22-3.05, I(2) = 76%). On the basis of these results, we have demonstrated that loss of ARID1A shortened time to cancer-specific mortality, and to recurrence of cancer when adjusting for potential confounders. For its role, this gene should be considered as an important potential target for personalized medicine in cancer treatment.


Subject(s)
Genes, Tumor Suppressor , Nuclear Proteins/genetics , Transcription Factors/genetics , Cohort Studies , DNA-Binding Proteins , Female , Humans , Male , Middle Aged , Mutation , Neoplasms/genetics , Prognosis
12.
J Pathol ; 237(2): 226-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26011734

ABSTRACT

The Gleason grading system is an important determinant of treatment decisions and prognosis in prostate cancer. It has a number of limitations, including significant inter-observer variability, creating a need for biological parameters to accurately assess the Gleason grade. The objective of this study was to determine the molecular correlates of the different Gleason grades. Global miRNA expression was analysed in pure regions of each Gleason grade. Bioinformatics analysis was performed to predict miRNA-mediated signalling. We experimentally validated the effect of miRNAs on target gene expression and cellular functions using cell line models. We also examined the correlation of miRNAs with biochemical failure, metastasis and prognosis. We identified miRNAs that are differentially expressed between grades 3 and 5, and the top biological processes associated with Gleason grade transition were extracellular matrix (ECM)-mediated signalling, focal adhesion kinase- and mitogen-activated kinase pathways. Transfection with miR-29c, miR-34a and miR-141 repressed genes involved in ECM-mediated pathways, such as SRC, PRKCA, COL1A1, ITGB1 and MAPK13, and decreased cell proliferation and migration. Furthermore, miR-29c and miR-34a influenced downstream pathways that affect actin cytoskeleton organization and androgen receptor localization. Finally, miR-29c, miR-34a, miR-141 and miR-148a showed inverse correlations with biochemical recurrence, but were independent of other clinical parameters. Our results demonstrate the potential role of miRNAs as independent prognostic markers and pave the road for a biological-based reclassification of the Gleason grading system.


Subject(s)
Cytoskeleton/metabolism , Extracellular Matrix/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Computational Biology , Cytoskeleton/genetics , Databases, Genetic , Disease Progression , Extracellular Matrix/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Neoplasm Grading , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Receptors, Androgen/genetics , Recurrence , Signal Transduction , Time Factors , Transfection , Treatment Failure
13.
Oncotarget ; 6(8): 5567-81, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25011053

ABSTRACT

Renal cell carcinoma (RCC) is an aggressive disease, with 35% chance of metastasis. The 'cancer stem cell' hypothesis suggests that a subset of cancer cells possess stem cell properties and is crucial in tumor initiation, metastasis and treatment resistance. We isolated RCC spheres and showed that they exhibit cancer stem cell/ tumor initiating cell-like properties including the formation of self-renewing spheres, high tumorigenicity and the ability to differentiate to cell types of the original tumor. Spheres showed increased expression of stem cell-related transcription factors and mesenchymal markers. miRNAs were differentially expressed between RCC spheres and their parental cells. Inhibition of miR-17 accelerated the formation of RCC spheres which shared molecular characteristics with the spontaneous RCC spheres. Target prediction pointed out TGFß pathway activation as a possible mechanism to drive RCC sphere formation. We demonstrate that miR-17 overexpression interferes with the TGFß-EMT axis and hinders RCC sphere formation; and validated TGFBR2 as a direct and biologically relevant target during this process. Thus, a single miRNA may have an impact on the formation of highly tumorigenic cancer spheres of kidney cancer.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MicroRNAs/antagonists & inhibitors , Animals , Carcinoma, Renal Cell/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Kidney Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Spheroids, Cellular/pathology , Transfection , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
14.
Biol Chem ; 395(9): 991-1001, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25153383

ABSTRACT

MicroRNAs (miRNAs) are short RNA nucleotides that negatively regulate their target genes. They are differentially expressed in prostate cancer. Kallikreins are genes that encode serine proteases and are dysregulated in cancer. We elucidated a miRNA-kallikrein network that can be involved in prostate cancer progression. Target prediction identified 23 miRNAs that are dysregulated between high and low risk biochemical failure and are predicted to target five kallikreins linked to prostate cancer; KLK2, KLK3, KLK4, KLK14 and KLK15. We also identified 14 miRNAs that are differentially expressed between Gleason grades and are predicted to target these kallikreins. This demonstrates that kallikreins are downstream effectors through which miRNAs influence tumor progression. We show, through in-silico and experimental analysis, that miR-378/422a and its gene targets PIK3CG, GRB2, AKT3, KLK4 and KLK14 form an integrated circuit in prostate cancer. Our analysis shows that a minisatellite sequence in the kallikrein locus consists of a number of microsatellite repeats that represent predicted miRNA response elements. A number of kallikrein and non-kallikrein prostate cancer-related genes share these microsatellite repeats. We validated some of these interactions in prostate cancer cell lines. Finally, we provide preliminary evidence on the presence of a miRNA-mediated cross-talk between kallikreins, including a kallikrein pseudogene.


