Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Acta Ophthalmol ; 100(5): e1172-e1185, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34687141

ABSTRACT

PURPOSE: Dysfunction of the retinal pigment epithelium (RPE) causes numerous forms of retinal degeneration. RPE replacement is a modern option to save vision. We aimed to test the results of transplanting cultured RPEs on biocompatible membranes. METHODS: We cultivated porcine primary RPE cells isolated from cadaver eyes from the slaughterhouse on two types of membranes: commercial polyester scaffolds Transwell (Corning Inc., Kenneburg, ME, USA) with 0.4 µm pore size and prepared Poly (L-lactide-co-DL-lactide) (PDLLA) nanofibrous membranes with an average pore size of 0.4 µm. RESULTS: Five types of assays were used for the analysis: immunocytochemistry (ICC), phagocytosis assay, Western blotting, real-time qPCR (RT-qPCR) and electron microscopy. RT-qPCR demonstrated that RPEs cultured on nanofibrous membranes have higher expressions of BEST1 (bestrophin 1), RLBP1 (retinaldehyde-binding protein 1), RPE65 (retinal pigment epithelium-specific 65 kDa protein), PAX6 (transcription factor PAX6), SOX9 (transcription factor SOX9), DCT (dopachrome tautomerase) and MITF (microphthalmia-associated transcription factor). ICC of the RPEs cultured on nanofibrous membranes showed more intensive staining of markers such as BEST1, MCT1 (monocarboxylate transporter 1), Na+ /K+ ATPase, RPE65 and acetylated tubulin in comparison with commercial ones. Additionally, the absence of α-SMA proved the stability of the RPE polarization state and the absence of epithelial-to-mesenchymal transition. RPE possessed high phagocytic activity. Electron microscopy of both membranes confirmed a confluent layer of RPE cells and their genuine morphological structure, which was comparable to native RPEs. CONCLUSIONS: Retinal pigment epitheliums cultured on polylactide nanofibrous membranes improved the final quality of the cell product by having better maturation and long-term survival of the RPE monolayer compared to those cultured on commercial polyester scaffolds. PDLLA-cultured RPEs are a plausible source for the replacement of non-functioning RPEs during cell therapy.


Subject(s)
Nanofibers , Retinal Degeneration , Animals , Bestrophins/metabolism , Cells, Cultured , Nanofibers/chemistry , Polyesters/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Swine
2.
Int J Mol Sci ; 22(4)2021 02 20.
Article in English | MEDLINE | ID: mdl-33672445

ABSTRACT

Hereditary retinal dystrophies (HRD) represent a significant cause of blindness, affecting mostly retinal pigment epithelium (RPE) and photoreceptors (PRs), and currently suffer from a lack of effective treatments. Highly specialized RPE and PR cells interact mutually in the functional retina, therefore primary HRD affecting one cell type leading to a secondary HRD in the other cells. Phagocytosis is one of the primary functions of the RPE and studies have discovered that mutations in the phagocytosis-associated gene Mer tyrosine kinase receptor (MERTK) lead to primary RPE dystrophy. Treatment strategies for this rare disease include the replacement of diseased RPE with healthy autologous RPE to prevent PR degeneration. The generation and directed differentiation of patient-derived human-induced pluripotent stem cells (hiPSCs) may provide a means to generate autologous therapeutically-relevant adult cells, including RPE and PR. However, the continued presence of the MERTK gene mutation in patient-derived hiPSCs represents a significant drawback. Recently, we reported the generation of a hiPSC model of MERTK-associated Retinitis Pigmentosa (RP) that recapitulates disease phenotype and the subsequent creation of gene-corrected RP-hiPSCs using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9. In this study, we differentiated gene-corrected RP-hiPSCs into RPE and found that these cells had recovered both wild-type MERTK protein expression and the lost phagocytosis of fluorescently-labeled photoreceptor outer segments observed in uncorrected RP-hiPSC-RPE. These findings provide proof-of-principle for the utility of gene-corrected hiPSCs as an unlimited cell source for personalized cell therapy of rare vision disorders.


