Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Neuron ; 112(12): 1943-1958.e10, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38697112

ABSTRACT

Mutations in the methyl-DNA-binding protein MECP2 cause the neurodevelopmental disorder Rett syndrome (RTT). How MECP2 contributes to transcriptional regulation in normal and disease states is unresolved; it has been reported to be an activator and a repressor. We describe here the first integrated CUT&Tag, transcriptome, and proteome analyses using human neurons with wild-type (WT) and mutant MECP2 molecules. MECP2 occupies CpG-rich promoter-proximal regions in over four thousand genes in human neurons, including a plethora of autism risk genes, together with RNA polymerase II (RNA Pol II). MECP2 directly interacts with RNA Pol II, and genes occupied by both proteins showed reduced expression in neurons with MECP2 patient mutations. We conclude that MECP2 acts as a positive cofactor for RNA Pol II gene expression at many neuronal genes that harbor CpG islands in promoter-proximal regions and that RTT is due, in part, to the loss of gene activity of these genes in neurons.


Subject(s)
Methyl-CpG-Binding Protein 2 , Neurons , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Humans , Neurons/metabolism , Promoter Regions, Genetic , Rett Syndrome/genetics , Rett Syndrome/metabolism , CpG Islands/genetics , Mutation , Gene Expression Regulation/genetics
2.
bioRxiv ; 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37790372

ABSTRACT

L-type Ca 2+ channels (Ca V 1.2/1.3) convey influx of calcium ions (Ca 2+ ) that orchestrate a bevy of biological responses including muscle contraction and gene transcription. Deficits in Ca V 1 function play a vital role in cardiac and neurodevelopmental disorders. Yet conventional pharmacological approaches to upregulate Ca V 1 are limited, as excessive Ca 2+ influx leads to cytotoxicity. Here, we develop a genetically encoded enhancer of Ca V 1.2/1.3 channels (GeeC) to manipulate Ca 2+ entry in distinct physiological settings. Specifically, we functionalized a nanobody that targets the Ca V macromolecular complex by attaching a minimal effector domain from a Ca V enhancer-leucine rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeC evoked a 3-fold increase in L-type current amplitude. In neurons, GeeC augmented excitation-transcription (E-T) coupling. In all, GeeC represents a powerful strategy to boost Ca V 1.2/1.3 function in distinct physiological settings and, in so doing, lays the groundwork to illuminate new insights on neuronal and cardiac physiology and disease.

3.
Sci Transl Med ; 15(679): eadd4666, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36652535

ABSTRACT

Rett syndrome (RTT) is an X-linked neurodevelopmental disorder caused by loss-of-function heterozygous mutations of methyl CpG-binding protein 2 (MECP2) on the X chromosome in young females. Reactivation of the silent wild-type MECP2 allele from the inactive X chromosome (Xi) represents a promising therapeutic opportunity for female patients with RTT. Here, we applied a multiplex epigenome editing approach to reactivate MECP2 from Xi in RTT human embryonic stem cells (hESCs) and derived neurons. Demethylation of the MECP2 promoter by dCas9-Tet1 with target single-guide RNA reactivated MECP2 from Xi in RTT hESCs without detectable off-target effects at the transcriptional level. Neurons derived from methylation-edited RTT hESCs maintained MECP2 reactivation and reversed the smaller soma size and electrophysiological abnormalities, two hallmarks of RTT. In RTT neurons, insulation of the methylation-edited MECP2 locus by dCpf1-CTCF (a catalytically dead Cpf1 fused with CCCTC-binding factor) with target CRISPR RNA enhanced MECP2 reactivation and rescued RTT-related neuronal defects, providing a proof-of-concept study for epigenome editing to treat RTT and potentially other dominant X-linked diseases.


