Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cells ; 11(15)2022 07 29.
Article in English | MEDLINE | ID: mdl-35954176

ABSTRACT

Three decades have passed from the initial discovery of a microRNA (miRNA) in Caenorhabditis elegans to our current understanding that miRNAs play essential roles in regulating fundamental physiological processes and that their dysregulation can lead to many human pathologies, including cancer. In effect, restoration of miRNA expression or downregulation of aberrantly expressed miRNAs using miRNA mimics or anti-miRNA inhibitors (anti-miRs/antimiRs), respectively, continues to show therapeutic potential for the treatment of cancer. Although the manipulation of miRNA expression presents a promising therapeutic strategy for cancer treatment, it is predominantly reliant on nucleic acid-based molecules for their application, which introduces an array of hurdles, with respect to in vivo delivery. Because naked nucleic acids are quickly degraded and/or removed from the body, they require delivery vectors that can help overcome the many barriers presented upon their administration into the bloodstream. As such, in this review, we discuss the strengths and weaknesses of the current state-of-the-art delivery systems, encompassing viral- and nonviral-based systems, with a specific focus on nonviral nanotechnology-based miRNA delivery platforms, including lipid-, polymer-, inorganic-, and extracellular vesicle-based delivery strategies. Moreover, we also shed light on peptide carriers as an emerging technology that shows great promise in being a highly efficacious delivery platform for miRNA-based cancer therapeutics.


Subject(s)
MicroRNAs , Neoplasms , Nucleic Acids , Humans , MicroRNAs/metabolism , Neoplasms/drug therapy , Neoplasms/therapy , Peptides , Polymers
2.
Mol Ther Nucleic Acids ; 24: 462-476, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-33868789

ABSTRACT

The 599 peptide has been previously shown to effectively deliver small interfering RNAs (siRNAs) to cancer cells, inducing targeted-oncogene silencing, with a consequent inhibition of tumor growth. Although effective, this study was undertaken to advance the 599 peptide siRNA-carrier design through L/D-amino acid stereochemical modifications. Consequently, 599 was modified to generate eight different peptide variants, incorporating either different stereochemical patterns of L/D-amino acids or a specific D-amino acid substitution. Upon analysis of the variants, it was observed that these modifications could, in some instances, increase/decrease the binding, nuclease/serum stability, and complex release of siRNAs, as well as influence the gene-silencing efficiencies of the complex. These modifications were also found to affect cellular uptake and intracellular localization patterns of siRNA cargo, with one particular variant capable of mediating binding of siRNAs to specific cellular projections, identified as filopodia. Interestingly, this variant also exhibited the most enhanced gene silencing in comparison to the parent 599 peptide, thus suggesting a possible connection between filopodia binding and enhanced gene silencing. Together, these data demonstrate the utility of peptide stereochemistry, as well as the importance of a key D-amino acid modification, in advancing the 599 carrier design for the enhancement of gene silencing in cancer cells.

3.
Transl Res ; 214: 92-104, 2019 12.
Article in English | MEDLINE | ID: mdl-31404520

ABSTRACT

Cancer is a significant health concern worldwide and its clinical treatment presents many challenges. Consequently, much research effort has focused on the development of new anticancer drugs to combat this disease. One area of exploration, in particular, has been in the therapeutic application of RNA interference (RNAi). Although RNAi appears to be an attractive therapeutic tool for the treatment of cancer, one of the primary obstacles towards its pervasive use in the clinic has been cell/tissue type-specific cytosolic delivery of therapeutic small interfering RNA (siRNA) molecules. Consequently, varied drug delivery platforms have been developed and widely explored for siRNA delivery. Among these candidate drug delivery systems, peptides have shown great promise as siRNA carriers due to their varied physiochemical properties and functions, simple formulations, and flexibility in design. In this review, we will focus on distinguishing between the different classes of peptide carriers based on their functions, as well as summarize and discuss the various design strategies and advancements that have been made in circumventing the barriers to siRNA delivery for cancer treatment. Resolution of these challenges by peptide carriers will accelerate the translation of RNAi-based therapies to the clinic.