Subject(s)
Disease Progression , Gene Regulatory Networks , Kallikreins/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Base Sequence , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Minisatellite Repeats/genetics , Models, Biological , Molecular Sequence Data , Neoplasm Grading
15.
Clin Chem ; 59(11): 1595-603, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23958847

ABSTRACT

PURPOSE: Prostate-specific antigen testing has led to overtreatment of prostate cancer (PCa). Only a small subset of PCa patients will have an aggressive disease that requires intensive therapy, and there is currently no biomarker to predict disease aggressiveness at the time of surgery. MicroRNAs (miRNAs) are reported to be involved in PCa pathogenesis. METHODS: This study involved 105 participants. For the discovery phase, prostatectomy samples were dichotomized to high-risk (n = 27, biochemical failure <36 months after prostatectomy) and low-risk groups (n = 14, ≥ 36 months without biochemical failure). Expression of 754 mature miRNAs was compared between the 2 groups. Linear regression models were built to accurately predict biochemical failure risk. miRNA mimics were transfected into PCa model cell lines to test effects on proliferation and to deduce responding signaling pathways. RESULTS: We identified 25 differentially expressed miRNAs between the biochemical failure risk groups. Based on the expression of 2-3 miRNAs, 3 logistic regression models were developed, each with a high positive predictive value. Candidate miRNAs and the best-performing model were also verified on an independent PCa set. miRNA-152, featured in the models, was further investigated by using cell line models and was shown to affect cell proliferation. Predicted interaction between miR-152 and (mRNA)ERBB3 (erythroblastic leukemia viral oncogene homolog 3) was experimentally validated in vitro. CONCLUSIONS: miRNAs can help to predict biochemical failure risk at the time of prostatectomy.


Subject(s)
MicroRNAs/metabolism , Neoplasm Recurrence, Local/diagnosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/diagnosis , Cell Line, Tumor , Cell Proliferation , Humans , Logistic Models , Male , MicroRNAs/analysis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptor, ErbB-3/genetics , Risk Assessment , Transcriptome
16.
Am J Pathol ; 182(4): 1163-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416164

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common tumor of the adult kidney, with an increasing rate of incidence. Recently, exome sequencing studies have revealed that the SWI/SNF (switch/sucrose nonfermentable) members PBRM1 and ARID1A are mutated in ccRCC, and it has also been suggested that aberrant chromatin regulation is a key step in kidney cancer pathogenesis. Herein, we show that down-regulation of another SW/SNF component, ARID1A, occurs frequently in ccRCC. We detected copy number loss of ARID1A in 16% of patients with ccRCC. Immunohistochemistry indicated that 67% of ccRCC (53 of 79) had significantly lower expression of BAF250a, the protein product of ARID1A, than did the matched normal kidney cortex. In parallel, we conducted in silico mRNA expression analysis on 404 ccRCC tumors and 167 normal kidney cortex samples using publicly available databases and confirmed significant down-regulation of ARID1A in 68.8% of patients. We also show that decreased BAF250a protein and ARID1A mRNA expression correlate with tumor stage and grade. Our results indicate that both the protein and mRNA levels of ARID1A are statistically significant prognostic markers for ccRCC. Even after controlling for other confounders in the multivariate analysis, BAF250 retained its prognostic significance. BAF250a IHC is easy to perform and represents a potential biomarker that could be incorporated in laboratory practice to enhance the accuracy of the existing prognostic models.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Chromatin Assembly and Disassembly/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Carcinoma, Renal Cell/classification , DNA Copy Number Variations/genetics , DNA-Binding Proteins , Disease-Free Survival , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/classification , Male , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Am J Kidney Dis ; 61(5): 798-808, 2013 May.
Article in English | MEDLINE | ID: mdl-23219107

ABSTRACT

MicroRNAs (miRNAs) are short noncoding RNA molecules that function by negatively regulating the expression of their target genes in a tightly controlled manner. Accumulating evidence, based in part on effects seen after miRNA overexpression and/or knockdown, points to the critical involvement of miRNAs in kidney function in health and disease. In this review, we provide a quick overview of the biogenesis of miRNAs and their potential involvement in kidney development and normal function. We also discuss the current literature that has begun to uncover the role of miRNAs in the pathogenesis of kidney diseases, including diabetic nephropathy, hypertension, glomerulonephritis, and cancer. As such, miRNAs have potential utility in the clinical realm as disease biomarkers. Moreover, miRNAs represent an attractive therapeutic target for a number of kidney diseases. We close by discussing a number of potential challenges that face the field of miRNA research and clinical use.