Subject(s)
Rett Syndrome , Humans , Female , Rett Syndrome/genetics , Rett Syndrome/therapy , Epigenome , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , Mutation , Heterozygote , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/therapeutic use , Proto-Oncogene Proteins/metabolism
4.
Biol Psychiatry ; 93(1): 71-81, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36372569

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is characterized by physical abnormalities, anxiety, intellectual disability, hyperactivity, autistic behaviors, and seizures. Abnormal neuronal development in FXS is poorly understood. Data on patients with FXS remain scarce, and FXS animal models have failed to yield successful therapies. In vitro models do not fully recapitulate the morphology and function of human neurons. METHODS: To mimic human neuron development in vivo, we coinjected neural precursor cells derived from FXS patient-derived induced pluripotent stem cells and neural precursor cells derived from corrected isogenic control induced pluripotent stem cells into the brain of neonatal immune-deprived mice. RESULTS: The transplanted cells populated the brain and a proportion differentiated into neurons and glial cells. Immunofluorescence and single and bulk RNA sequencing analyses showed accelerated maturation of FXS neurons after an initial delay. Additionally, we found increased percentages of Arc- and Egr-1-positive FXS neurons and wider dendritic protrusions of mature FXS striatal medium spiny neurons. CONCLUSIONS: This transplantation approach provides new insights into the alterations of neuronal development in FXS by facilitating physiological development of cells in a 3-dimensional context.


Subject(s)
Fragile X Syndrome , Neural Stem Cells , Humans , Mice , Animals , Fragile X Syndrome/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Phenotype , Brain/metabolism , Mice, Knockout
5.
Commun Biol ; 5(1): 1009, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36163262

ABSTRACT

The hippocampus is involved in processing a variety of mnemonic computations specifically the spatiotemporal components and emotional dimensions of contextual memory. Recent studies have demonstrated cellular heterogeneity along the hippocampal axis. The ventral hippocampus has been shown to be important in the processing of emotion and valence. Here, we combine transgenic and all-virus based activity-dependent tagging strategies to visualize multiple valence-specific engrams in the vHPC and demonstrate two partially segregated cell populations and projections that respond to appetitive and aversive experiences. Next, using RNA sequencing and DNA methylation sequencing approaches, we find that vHPC appetitive and aversive engram cells display different transcriptional programs and DNA methylation landscapes compared to a neutral engram population. Additionally, optogenetic manipulation of tagged cell bodies in vHPC is not sufficient to drive appetitive or aversive behavior in real-time place preference, stimulation of tagged vHPC terminals projecting to the amygdala and nucleus accumbens (NAc), but not the prefrontal cortex (PFC), showed the capacity drive preference and avoidance. These terminals also were able to change their capacity to drive behavior. We conclude that the vHPC contains genetically, cellularly, and behaviorally segregated populations of cells processing appetitive and aversive memory engrams.


Subject(s)
Hippocampus , Memory , Amygdala/physiology , Hippocampus/physiology , Memory/physiology , Optogenetics , Prefrontal Cortex/physiology
6.
Nature ; 586(7829): 440-444, 2020 10.
Article in English | MEDLINE | ID: mdl-32698189

ABSTRACT

Methyl CpG binding protein 2 (MeCP2) is a key component of constitutive heterochromatin, which is crucial for chromosome maintenance and transcriptional silencing1-3. Mutations in the MECP2 gene cause the progressive neurodevelopmental disorder Rett syndrome3-5, which is associated with severe mental disability and autism-like symptoms that affect girls during early childhood. Although previously thought to be a dense and relatively static structure1,2, heterochromatin is now understood to exhibit properties consistent with a liquid-like condensate6,7. Here we show that MeCP2 is a dynamic component of heterochromatin condensates in cells, and is stimulated by DNA to form liquid-like condensates. MeCP2 contains several domains that contribute to the formation of condensates, and mutations in MECP2 that lead to Rett syndrome disrupt the ability of MeCP2 to form condensates. Condensates formed by MeCP2 selectively incorporate and concentrate heterochromatin cofactors rather than components of euchromatic transcriptionally active condensates. We propose that MeCP2 enhances the separation of heterochromatin and euchromatin through its condensate partitioning properties, and that disruption of condensates may be a common consequence of mutations in MeCP2 that cause Rett syndrome.