Subject(s)
Gene Transfer Techniques , Neoplasms/therapy , Peptides/chemistry , RNA, Small Interfering/administration & dosage , Amino Acid Sequence , Animals , Endosomes/metabolism , Humans
4.
Oral Oncol ; 72: 123-131, 2017 09.
Article in English | MEDLINE | ID: mdl-28797448

ABSTRACT

OBJECTIVES: Despite significant advances in cancer treatment, the prognosis for oral cancer remains poor in comparison to other cancer types, including breast, skin, and prostate. As a result, more effective therapeutic modalities are needed for the treatment of oral cancer. Consequently, in the present study, we examined the feasibility of using a dual peptide carrier approach, combining an epidermal growth factor receptor (EGFR)-targeting peptide with an endosome-disruptive peptide, to mediate targeted delivery of small interfering RNAs (siRNAs) into EGFR-overexpressing oral cancer cells and induce silencing of the targeted oncogene, cancerous inhibitor of protein phosphatase 2A (CIP2A). MATERIALS AND METHODS: Fluorescence microscopy, real-time PCR, Western blot analysis, and in vivo bioimaging of mice containing orthotopic xenograft tumors were used to examine the ability of the dual peptide carrier to mediate specific delivery of bioactive siRNAs into EGFR-overexpressing oral cancer cells/tissues. RESULTS: Co-complexation of the EGFR-targeting peptide, GE11R9, with the endosome-disruptive 599 peptide facilitated the specific uptake of siRNAs into oral cancer cells overexpressing EGFR in vitro with optimal gene silencing observed at a 60:30:1 (GE11R9:599:siRNA) molar ratio. Furthermore, when administered systemically to mice bearing xenograft oral tumors, this dual peptide complex mediated increased targeted delivery of siRNAs into tumor tissues in comparison to the 599 peptide alone and significantly enhanced CIP2A silencing. CONCLUSION: Herein we provide the first report demonstrating the clinical potential of a dual peptide strategy for siRNA-based therapeutics by synergistically mediating the effective targeting and delivery of bioactive siRNAs into EGFR-overexpressing oral cancer cells.


Subject(s)
Mouth Neoplasms/drug therapy , Peptides/administration & dosage , RNA, Small Interfering/administration & dosage , Administration, Intravenous , Amino Acid Sequence , Animals , ErbB Receptors/genetics , Genetic Therapy , Heterografts , Humans , Mice , Mouth Neoplasms/genetics , Peptides/chemistry , RNA, Small Interfering/genetics
5.
J Control Release ; 218: 72-81, 2015 Nov 28.
Article in English | MEDLINE | ID: mdl-26386438

ABSTRACT

Intracellular delivery and endosomal escape of functional small interfering RNAs (siRNAs) remain major barriers limiting the clinical translation of RNA interference (RNAi)-based therapeutics. Recently, we demonstrated that a cell-penetrating endosome-disruptive peptide we synthesized, termed 599, enhanced the intracellular delivery and bioavailability of siRNAs designed to target the CIP2A oncoprotein (siCIP2A) into oral cancer cells and consequently inhibited oral cancer cell invasiveness and anchorage-independent growth in vitro. Thus, to further assess the therapeutic potential of the 599 peptide in mediating RNAi-based therapeutics for oral cancer and its prospective applicability in clinical settings, the objective of the current study was to determine whether intratumoral dosing of the 599 peptide-siCIP2A complex could induce silencing of CIP2A and consequently impair tumor growth using a xenograft oral cancer mouse model. Our results demonstrate that the 599 peptide is able to protect siRNAs from degradation by serum and ribonucleases in vitro and upon intratumoral injection in vivo, confirming the stability of the 599 peptide-siRNA complex and its potential for therapeutic utility. Moreover, 599 peptide-mediated delivery of siCIP2A to tumor tissue induces CIP2A silencing without any associated toxicity, consequently resulting in reduction of the mitotic index and significant inhibition of tumor growth. Together, these data suggest that the 599 peptide carrier is a clinically effective mediator of RNAi-based cancer therapeutics.