Subject(s)
Gene Editing , Induced Pluripotent Stem Cells/pathology , Phagocytosis , Retinal Pigment Epithelium/pathology , Retinitis Pigmentosa/pathology , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/ultrastructure , Mutation/genetics , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/pathology , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Retinal Pigment Epithelium/ultrastructure , Retinitis Pigmentosa/genetics , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism
3.
Prog Neurobiol ; 193: 101817, 2020 10.
Article in English | MEDLINE | ID: mdl-32360241

ABSTRACT

Glaucoma, one of the most common causes of blindness in developing countries today, involves a progressive loss of neural cells in the optic nerve that leads to progressive, irreversible vision loss. Increased intraocular pressure (IOP) presents as a major risk factor for glaucoma, although there exist cases of glaucoma patients with normal IOP that exhibit damage to retinal ganglion cells (RGCs) and the optic nerve. However, treatment approaches have maintained their focus on modifying IOP due to a lack of other modifiable risks factors. Traditional concepts in glaucoma involve the neuronal environment and external effects as a source of causative factors; however, studies have yet to investigate whether the molecular profile of RGCs in glaucoma patients makes them more vulnerable and/or susceptible to external damage. Our hypothesis states that molecular changes at the whole cell, gene expression, and electrophysiological level of the neurons can contribute to their degeneration. Herein, we briefly describe different types of glaucoma and any similarities to different molecular and cellular features of neurodegeneration. To test our hypothesis, we describe human induced pluripotent stem cells (hiPSCs) as a reliable cellular tool to model neurodegenerative aspects of glaucoma to reveal the multiple pathological molecular mechanisms underlying disease development.


Subject(s)
Genetic Predisposition to Disease , Glaucoma , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Retinal Ganglion Cells , Glaucoma/etiology , Glaucoma/genetics , Glaucoma/metabolism , Glaucoma/pathology , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
4.
Sci Rep ; 10(1): 5426, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214115

ABSTRACT

Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a photoreceptor-specific chaperone that stabilizes the effector enzyme of phototransduction, cGMP phosphodiesterase 6 (PDE6). Mutations in the AIPL1 gene cause a severe inherited retinal dystrophy, Leber congenital amaurosis type 4 (LCA4), that manifests as the loss of vision during the first year of life. In this study, we generated three-dimensional (3D) retinal organoids (ROs) from human induced pluripotent stem cells (hiPSCs) derived from an LCA4 patient carrying a Cys89Arg mutation in AIPL1. This study aimed to (i) explore whether the patient hiPSC-derived ROs recapitulate LCA4 disease phenotype, and (ii) generate a clinically relevant resource to investigate the molecular mechanism of disease and safely test novel therapies for LCA4 in vitro. We demonstrate reduced levels of the mutant AIPL1 and PDE6 proteins in patient organoids, corroborating the findings in animal models; however, patient-derived organoids maintained retinal cell cytoarchitecture despite significantly reduced levels of AIPL1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Eye Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Retina/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/metabolism , Cell Line , Eye Proteins/genetics , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Mutation/genetics
5.
Stem Cells ; 37(12): 1496-1504, 2019 12.
Article in English | MEDLINE | ID: mdl-31617949

ABSTRACT

Three-dimensional (3D) retinal organoids, in vitro tissue structures derived from self-organizing cultures of differentiating human embryonic stem cells or induced pluripotent stem cells, could recapitulate some aspects of the cytoarchitectural structure and function of the retina in vivo. 3D retinal organoids display huge potential for the investigation of the pathogenesis of monogenic hereditary eye diseases that are related to the malfunction or degeneration of photoreceptors or retinal ganglion cells by providing an effective in vitro tool with multiple applications. In combination with recent genome editing tools, 3D retinal organoids could also represent a reliable and renewable source of transplantable cells for personalized therapies. In this review, we describe the recent advances in human pluripotent stem cells-derived retinal organoids, determination of their histoarchitecture, complexity, and maturity. We also discuss their application as a means to decipher the pathogenesis of retinal diseases, as well as the main drawbacks and challenges. Stem Cells 2019;37:1496-1504.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Retina/pathology , Retinal Diseases/pathology , Retinal Rod Photoreceptor Cells/cytology , Gene Editing/methods , Induced Pluripotent Stem Cells/transplantation , Organoids/ultrastructure , Retina/ultrastructure
6.
Stem Cell Res ; 34: 101341, 2019 01.
Article in English | MEDLINE | ID: mdl-30612079