Subject(s)
Heterochromatin/metabolism , Intellectual Disability/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mutation , Adaptive Immunity , Animals , Female , Immunity, Innate , Intellectual Disability/pathology , Methyl-CpG-Binding Protein 2/genetics , Mice , Neurons/metabolism , Neurons/pathology , Phenotype , Rett Syndrome/genetics
7.
Trends Neurosci ; 42(12): 861-870, 2019 12.
Article in English | MEDLINE | ID: mdl-31706628

ABSTRACT

Genetic studies of epigenetic modifiers such as DNA methyltransferases and histone acetyltransferases have revealed a critical role for epigenetic regulation during brain development and function. Alteration of epigenetic modifications have been documented in a variety of brain disorders, including neurodevelopmental, psychiatric, and neurodegenerative diseases. Development of epigenome editing tools enables a functional dissection of the link between altered epigenetic changes and disease outcomes. Here, we review the development of epigenome editing tools, summarize proof of concept applications focusing on brain disease-associated genes, and discuss the promising application and challenges of epigenome editing to tackle brain disorders.


Subject(s)
Brain Diseases/genetics , CRISPR-Cas Systems , Epigenesis, Genetic , Epigenomics/methods , Gene Editing/methods , Animals , DNA Methylation , Humans
8.
Hand (N Y) ; 14(6): 836-840, 2019 11.
Article in English | MEDLINE | ID: mdl-29998773

ABSTRACT

Background: Postoperative care is essential to optimizing patient outcome. We sought to determine the incidence and associated demographic and surgical factors of postoperative patient loss to follow-up following hand and upper extremity surgery. Methods: In all, 2834 surgical cases (2467 patients) were retrospectively reviewed. All surgical cases from July 2014 to June 2015 at a single practice with five surgeons were assessed. Charts were reviewed for compliance with postoperative follow-up. Variables were described with proportions and compared using logistic regression analysis. Results: In total, 2563 cases (2388 patients) met the inclusion criteria. Overall loss to follow-up rate was 26%. Patients lost to follow-up based on insurance type were 13% for worker's compensation, 22% for private insurance, 21% for Medicare, 38% for Medicaid, and 44% for self-pay. Patients with expected short-term follow-up were lost at a 23% rate. Expected mid- and long-term follow-up patients were lost at 34% and 20% rates, respectively. Patients below 30 years old were lost to follow-up at a 42% rate compared to patients 30 to 64 years old (26%) and greater than or equal to 65 years (13%). Males had a higher rate of loss to follow-up, 32%, compared with females (22%). Patients living greater than 50 miles from our surgery center were lost to follow-up at a rate of 31%, compared with those who lived less than 50 miles (25%). Conclusions: We have identified demographic variables associated with patients being lost to follow-up after hand and upper extremity surgery. With this knowledge, we hope to develop methods of either improving in-office follow-up rates or discover new avenues to deliver postoperative care.


Subject(s)
Lost to Follow-Up , Orthopedic Procedures/statistics & numerical data , Upper Extremity/surgery , Adult , Aged , Female , Humans , Insurance, Health/statistics & numerical data , Logistic Models , Male , Medicaid/statistics & numerical data , Medicare/statistics & numerical data , Middle Aged , Retrospective Studies , United States/epidemiology , Workers' Compensation/statistics & numerical data
9.
Cell ; 172(5): 979-992.e6, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29456084

ABSTRACT

Fragile X syndrome (FXS), the most common genetic form of intellectual disability in males, is caused by silencing of the FMR1 gene associated with hypermethylation of the CGG expansion mutation in the 5' UTR of FMR1 in FXS patients. Here, we applied recently developed DNA methylation editing tools to reverse this hypermethylation event. Targeted demethylation of the CGG expansion by dCas9-Tet1/single guide RNA (sgRNA) switched the heterochromatin status of the upstream FMR1 promoter to an active chromatin state, restoring a persistent expression of FMR1 in FXS iPSCs. Neurons derived from methylation-edited FXS iPSCs rescued the electrophysiological abnormalities and restored a wild-type phenotype upon the mutant neurons. FMR1 expression in edited neurons was maintained in vivo after engrafting into the mouse brain. Finally, demethylation of the CGG repeats in post-mitotic FXS neurons also reactivated FMR1. Our data establish that demethylation of the CGG expansion is sufficient for FMR1 reactivation, suggesting potential therapeutic strategies for FXS.