Subject(s)
Genetic Predisposition to Disease , Kidney Diseases/genetics , MicroRNAs/genetics , Aged , Female , Humans , Kidney Diseases/metabolism , MicroRNAs/metabolism
18.
J Mol Diagn ; 14(4): 385-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22580180

ABSTRACT

Renal cell carcinoma (RCC) is the most common neoplasm of the kidney. Increasing evidence suggests that microRNAs are dysregulated in RCC and are important factors in RCC pathogenesis. miR-21 is a known oncogene with tumor-promoting effects in many types of cancer. In this study, we analyzed miR-21 in 121 cases of healthy kidney and different RCC subtypes, including clear cell (ccRCC), papillary (pRCC), chromophobe (chRCC), and oncocytoma. Total RNA was extracted, and the expression of miR-21 was measured with real-time quantitative RT-PCR using miR-21-specific probes. The expression of miR-21 was significantly up-regulated in RCC compared with healthy kidney. There was a significant difference in the expression levels between RCC subtypes, with the highest levels of expression in ccRCC and pRCC subtypes. miR-21 expression distinguished ccRCC and pRCC from chRCC and oncocytoma with 90% specificity (95% CI, 63.9% to 98.1%) and 83% sensitivity (95% CI, 53.5% to 97.6%). Significantly higher miR-21 levels were associated with higher stage and grade. Patients who were miR-21 positive had statistically significant shorter disease-free and overall survival rates. Thus, miR-21 is up-regulated in RCC, and its expression levels can be used as a diagnostic marker to distinguish ccRCC and pRCC from chRCC and oncocytoma. Moreover, it has potential as a prognostic marker in RCC, although it is not independent of tumor stage and grade.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , Computational Biology , Humans , Reverse Transcriptase Polymerase Chain Reaction
19.
Tumour Biol ; 33(4): 1209-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22437349

ABSTRACT

The prognosis of patients with colorectal cancer (CRC) is assessed through conventional clinicopathological parameters, which are not always accurate. Members of the human kallikrein-related peptidases gene family represent potential cancer biomarkers. The aim of this study was to investigate the expression of human tissue kallikrein-related peptidase 10 (KLK10) by immunohistochemistry in CRC, to correlate this expression with various histopathological and clinical variables, and to evaluate its significance as a predictor of disease outcome. KLK10 expression was evaluated by immunohistochemistry and a combined expression score was calculated for each case based on intensity and percentage of positivity. A statistically significant positive association was observed between KLK10 and tumor stage and liver metastases (p = 0.015 and p = 0.035, respectively). Paradoxically, a negative association was observed between KLK10 and tumor grade (p = 0.009). Kaplan-Meier survival curves and univariate analysis showed that both KLK10 expression and stage had statistically significant correlations with disease-free survival (DFS) (p = 0.030 and p < 0.001, respectively) and overall survival (p = 0.010 and p = 0.001, respectively). Cox multivariate analysis showed that both KLK10 expression and stage were independent predictors of unfavorable DFS (p = 0.057 and p = 0.001, respectively) and overall survival (p = 0.009 and p = 0.001, respectively). In conclusion, KLK10 immunostaining is an independent prognostic marker in patients with CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/enzymology , Kallikreins/metabolism , Liver Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Female , Humans , Immunohistochemistry/statistics & numerical data , Kaplan-Meier Estimate , Liver Neoplasms/secondary , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Predictive Value of Tests , Prognosis , Proportional Hazards Models
20.
Am J Pathol ; 180(4): 1675-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326755

ABSTRACT

The von Hippel-Lindau (VHL) gene is lost in ≈ 70% of all renal cell carcinomas (RCCs); however, increasing evidence supports the involvement of alternative mechanisms in the regulation of VHL expression, including suppression by microRNAs (miRNAs). miRNAs are small, noncoding RNA molecules that regulate gene expression through binding to target mRNAs. In this study, we found that miRNAs, which are dysregulated in cases of RCC, can target multiple members of RCC-related signaling pathways. Importantly, both VHL and the hypoxia-inducible factor 1-α gene are experimentally validated and are likely direct targets of miR-17-5p and miR-224, as shown by both luciferase assay and Western blot analysis. We found a negative correlation between miR-17-5p and its two predicted targets, VEGF-A and EGLN3, and between miR-224 and its targets SMAD4 and SMAD5 in RCC specimens, suggesting that downstream signaling pathways are also modulated by clear cell RCC-dysregulated miRs. Results from our bioinformatics analysis show that a single miRNA molecule can target multiple components of the same pathway and that multiple miRNAs can target the same molecule. Our results also indicate that miRNAs represent a mechanism for the inactivation of VHL in cases of RCC and can elucidate a new dimension in cancer pathogenesis. As such, miRNAs exemplify new potential therapeutic targets with a significant effect on both tumor growth and metastatic potential.


Subject(s)
Carcinoma, Renal Cell/genetics , Genetic Pleiotropy/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , Carcinoma, Renal Cell/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Gene Expression Regulation, Neoplastic/physiology , Genetic Pleiotropy/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Messenger/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...