Subject(s)
Autoantigens/genetics , Cell-Penetrating Peptides/administration & dosage , Membrane Proteins/genetics , Mouth Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Animals , Arginine/chemistry , Cell Line, Tumor , Cell-Penetrating Peptides/therapeutic use , Gene Silencing , Humans , Mice, Nude , Mouth Neoplasms/pathology , RNA, Messenger/metabolism , RNA, Small Interfering/therapeutic use , Tumor Burden/drug effects
6.
Mol Cancer ; 13: 190, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25115815

ABSTRACT

BACKGROUND: The human dicer1 gene has been predicted to produce several mRNA variants that encode truncated Dicer1 proteins of varying lengths. One of these Dicer1 variants, Dicer1e, was recently found to be differentially expressed in breast cancer cells. Because the expression and function of the Dicer1e protein variant has not been well characterized and the underlying molecular mechanisms for the development of oral squamous cell carcinomas (OSCCs) are poorly understood, the present study sought to characterize the biological role of Dicer1e and determine its relationship, if any, to OSCC pathogenesis. METHODS: Western blot analyses were used to examine Dicer1e expression levels in a panel of oral cancer cells/tissues and during epithelial-mesenchymal transition (EMT), followed by 5'/3'-RACE analyses to obtain the full-length Dicer1e transcript. Biochemical fractionation and indirect immunofluorescent studies were performed to determine the cellular localization of Dicer1e and the effects of Dicer1e silencing on cancer cell proliferation, clonogenicity, and drug sensitivity were also assessed. RESULTS: Dicer1e protein levels were found to be overexpressed in OSCC cell lines of epithelial phenotype and in OSCC tissues with its levels downregulated during EMT. Moreover, the Dicer1e protein was observed to predominantly localize in the nucleus. 5'/3'-RACE analyses confirmed the presence of the Dicer1e transcript and silencing of Dicer1e impaired both cancer cell proliferation and clonogenicity by inducing either apoptosis and/or G2/M cell cycle arrest. Lastly, Dicer1e knockdown enhanced the chemosensitivity of oral cancer cells to cisplatin. CONCLUSION: The expression levels of Dicer1e influence the pathogenesis of oral cancer cells and alter their response to chemosensitivity, thus supporting the importance of Dicer1e as a therapeutic target for OSCCs.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Mouth Neoplasms/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Alternative Splicing , Cell Line, Tumor , Cell Nucleus/metabolism , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/pathology , RNA, Messenger/metabolism
7.
PLoS One ; 8(9): e73348, 2013.
Article in English | MEDLINE | ID: mdl-24019920

ABSTRACT

Despite a better understanding of the pathogenesis of oral cancer, its treatment outcome remains poor. Thus, there is a need for new therapeutic strategies to improve the prognosis of this disease. RNA interference (RNAi) appears to be a promising therapeutic tool for the treatment of many diseases, including oral cancer. However, an obstacle for RNAi-mediated therapies has been delivery, in particular, the retention of small interfering RNAs (siRNAs) in endosomes and their subsequent degradation in lysosomes, resulting in inefficient gene silencing. Thus, the current study examined the feasibility of designing and utilizing a peptide, termed 599, consisting of a synthetic influenza virus-derived endosome-disruptive fusogenic peptide sequence and a stretch of cationic cell-penetrating nona(D-arginine) residues, to deliver siRNAs into oral cancer cells and induce silencing of the therapeutic target, CIP2A, an oncoprotein overexpressed in various human malignancies including oral cancer. Increasing the 599 peptide-to-siRNA molar ratio demonstrated a higher binding capacity for siRNA molecules and enhanced siRNA delivery into the cytoplasm of oral cancer cells. In fact, quantitative measurements of siRNA delivery into cells demonstrated that a 50∶1 peptide-to-siRNA molar ratio could deliver 18-fold higher amounts of siRNAs compared to cells treated with siRNA alone with no significant long-term cytotoxic effects. Most importantly, the 599 peptide-mediated siRNA delivery promoted significant CIP2A mRNA and protein silencing which resulted in decreased oral cancer cell invasiveness and anchorage-independent growth. Together, these data demonstrate that a chimeric peptide consisting of a fusogenic sequence, in combination with cell-penetrating residues, can be used to effectively deliver siRNAs into oral cancer cells and induce the silencing of its target gene, potentially offering a new therapeutic strategy in combating oral cancer.