Subject(s)
DNA Methylation/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Editing , Neurons/pathology , Animals , CRISPR-Associated Protein 9/metabolism , Epigenesis, Genetic , HEK293 Cells , Heterochromatin/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kinetics , Male , Mice , Neurons/metabolism , Phenotype , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida/metabolism , Trinucleotide Repeat Expansion/genetics
10.
Oncotarget ; 8(42): 73345-73361, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069873

ABSTRACT

BACKGROUND: Clinical and pathologic markers of prognosis and patterns of failure help guide clinicians in selecting patients for adjuvant therapy after surgical resection for pancreatic adenocarcinoma (PDAC). Recent studies have reported the prognostic utility of microRNA profiling in numerous malignancies. Here, we review and summarize the current literature regarding associations between microRNA expression and overall survival in PDAC patients. MATERIALS AND METHODS: We conducted a systematic search in the PubMed database to identify all primary research studies reporting prognostic associations between tumor and/or serum microRNA expression and overall survival in PDAC patients. Eligible articles were reviewed by the authors and relevant findings are summarized below. RESULTS: We found 53 publications that fit our search criteria. In total, 23 up-regulated and 49 down-regulated miRNAs have been associated with worse overall survival. MiR-21 is the most commonly reported miRNA, appearing in 19 publications, all of which report aberrant over-expression and association with shorter survival in PDAC. Other miRNAs that appear in multiple publications include miR-10b, -21, -34a, -155, -196a, -198, -200c, -203, -210, -218, -222, and -328. We summarize the preclinical and clinical data implicating these miRNAs in various molecular signaling pathways and cellular functions. CONCLUSIONS: There is growing evidence that miRNA expression profiles have the potential to provide tumor-specific prognostic information to assist clinicians in more appropriately selecting patients for adjuvant therapy. These molecules are often aberrantly expressed and exhibit oncogenic and/or tumor suppressor functions in PDAC. Additional efforts to develop prognostic and predictive molecular signatures, and further elucidate miRNA mechanisms of action, are warranted.

11.
Cell ; 167(1): 233-247.e17, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662091

ABSTRACT

Mammalian DNA methylation is a critical epigenetic mechanism orchestrating gene expression networks in many biological processes. However, investigation of the functions of specific methylation events remains challenging. Here, we demonstrate that fusion of Tet1 or Dnmt3a with a catalytically inactive Cas9 (dCas9) enables targeted DNA methylation editing. Targeting of the dCas9-Tet1 or -Dnmt3a fusion protein to methylated or unmethylated promoter sequences caused activation or silencing, respectively, of an endogenous reporter. Targeted demethylation of the BDNF promoter IV or the MyoD distal enhancer by dCas9-Tet1 induced BDNF expression in post-mitotic neurons or activated MyoD facilitating reprogramming of fibroblasts into myoblasts, respectively. Targeted de novo methylation of a CTCF loop anchor site by dCas9-Dnmt3a blocked CTCF binding and interfered with DNA looping, causing altered gene expression in the neighboring loop. Finally, we show that these tools can edit DNA methylation in mice, demonstrating their wide utility for functional studies of epigenetic regulation.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Editing/methods , Proto-Oncogene Proteins/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brain-Derived Neurotrophic Factor/genetics , CCCTC-Binding Factor , CRISPR-Associated Protein 9 , Cell Line , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endonucleases/metabolism , Enhancer Elements, Genetic , Genome , Mice , MyoD Protein/metabolism , Neurons/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 111(30): E3091-100, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024225