ABSTRACT

The human induced pluripotent stem cell (hiPSC) line RP1-FiPS4F1 generated from the patient with autosomal recessive retinitis pigmentosa (arRP) caused by homozygous Ser331Cysfs*5 mutation in Mer tyrosine kinase receptor (MERTK) was genetically corrected using CRISPR/Cas9 system. Two isogenic hiPSCs lines, with heterozygous and homozygous correction of c.992_993delCA mutation in the MERTK gene were generated. These cell lines demonstrate normal karyotype, maintain a pluripotent state, and can differentiate toward three germ layers in vitro. These genetically corrected hiPSCs represent accurate controls to study the contribution of the specific genetic change to the disease, and potentially therapeutic material for cell-replacement therapy.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Retinitis Pigmentosa/pathology , Targeted Gene Repair , c-Mer Tyrosine Kinase/genetics , Base Sequence , Cell Line , Humans
7.
Expert Opin Drug Discov ; 14(2): 169-177, 2019 02.
Article in English | MEDLINE | ID: mdl-30616395

ABSTRACT

INTRODUCTION: Combinatory strategies using pharmacology and stem cell therapy have emerged due to their potential in the treatment of retinal pigment epithelium (RPE) cell related diseases, and a variety of different stem cell sources have been evaluated both in animal models and in humans. RPE cells derived from human embryonic stem cells (hESCs) and human induced pluripotent cells (hiPSCs) are already in clinical trials, holding great promise for the treatment of age-related macular disease (AMD) and hereditary RPE-related retinal dystrophies. Highly efficient protocol for RPE generations have been developed, but they are still time-consuming and laborious. Areas covered: The authors review RPE related diseases, as well as the known functions of RPE cells in retinal homeostasis. The authors also discuss small molecules that target RPE in vivo as well as in vitro to aid RPE differentiation from pluripotent stem cells clinically. The authors base this review on literature searches performed through PubMed. Expert opinion: Using high-throughput systems, technology will provide the possibility of identifying and optimizing molecules/drugs that could lead to faster and simpler protocols for RPE differentiation. This could be crucial in moving forward to create safer and more efficient RPE-based personalized therapies.


Subject(s)
Macular Degeneration/therapy , Retinal Diseases/therapy , Retinal Pigment Epithelium/cytology , Animals , Cell Differentiation/physiology , Combined Modality Therapy , High-Throughput Screening Assays , Humans , Macular Degeneration/physiopathology , Pluripotent Stem Cells/cytology , Retinal Diseases/physiopathology , Stem Cell Transplantation/methods
8.
Stem Cell Res ; 33: 166-170, 2018 12.
Article in English | MEDLINE | ID: mdl-30384130

ABSTRACT

The human iPSC cell line, CARS-FiPS4F1 (ESi064-A), derived from dermal fibroblast from the apparently healthy carrier of the mutation of the gene SACSIN, was generated by non-integrative reprogramming technology using OCT3/4, SOX2, CMYC and KLF4 reprogramming factors. The pluripotency was assessed by immunocytochemistry and RT-PCR. This iPSC line can be used as control for Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) disease.


Subject(s)
Heat-Shock Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Adult , Female , Humans , Kruppel-Like Factor 4 , Mutation
9.
Stem Cell Res ; 33: 151-155, 2018 12.
Article in English | MEDLINE | ID: mdl-30366342

ABSTRACT

The human induced pluripotent stem cell (hiPSC) line, derived from dermal fibroblasts from Leber congenital amaurosis patient with homozygous mutation c.265 T > C, p.Cys89Arg in aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) was generated by Sendai virus reprogramming. The generated hiPSC line was free of Sendai virus genes, had stable karyotype, carried the homozygous mutation, was immunopositive to pluripotency markers and able to generate all three germ layers upon embryoid body formation. Resource table.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Leber Congenital Amaurosis/genetics , Adaptor Proteins, Signal Transducing , Adult , Female , Humans , Mutation
12.
Stem Cells ; 36(4): 474-481, 2018 04.
Article in English | MEDLINE | ID: mdl-29345014

ABSTRACT

Hereditary retinal dystrophies, specifically retinitis pigmentosa (RP) are clinically and genetically heterogeneous diseases affecting primarily retinal cells and retinal pigment epithelial cells with blindness as a final outcome. Understanding the pathogenicity behind these diseases has been largely precluded by the unavailability of affected tissue from patients, large genetic heterogeneity and animal models that do not faithfully represent some human diseases. A landmark discovery of human induced pluripotent stem cells (hiPSCs) permitted the derivation of patient-specific cells. These cells have unlimited self-renewing capacity and the ability to differentiate into RP-affected cell types, allowing the studies of disease mechanism, drug discovery, and cell replacement therapies, both as individual cell types and organoid cultures. Together with precise genome editing, the patient specific hiPSC technology offers novel strategies for targeting the pathogenic mutations and design therapies toward retinal dystrophies. This study summarizes current hiPSC-based RP models and highlights key achievements and challenges of these cellular models, as well as questions that still remain unanswered. Stem Cells 2018;36:474-481.