Subject(s)
Arginine/administration & dosage , Autoantigens/genetics , Membrane Proteins/genetics , Mouth Neoplasms/genetics , Oncogenes , RNA, Small Interfering/administration & dosage , Cell Line, Tumor , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins , Mouth Neoplasms/pathology , RNA Interference
8.
J Biol Chem ; 287(35): 29261-72, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22761427

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that posttranscriptionally regulate gene expression during many biological processes. Recently, the aberrant expressions of miRNAs have become a major focus in cancer research. The purpose of this study was to identify deregulated miRNAs in oral cancer and further focus on specific miRNAs that were related to patient survival. Here, we report that miRNA expression profiling provided more precise information when oral squamous cell carcinomas were subcategorized on the basis of clinicopathological parameters (tumor primary site, histological subtype, tumor stage, and HPV16 status). An innovative radar chart analysis method was developed to depict subcategories of cancers taking into consideration the expression patterns of multiple miRNAs combined with the clinicopathological parameters. Keratinization of tumors and the high expression of miR-21 were the major factors related to the poor prognosis of patients. Interestingly, a majority of the keratinized tumors expressed high levels of miR-21. Further investigations demonstrated the regulation of the tumor suppressor gene reversion-inducing cysteine-rich protein with kazal motifs (RECK) by two keratinization-associated miRNAs, miR-7 and miR-21. Transfection of miR-7 and miR-21-mimics reduced the expression of RECK through direct miRNA-mediated regulation, and these miRNAs were inversely correlated with RECK in CAL 27 orthotopic xenograft tumors. Furthermore, a similar inverse correlation was demonstrated in CAL 27 cells treated in vitro by different external stimuli such as trypsinization, cell density, and serum concentration. Taken together, our data show that keratinization is associated with poor prognosis of oral cancer patients and keratinization-associated miRNAs mediate deregulation of RECK which may contribute to the aggressiveness of tumors.


Subject(s)
GPI-Linked Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Mouth Neoplasms/metabolism , RNA, Neoplasm/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Animals , GPI-Linked Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Transplantation , RNA, Neoplasm/genetics , Transcriptome , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics
9.
Int J Cell Biol ; 2012: 603961, 2012.
Article in English | MEDLINE | ID: mdl-22505930

ABSTRACT

The human genome contains microRNAs (miRNAs), small noncoding RNAs that orchestrate a number of physiologic processes through regulation of gene expression. Burgeoning evidence suggests that dysregulation of miRNAs may promote disease progression and cancer pathogenesis. Virus-encoded miRNAs, exhibiting unique molecular signatures and functions, have been increasingly recognized as contributors to viral cancer pathogenesis. A large segment of the existing knowledge in this area has been generated through characterization of miRNAs encoded by the human gamma-herpesviruses, including the Kaposi's sarcoma-associated herpesvirus (KSHV). Recent studies focusing on KSHV miRNAs have led to a better understanding of viral miRNA expression in human tumors, the identification of novel pathologic check points regulated by viral miRNAs, and new insights for viral miRNA interactions with cellular ("human") miRNAs. Elucidating the functional effects of inhibiting KSHV miRNAs has also provided a foundation for further translational efforts and consideration of clinical applications. This paper summarizes recent literature outlining mechanisms for KSHV miRNA regulation of cellular function and cancer-associated pathogenesis, as well as implications for interactions between KSHV and human miRNAs that may facilitate cancer progression. Finally, insights are offered for the clinical feasibility of targeting miRNAs as a therapeutic approach for viral cancers.

10.
Arch Oral Biol ; 56(12): 1506-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21704302

ABSTRACT

OBJECTIVE: Identifying discriminatory human salivary RNA biomarkers reflective of disease in a low-cost non-invasive screening assay is crucial to salivary diagnostics. Recent studies have reported both mRNA and microRNA (miRNA) in saliva, but little information has been documented on the quality and yield of RNA collected. Therefore, the aim of the present study was to develop an improved RNA isolation method from saliva and to identify major miRNA species in human whole saliva. DESIGN: RNA samples were isolated from normal human saliva using a combined protocol based on the Oragene RNA collection kit and the mirVana miRNA isolation kit in tandem. RNA samples were analysed for quality and subjected to miRNA array analysis. RESULTS: RNA samples isolated from twenty healthy donors ranged from 2.59 to 29.4 µg/ml saliva and with 1.92-2.16OD(260/280 nm) ratios. RNA yield and concentration of saliva samples were observed to be stable over 48 h at room temperature. Analysis of total salivary RNA isolated from these twenty donors showed no statistical significance between sexes; however, the presence of high-, medium-, and low-yield salivary RNA producers was detected. MiRNA array analysis of salivary RNA detected five abundantly expressed miRNAs, miR-223, miR-191, miR-16, miR-203, and miR-24, that were similarly described in other published reports. Additionally, many previously undetected miRNAs were also identified. CONCLUSION: High quality miRNAs can be isolated from saliva using available commercial kits, and in future studies, the availability of this isolation protocol may allow specific changes in their levels to be measured accurately in various relevant diseases.