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is the most lethal of common human malignancies, with no truly effective therapies for advanced disease. Preclinical studies have suggested a therapeutic benefit of targeting the Hedgehog (Hh) signaling pathway, which is activated throughout the course of PDA progression by expression of Hh ligands in the neoplastic epithelium and paracrine response in the stromal fibroblasts. Clinical trials to test this possibility, however, have yielded disappointing results. To further investigate the role of Hh signaling in the formation of PDA and its precursor lesion, pancreatic intraepithelial neoplasia (PanIN), we examined the effects of genetic or pharmacologic inhibition of Hh pathway activity in three distinct genetically engineered mouse models and found that Hh pathway inhibition accelerates rather than delays progression of oncogenic Kras-driven disease. Notably, pharmacologic inhibition of Hh pathway activity affected the balance between epithelial and stromal elements, suppressing stromal desmoplasia but also causing accelerated growth of the PanIN epithelium. In striking contrast, pathway activation using a small molecule agonist caused stromal hyperplasia and reduced epithelial proliferation. These results indicate that stromal response to Hh signaling is protective against PDA and that pharmacologic activation of pathway response can slow tumorigenesis. Our results provide evidence for a restraining role of stroma in PDA progression, suggesting an explanation for the failure of Hh inhibitors in clinical trials and pointing to the possibility of a novel type of therapeutic intervention.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Mice , Mice, Knockout , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
13.
Mol Cancer Ther ; 12(1): 58-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23188630

ABSTRACT

Although gemcitabine is the standard chemotherapeutic drug for treatment of pancreatic cancer, almost all patients eventually develop resistance to this agent. Previous studies identified Polo-like kinase 1 (Plk1) as the mediator of gemcitabine resistance, but the molecular mechanism remains unknown. In this study, we show that Plk1 phosphorylation of Orc2 and Hbo1 mediates the resistance to gemcitabine. We show that the level of Plk1 expression positively correlates with gemcitabine resistance, both in pancreatic cancer cells and xenograft tumors. Overexpression of Plk1 increases gemcitabine resistance, while inhibition of Plk1 sensitizes pancreatic cancer cells to gemcitabine treatment. To validate our findings, we show that inhibition of Plk1 sensitizes tumors to gemcitabine treatment in a mouse xenograft study. Mechanistically, we find that Plk1 phosphorylation of Orc2 maintains DNA replication on gemcitabine treatment. Furthermore, Plk1 phosphorylation of Hbo1 transcriptionally increases cFos expression and consequently elevates its target multidrug resistance 1 (MDR1), which was previously reported to confer chemotherapeutic drug resistance. Knockdown of cFos or MDR1 sensitizes gemcitabine-resistant cells to gemcitabine treatment. Finally, pancreatic cancer cells expressing Plk1-unphosphorylatable mutants of Orc2 or Hbo1 are more sensitive to gemcitabine than cells expressing wild-type Orc2 or Hbo1. In short, our study provides a mechanism for Plk1-mediated gemcitabine resistance, suggesting that Plk1 is a promising target for treatment of gemcitabine-resistant pancreatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle Proteins/metabolism , Deoxycytidine/analogs & derivatives , Histone Acetyltransferases/metabolism , Origin Recognition Complex/metabolism , Pancreatic Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA Replication/drug effects , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Pteridines/pharmacology , Tissue Array Analysis , Xenograft Model Antitumor Assays , Gemcitabine , Polo-Like Kinase 1
14.
Mol Cell Biol ; 32(19): 4053-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22869522

ABSTRACT

Accurate chromosome segregation during cell division maintains genomic integrity and requires the proper establishment of kinetochore-microtubule attachment in mitosis. As a key regulator of mitosis, Polo-like kinase 1 (Plk1) is essential for this attachment process, but the molecular mechanism remains elusive. Here we identify Sgt1, a cochaperone for Hsp90, as a novel Plk1 substrate during mitosis. We show that Sgt1 dynamically localizes at the kinetochores, which lack microtubule attachments during prometaphase. Plk1 is required for the kinetochore localization of Sgt1 and phosphorylates serine 331 of Sgt1 at the kinetochores. This phosphorylation event enhances the association of the Hsp90-Sgt1 chaperone with the MIS12 complex to stabilize this complex at the kinetochores and thus coordinates the recruitment of the NDC80 complex to form efficient microtubule-binding sites. Disruption of Sgt1 phosphorylation reduces the MIS12 and NDC80 complexes at the kinetochores, impairs stable microtubule attachment, and eventually results in chromosome misalignment to delay the anaphase onset. Our results demonstrate a mechanism for Plk1 in promoting kinetochore-microtubule attachment to ensure chromosome stability.