Subject(s)
Cell Differentiation , Gene Editing , Genome, Human , Induced Pluripotent Stem Cells/metabolism , Retinitis Pigmentosa , Stem Cell Transplantation , Animals , Autografts , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/therapy
13.
Stem Cell Res ; 21: 23-25, 2017 05.
Article in English | MEDLINE | ID: mdl-28677533

ABSTRACT

The human iPSC cell line, RP2-FiPS4F1 (RCPFi001-A), derived from dermal fibroblasts from the patient with retinitis pigmentosa caused by the mutation of the gene PRPF8, was generated by non-integrative reprogramming technology using OCT3/4, SOX2, CMYC and KLF4 reprogramming factors.


Subject(s)
Dermis/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , RNA-Binding Proteins , Retinitis Pigmentosa/metabolism , Cell Line , Cellular Reprogramming Techniques , Dermis/pathology , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Kruppel-Like Factor 4 , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
14.
Sci Rep ; 5: 12910, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26263531

ABSTRACT

Retinitis pigmentosa (RP) represents a genetically heterogeneous group of retinal dystrophies affecting mainly the rod photoreceptors and in some instances also the retinal pigment epithelium (RPE) cells of the retina. Clinical symptoms and disease progression leading to moderate to severe loss of vision are well established and despite significant progress in the identification of causative genes, the disease pathology remains unclear. Lack of this understanding has so far hindered development of effective therapies. Here we report successful generation of human induced pluripotent stem cells (iPSC) from skin fibroblasts of a patient harboring a novel Ser331Cysfs*5 mutation in the MERTK gene. The patient was diagnosed with an early onset and severe form of autosomal recessive RP (arRP). Upon differentiation of these iPSC towards RPE, patient-specific RPE cells exhibited defective phagocytosis, a characteristic phenotype of MERTK deficiency observed in human patients and animal models. Thus we have created a faithful cellular model of arRP incorporating the human genetic background which will allow us to investigate in detail the disease mechanism, explore screening of a variety of therapeutic compounds/reagents and design either combined cell and gene- based therapies or independent approaches.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Models, Biological , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Retinitis Pigmentosa/pathology , Animals , Case-Control Studies , Cell Differentiation , Humans , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Retinitis Pigmentosa/genetics , c-Mer Tyrosine Kinase
15.
Autophagy ; 11(9): 1499-519, 2015.
Article in English | MEDLINE | ID: mdl-26176264

ABSTRACT

The human ribosomal P complex, which consists of the acidic ribosomal P proteins RPLP0, RPLP1, and RPLP2 (RPLP proteins), recruits translational factors, facilitating protein synthesis. Recently, we showed that overexpression of RPLP1 immortalizes primary cells and contributes to transformation. Moreover, RPLP proteins are overexpressed in human cancer, with the highest incidence in breast carcinomas. It is thought that disruption of the P complex would directly affect protein synthesis, causing cell growth arrest and eventually apoptosis. Here, we report a distinct mechanism by which cancer cells undergo cell cycle arrest and induced autophagy when RPLP proteins are downregulated. We found that absence of RPLP0, RPLP1, or RPLP2 resulted in reactive oxygen species (ROS) accumulation and MAPK1/ERK2 signaling pathway activation. Moreover, ROS generation led to endoplasmic reticulum (ER) stress that involved the EIF2AK3/PERK-EIF2S1/eIF2α-EIF2S2-EIF2S3-ATF4/ATF-4- and ATF6/ATF-6-dependent arms of the unfolded protein response (UPR). RPLP protein-deficient cells treated with autophagy inhibitors experienced apoptotic cell death as an alternative to autophagy. Strikingly, antioxidant treatment prevented UPR activation and autophagy while restoring the proliferative capacity of these cells. Our results indicate that ROS are a critical signal generated by disruption of the P complex that causes a cellular response that follows a sequential order: first ROS, then ER stress/UPR activation, and finally autophagy. Importantly, inhibition of the first step alone is able to restore the proliferative capacity of the cells, preventing UPR activation and autophagy. Overall, our results support a role for autophagy as a survival mechanism in response to stress due to RPLP protein deficiency.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Multiprotein Complexes/metabolism , Ribosomal Proteins/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Female , HEK293 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Oxidation-Reduction , Phenotype , Protein Biosynthesis/drug effects , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Ribosomal Proteins/antagonists & inhibitors , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
16.
PLoS One ; 9(6): e99956, 2014.
Article in English | MEDLINE | ID: mdl-24959908