Subject(s)
Biomarkers/analysis , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis/methods , Saliva/chemistry , Female , Humans , Linear Models , Male , MicroRNAs/isolation & purification
11.
Cancer Biol Ther ; 10(7): 694-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-21068540

ABSTRACT

AIMS: Oral squamous cell carcinoma (OSCC) is the most prevalent malignancy of the oral cavity resulting in severe morbidity and mortality. To date only few proteins have been suggested as potential biomarkers or targets for this type of cancer. Cancerous inhibitor of PP2A (CIP2A) is a protein expressed in epithelial tissues that stabilizes the oncogene c-Myc and causes cell transformation. This study was designed to investigate the expression of CIP2A in OSCC cell lines and tissues representing human normal, dysplasia and OSCC. METHODS: Using quantitative real time PCR, mRNA quantification for CIP2A was performed in a primary gingival cell line and OSCCs CAL 27 and SCC-25. Paraffin embedded human specimen classified as normal, dysplastic or OSCC were immunohistochemically stained for CIP2A expression. EGFR and CIP2A were also stained by immunofluorescence for co-localization. Samples of human normal oral tissue and OSCC were studied by PCR for mRNA expression of CIP2A. RESULTS: CIP2A was significantly increased in the human carcinoma cell lines compared to the primary gingival cell line. CIP2A was overexpressed in the human oral dysplasia and OSCC tissues compared to normal oral tissues. CIP2A was also preferentially localized in the dysplastic and OSCC epithelial areas compared to EGFR that was expressed mainly in areas of relatively normal epithelium and in dysplastic tissues above the basal layers. CONCLUSIONS: CIP2A may play a significant role in oral malignant transformation and therefore, it may be a potential target for chemotherapy of OSCC.


Subject(s)
Autoantigens/metabolism , Carcinoma, Squamous Cell/metabolism , Membrane Proteins/metabolism , Mouth Diseases/metabolism , Mouth Neoplasms/metabolism , Autoantigens/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mouth Diseases/genetics , Mouth Diseases/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
12.
Genes Chromosomes Cancer ; 49(6): 549-59, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20232482

ABSTRACT

Recent reports have demonstrated that Dicer, an RNase III endonuclease required for microRNA (miRNA) maturation, is aberrantly expressed in different types of cancer. Furthermore, Dicer has been reported to be regulated by the let-7 family of miRNA genes. We hypothesize that Dicer is aberrantly expressed in oral cancer cells due to altered expressions of let-7 and that Dicer contributes to the development and progression of the disease. Western blot examination of Dicer protein levels in four head and neck squamous cell carcinoma (HNSCC) cell lines, including two oral cancer cell lines, demonstrated that Dicer had between 4- and 24-fold higher expression levels when compared to normal human primary gingival epithelial cells. Furthermore, five of six oral cancer tissues analyzed by indirect immunofluorescence had increased Dicer protein expression, compared to normal gingival epithelial tissue. The Dicer mRNA levels were not found to correlate well with protein expression in the HNSCC cell lines, suggesting that Dicer protein expression was post-transcriptionally regulated. Analysis of let-7a and let-7b levels in HNSCC cell lines by real-time PCR demonstrated that let-7b, but not let-7a, was significantly reduced in the HNSCC cell lines compared to control cells. Lastly, transfection of oral cancer cells with chemically synthesized let-7b and small interfering RNAs targeting Dicer significantly inhibited cell proliferation up to 83% and >100%, respectively, as early as 3 days post-transfection. Together, these data demonstrate that elevated expression levels of Dicer in oral cancer cells correlate with downregulation of let-7b and increased cell proliferation.


Subject(s)
Carcinoma, Squamous Cell/genetics , DEAD-box RNA Helicases/genetics , MicroRNAs/genetics , Mouth Neoplasms/genetics , Ribonuclease III/genetics , Analysis of Variance , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Growth Processes/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histocytochemistry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Microscopy, Fluorescence , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Transplantation , Ribonuclease III/biosynthesis , Ribonuclease III/metabolism , Transfection
13.
J Gene Med ; 10(2): 132-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18064719

ABSTRACT

BACKGROUND: Reduced contractility due to dysregulation of intracellular calcium (Ca(2+)) is a common pathologic feature of chronic heart failure. Calcium stores in the sarcoplasmic reticulum play a major role in regulating cardiac contractility. Several animal models of heart failure have been treated by altering the regulation of the sarcoplamic reticulum ATPase through ablation or down-regulation of its inhibitor peptide, phospholamban (PLN). METHODS: We have designed two small hairpin RNAs (shRNAs) to block the synthesis of PLN via RNA interference. These were tested in cell culture using a co-transfection assay and using adeno-associated virus (AAV)-mediated delivery to cardiomyocytes. Reverse-transcription polymerase chain reaction (RT-PCR) and Western blots were used to measure reduction in PLN mRNA and protein levels. Reduction of PLN was also documented by indirect immunofluorescence. Free cytosolic calcium and contractile properties of transduced cardiomyocytes was examined on fura-2-loaded cells. Direct cardiac injection was used to deliver AAV1-shRNAs to mice, and reduction of PLN was measured by indirect immunofluorescence. RESULTS: Both siRNAs led to significant reduction of PLN RNA and protein levels in cultured cells. Down-regulation of PLN led to enhanced cell shortening and relaxation and to a decrease in the time constant of calcium decay, signs of improved contractility and calcium handling. In the hearts of AAV-infected mice, shRNA-transduced cells showed significant reduction in the level of PLN. CONCLUSIONS: Our results suggest that AAV-delivered shRNAs mediated physiologically significant suppression of phospholamban that may be useful in combating the effects of chronic heart failure.


Subject(s)
Calcium-Binding Proteins/deficiency , Calcium/metabolism , Dependovirus/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cell Separation , Gene Expression Regulation , Gene Silencing , Genetic Vectors , Humans , Mice , Myocytes, Cardiac/cytology , Plasmids , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats
14.
J Cell Sci ; 120(Pt 8): 1317-23, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17401112

ABSTRACT

GW bodies, also known as mammalian P-bodies, are cytoplasmic foci involved in the post-transcriptional regulation of eukaryotic gene expression. Recently, GW bodies have been linked to RNA interference and demonstrated to be important for short-interfering-RNA- and microRNA-mediated mRNA decay and translational repression. Evidence indicates that both passenger and guide strands of short-interfering RNA duplexes can localize to GW bodies, thereby indicating that RNA-induced silencing complexes may be activated within these cytoplasmic centers. Formation of GW bodies appears to depend on both specific protein factors and RNA, in particular, microRNA. Work over the past few years has significantly increased our understanding of the biology of GW bodies, revealing that they are specialized cell components that spatially regulate mRNA turnover in various biological processes. The formation of GW bodies appears to depend on both specific protein factors and RNA, in particular, microRNA. Here, we propose a working model for GW body assembly in terms of its relationship to RNA interference. In this process, one or more heteromeric protein complexes accumulate in successive steps into larger ribonucleoprotein structures.


Subject(s)
Cytoplasmic Structures/metabolism , Gene Silencing , RNA Processing, Post-Transcriptional , Animals , MicroRNAs/genetics , RNA Interference
15.
EMBO Rep ; 7(9): 904-10, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906129

ABSTRACT

GW bodies (GWBs), or mammalian P bodies, proposed to be involved in messenger RNA storage and/or degradation, have recently been linked to RNA interference and microRNA (miRNA) processing. We report that endogenous let-7 miRNA co-precipitates with the GW182 protein complex. In addition, knockdown of two proteins, Drosha and its protein partner DGCR8, which are vital to the generation of mature miRNA, results in the loss of GWBs. Subsequent introduction of short interference RNA specific to lamin A/C is accompanied by reassembly of GWBs and concurrent knockdown of lamin A/C protein. Taken together, these studies show that miRNAs are crucial components in GWB formation.