ABSTRACT

Ribosomal proteins are pivotal to development and tissue homeostasis. RP Large P1 (Rplp1) overexpression is associated with tumorigenesis. However, the physiological function of Rplp1 in mammalian development remains unknown. In this study, we disrupted Rplp1 in the mouse germline and central nervous system (Rplp1CNSΔ). Rplp1 heterozygosity caused body size reductions, male infertility, systemic abnormalities in various tissues and a high frequency of early postnatal death. Rplp1CNSΔ newborn mice exhibited perinatal lethality and brain atrophy with size reductions of the neocortex, midbrain and ganglionic eminence. The Rplp1 knockout neocortex exhibited progenitor cell proliferation arrest and apoptosis due to the dysregulation of key cell cycle and apoptosis regulators (cyclin A, cyclin E, p21CIP1, p27KIP1, p53). Similarly, Rplp1 deletion in pMEFs led to proliferation arrest and premature senescence. Importantly, Rplp1 deletion in primary mouse embryonic fibroblasts did not alter global protein synthesis, but did change the expression patterns of specific protein subsets involved in protein folding and the unfolded protein response, cell death, protein transport and signal transduction, among others. Altogether, we demonstrated that the translation "fine-tuning" exerted by Rplp1 is essential for embryonic and brain development and for proper cell proliferation.


Subject(s)
Congenital Abnormalities/etiology , Fibroblasts/cytology , Gene Deletion , Nervous System/embryology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Animals , Animals, Newborn , Body Size , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Gene Knockout Techniques , Genes, Essential , Genes, Lethal , Infertility, Male , Male , Mice , Nervous System/pathology
17.
PLoS One ; 8(10): e76247, 2013.
Article in English | MEDLINE | ID: mdl-24098452

ABSTRACT

MicroRNAs (miRNAs) play important roles in diverse biological processes and are emerging as key regulators of tumorigenesis and tumor progression. To explore the dysregulation of miRNAs in breast cancer, a genome-wide expression profiling of 939 miRNAs was performed in 50 breast cancer patients. A total of 35 miRNAs were aberrantly expressed between breast cancer tissue and adjacent normal breast tissue and several novel miRNAs were identified as potential oncogenes or tumor suppressor miRNAs in breast tumorigenesis. miR-125b exhibited the largest decrease in expression. Enforced miR-125b expression in mammary cells decreased cell proliferation by inducing G2/M cell cycle arrest and reduced anchorage-independent cell growth of cells of mammary origin. miR-125b was found to perform its tumor suppressor function via the direct targeting of the 3'-UTRs of ENPEP, CK2-α, CCNJ, and MEGF9 mRNAs. Silencing these miR-125b targets mimicked the biological effects of miR-125b overexpression, confirming that they are modulated by miR-125b. Analysis of ENPEP, CK2-α, CCNJ, and MEGF9 protein expression in breast cancer patients revealed that they were overexpressed in 56%, 40-56%, 20%, and 32% of the tumors, respectively. The expression of ENPEP and CK2-α was inversely correlated with miR-125b expression in breast tumors, indicating the relevance of these potential oncogenic proteins in breast cancer patients. Our results support a prognostic role for CK2-α, whose expression may help clinicians predict breast tumor aggressiveness. In particular, our results show that restoration of miR-125b expression or knockdown of ENPEP, CK2-α, CCNJ, or MEGF9 may provide novel approaches for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Casein Kinase II/genetics , Cell Transformation, Neoplastic/genetics , Cyclins/genetics , Genes, Tumor Suppressor , Glutamyl Aminopeptidase/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , 3' Untranslated Regions , Breast Neoplasms/metabolism , Casein Kinase II/metabolism , Cell Line , Cell Proliferation , Cluster Analysis , Cyclins/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glutamyl Aminopeptidase/metabolism , Humans , Membrane Proteins/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , RNA Interference
18.
Hum Pathol ; 42(2): 194-203, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21040949