Subject(s)
Cytoplasmic Structures/metabolism , MicroRNAs/metabolism , RNA Transport , Ribonuclease III/genetics , Cytoplasmic Structures/physiology , HeLa Cells , Humans , MicroRNAs/physiology , Models, Biological , RNA Stability , Transfection
16.
Arthritis Res Ther ; 8(4): R87, 2006.
Article in English | MEDLINE | ID: mdl-16684366

ABSTRACT

RNA interference (RNAi) is an evolutionarily conserved mechanism that is involved in the post-transcriptional silencing of genes. This process elicits the degradation or translational inhibition of mRNAs based on the complementarity with short interfering RNAs (siRNAs) or microRNAs (miRNAs). Recently, differential expression of specific miRNAs and disruption of the miRNA synthetic pathway have been implicated in cancer; however, their role in autoimmune disease remains largely unknown. Here, we report that anti-Su autoantibodies from human patients with rheumatic diseases and in a mouse model of autoimmunity recognize the human Argonaute (Ago) protein, hAgo2, the catalytic core enzyme in the RNAi pathway. More specifically, 91% (20/22) of the human anti-Su sera were shown to immunoprecipitate the full-length recombinant hAgo2 protein. Indirect immunofluorescence studies in HEp-2 cells demonstrated that anti-Su autoantibodies target cytoplasmic foci identified as GW bodies (GWBs) or mammalian P bodies, structures recently linked to RNAi function. Furthermore, anti-Su sera were also capable of immunoprecipitating additional key components of the RNAi pathway, including hAgo1, -3, -4, and Dicer. Together, these results demonstrate an autoimmune response to components of the RNAi pathway which could potentially implicate the involvement of an innate anti-viral response in the pathogenesis of autoantibody production.


Subject(s)
Autoimmunity/immunology , RNA Interference/immunology , Animals , Argonaute Proteins , Autoantibodies/immunology , Cell Line , Cytoplasm/immunology , Eukaryotic Initiation Factor-2 , Eukaryotic Initiation Factors , Fluorescent Antibody Technique, Indirect , Humans , Immunoprecipitation , Mice , Mice, Inbred BALB C , Peptide Initiation Factors/immunology , Proteins/immunology , Recombinant Proteins/immunology , Rheumatic Diseases/immunology , Ribonuclease III/immunology
17.
Cell Cycle ; 5(3): 242-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16418578

ABSTRACT

GW bodies (GWBs) are cytoplasmic foci initially identified through the use of an autoimmune serum targeting the marker protein, GW182. GWBs were first considered as both storage centers for a specific subset of mRNAs and degradation sites for mRNAs. Interestingly, they are known to vary in size and number throughout the cell cycle and are largest in size and most abundant in number during the late S and G2 phases. Recent studies have linked RNA interference to GWBs, in that disruption or disassembly of GWBs was demonstrated to impair siRNA and miRNA silencing activity. As miRNAs are implicated in the regulation of cell cycle progression and cell proliferation, it is very likely that GWBs, the critical intracellular structures for miRNA function, may very well be also linked to this cellular process.


Subject(s)
Cell Cycle/physiology , Cytoplasmic Structures/metabolism , MicroRNAs/metabolism , Autoantigens/genetics , Autoantigens/metabolism , HeLa Cells , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , MicroRNAs/genetics , RNA Interference , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins , Transfection
18.
Nat Cell Biol ; 7(12): 1267-74, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16284622

ABSTRACT

The GW182 RNA-binding protein was initially shown to associate with a specific subset of mRNAs and to reside within discrete cytoplasmic foci named GW bodies (GWBs). GWBs are enriched in proteins that are involved in mRNA degradation. Recent reports have shown that exogenously introduced human Argonaute-2 (Ago2) is also enriched in GWBs, indicating that RNA interference function may be somehow linked to these structures. In this report, we demonstrate that endogenous Ago2 and transfected small interfering RNAs (siRNAs) are also present within these same cytoplasmic bodies and that the GW182 protein interacts with Ago2. Disruption of these cytoplasmic foci in HeLa cells interferes with the silencing capability of a siRNA that is specific to lamin-A/C. Our data support a model in which GW182 and/or the microenvironment of the cytoplasmic GWBs contribute to the RNA-induced silencing complex and to RNA silencing.


Subject(s)
Autoantigens/physiology , Cytoplasmic Structures/physiology , RNA Interference , Argonaute Proteins , Autoantigens/metabolism , Cytoplasmic Structures/chemistry , Eukaryotic Initiation Factor-2 , HeLa Cells , Humans , Lamin Type A/genetics , Peptide Initiation Factors/metabolism , RNA, Small Interfering , RNA-Binding Proteins
19.
J Cell Sci ; 117(Pt 23): 5567-78, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494374

ABSTRACT

A novel cytoplasmic compartment referred to as GW bodies was initially identified using human autoantibodies to a 182 kDa protein named GW182. GW bodies are small, generally spherical, cytoplasmic domains that vary in number and size in several mammalian cell types examined to date. Based on our earlier studies, GW bodies were proposed to be cytoplasmic sites for mRNA storage and/or degradation. In the present study, immunogold electron microscopy identified electron dense structures of 100-300 nm diameter devoid of a lipid bilayer membrane. These structures appeared to comprise clusters of electron dense strands of 8-10 nm in diameter. By costaining with CENP-F and PCNA, and employing a double-thymidine block to synchronize HeLa cells, GW bodies were observed to be small in early S phase and larger during late S and G2 phases of the cell cycle. The majority of GW bodies disassembled prior to mitosis and small GW bodies reassembled in early G1. The analysis of GW bodies in two experimental models of cell proliferation using reversal of 3T3/serum-starvation and concanavalin A stimulation of mouse splenocytes and T cells, revealed that proliferating cells contained larger, brighter, and more numerous GW bodies as well as up to a fivefold more total GW182 protein than quiescent cells. In vitro gene knockdown of GW182 led to the disappearance of GW bodies demonstrating that GW182 is a critical component of GW bodies. The incremental expression of the GW182 protein in cells induced to proliferate and the cyclic formation and breakdown of GW bodies during mitosis are intriguing in view of the notion that GW bodies are specialized centers involved in maintaining stability and/or controlling degradation of mRNA.


Subject(s)
Autoantigens/metabolism , Cell Proliferation , Cytoplasmic Structures/metabolism , G1 Phase/physiology , G2 Phase/physiology , S Phase/physiology , 3T3 Cells , Animals , Autoantigens/genetics , Cell Compartmentation/physiology , Chromosomal Proteins, Non-Histone/metabolism , Concanavalin A , Culture Media, Serum-Free/pharmacology , Cytoplasmic Structures/ultrastructure , Down-Regulation/physiology , Female , HeLa Cells , Humans , Immunohistochemistry , Mice , Microfilament Proteins , Microscopy, Electron, Transmission , Proliferating Cell Nuclear Antigen/metabolism , RNA Stability/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins
20.
Oncogene ; 23(21): 3781-9, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15021911

ABSTRACT

Sam68 (Src-associated in mitosis; 68 kDa) is a member of the STAR (signal transduction and activation of RNA) family of KH domain-containing RNA binding proteins. Accumulating evidence suggests that it plays an important role in cell cycle control. Tyrosine phosphorylation by Src family kinases and breast tumor kinase can negatively regulate its RNA binding activity. To date, there are no reports of a factor, such as a phosphatase, which can positively regulate Sam68 association with RNA. Acetylation is a reversible post-translational modification known to influence the activity of DNA binding proteins. However, acetylation of a cellular RNA binding protein as a mechanism for regulating its activity has not yet been reported. Here we demonstrate Sam68 to be acetylated in vivo. A screen of several human mammary epithelial cell lines revealed variations in Sam68 acetylation. Interestingly, the highest level of acetylation was found in tumorigenic breast cancer cell lines. The screen also showed a positive correlation between Sam68 acetylation and its ability to bind RNA. The acetyltransferase CBP was shown to acetylate Sam68 and enhance its binding to poly(U) RNA. These results suggest that Sam68 association with RNA substrates may be positively regulated by acetylation, and that enhanced acetylation and RNA binding activity of Sam68 may play a role in tumor cell proliferation.


Subject(s)
DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Acetylation , Adaptor Proteins, Signal Transducing , Animals , CREB-Binding Protein , Cell Line, Tumor , Histone Deacetylases/physiology , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/physiology , Trans-Activators/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...