ABSTRACT

Previous work from our laboratory has demonstrated that the expression of the ribosomal protein Rplp1 immortalizes primary cells and is involved in transformation. To investigate the role of the P proteins in tumorigenesis, we examined the messenger RNA expression levels of Rplp0, Rplp1, and Rplp2 in a series of 32 patients with gynecologic tumors. The messenger RNA expression level of all 3 P proteins was increased significantly in the tumor tissue, compared with normal tissue. In addition, a total of 140 biopsies of gynecologic cancers (46 endometrioid and 94 ovarian) were investigated. An up-regulation of P protein expression was observed by immunohistochemistry in an average of 27% of the tumors, as compared with normal tissues. Moreover, the level of P protein up-regulation correlated significantly with p53 expression in serous ovarian cancers. This is an important fact because the level of overexpression of the P proteins correlated with the presence of lymph node metastases in serous ovarian cancers. We also observed that endometrial carcinomas that had invaded the myometrium overexpressed P proteins in the invasive front. In addition, we found that the P proteins are up-regulated in a considerable number of patients with the most common types of cancer. Overall, our study shows that P proteins are involved in human cancer and indicates that the expression level of these proteins could be useful as a prognostic marker in specific subtypes of gynecologic tumors.


Subject(s)
Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Phosphoproteins/genetics , Ribosomal Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , DNA, Neoplasm/analysis , Endometrial Neoplasms/pathology , Female , Humans , Male , Ovarian Neoplasms/pathology , Prognosis , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
19.
Mol Cancer ; 8: 3, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-19133111

ABSTRACT

Cellular immortalization is a crucial step during the development of human cancer. Primary mammalian cells reach replicative exhaustion after several passages in vitro, a process called replicative senescence. During such a state of permanent growth arrest, senescent cells are refractory to physiological proliferation stimuli: they have altered cell morphology and gene expression patterns, although they remain viable with preserved metabolic activity. Interestingly, senescent cells have also been detected in vivo in human tumors, particularly in benign lesions. Senescence is a mechanism that limits cellular lifespan and constitutes a barrier against cellular immortalization. During immortalization, cells acquire genetic alterations that override senescence. Tumor suppressor genes and oncogenes are closely involved in senescence, as their knockdown and ectopic expression confer immortality and senescence induction, respectively. By using high throughput genetic screening to search for genes involved in senescence, several candidate oncogenes and putative tumor suppressor genes have been recently isolated, including subtypes of micro-RNAs. These findings offer new perspectives in the modulation of senescence and open new approaches for cancer therapy.


Subject(s)
Cellular Senescence/genetics , Neoplasms/genetics , Animals , Humans
20.
Mol Cell Biol ; 29(7): 1855-68, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158277

ABSTRACT

Embryonic stem cells are immortalized cells whose proliferation rate is comparable to that of carcinogenic cells. To study the expression of embryonic stem cell genes in primary cells, genetic screening was performed by infecting mouse embryonic fibroblasts (MEFs) with a cDNA library from embryonic stem cells. Cold-inducible RNA-binding protein (CIRP) was identified due to its ability to bypass replicative senescence in primary cells. CIRP enhanced extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation, and treatment with an MEK inhibitor decreased the proliferation caused by CIRP. In contrast to CIRP upregulation, CIRP downregulation decreased cell proliferation and resulted in inhibition of phosphorylated ERK1/2 inhibition. This is the first evidence that ERK1/2 activation, through the same mechanism as that described for a Val12 mutant K-ras to induce premature senescence, is able to bypass senescence in the absence of p16(INK4a), p21(WAF1), and p19(ARF) upregulation. Moreover, these results show that CIRP functions by stimulating general protein synthesis with the involvement of the S6 and 4E-BP1 proteins. The overall effect is an increase in kinase activity of the cyclin D1-CDK4 complex, which is in accordance with the proliferative capacity of CIRP MEFs. Interestingly, CIRP mRNA and protein were upregulated in a subgroup of cancer patients, a finding that may be of relevance for cancer research.


Subject(s)
Cellular Senescence , Mitogen-Activated Protein Kinase 1/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Line, Transformed , Cell Proliferation , Down-Regulation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/enzymology , Neoplasms/genetics , Phosphorylation , Protein Biosynthesis , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...