Subject(s)
Cell Cycle Proteins/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/analysis , HEK293 Cells , HeLa Cells , Humans , Kinetochores/ultrastructure , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/analysis , Proto-Oncogene Proteins/analysis , Serine/metabolism , Polo-Like Kinase 1
15.
Development ; 139(16): 2857-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22764051

ABSTRACT

Microenvironmental oxygen (O(2)) regulates stem cell activity, and a hypoxic niche with low oxygen levels has been reported in multiple stem cell types. Satellite cells are muscle-resident stem cells that maintain the homeostasis and mediate the regeneration of skeletal muscles. We demonstrate here that hypoxic culture conditions favor the quiescence of satellite cell-derived primary myoblasts by upregulating Pax7, a key regulator of satellite cell self-renewal, and downregulating MyoD and myogenin. During myoblast division, hypoxia promotes asymmetric self-renewal divisions and inhibits asymmetric differentiation divisions without affecting the overall rate of proliferation. Mechanistic studies reveal that hypoxia activates the Notch signaling pathway, which subsequently represses the expression of miR-1 and miR-206 through canonical Hes/Hey proteins, leading to increased levels of Pax7. More importantly, hypoxia conditioning enhances the efficiency of myoblast transplantation and the self-renewal of implanted cells. Given the robust effects of hypoxia on maintaining the quiescence and promoting the self-renewal of cultured myoblasts, we predict that oxygen levels in the satellite cell niche play a central role in precisely balancing quiescence versus activation, and self-renewal versus differentiation, in muscle stem cells in vivo.


Subject(s)
Cell Hypoxia/physiology , Myoblasts, Skeletal/transplantation , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Animals , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred mdx , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , MyoD Protein/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/physiology , Myogenin/metabolism , PAX7 Transcription Factor/metabolism , Receptors, Notch/metabolism , Resting Phase, Cell Cycle , Signal Transduction , Stem Cell Niche/physiology
16.
Mol Cell Proteomics ; 11(9): 629-39, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22593177

ABSTRACT

With new discoveries of important roles of phosphorylation on a daily basis, phospho-specific antibodies, as the primary tool for on-membrane detection of phosphoproteins, face enormous challenges. To address an urgent need for convenient and reliable analysis of phosphorylation events, we report a novel strategy for sensitive phosphorylation analysis in the Western blotting format. The chemical reagent, which we termed pIMAGO, is based on a multifunctionalized soluble nanopolymer and is capable of selectively binding to phosphorylated residues independent of amino acid microenvironment, thus offering great promise as a universal tool in biological analyses where the site of phosphorylation is not known or its specific antibody is not available. The specificity and sensitivity of the approach was first examined using a mixture of standard proteins. The method was then applied to monitor phosphorylation changes in in vitro kinase and phosphatase assays. Finally, to demonstrate the unique ability of pIMAGO to measure endogenous phosphorylation, we used it to visualize and determine the differences in phosphorylated proteins that interact with wild-type and kinase dead mutant of Polo-like kinase 1 during mitosis, the results of which were further confirmed by a quantitative phosphoproteomics experiment.


Subject(s)
Cell Cycle Proteins/analysis , Membrane Proteins/analysis , Phosphoproteins/analysis , Protein Serine-Threonine Kinases/analysis , Proto-Oncogene Proteins/analysis , Antibodies/immunology , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Nanostructures , Phosphoproteins/immunology , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteome , Proteomics , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
17.
Cell Div ; 7: 3, 2012 Feb 06.
Article in English | MEDLINE | ID: mdl-22309699

ABSTRACT

Regulation of cell cycle progression is important for the maintenance of genome integrity, and Polo-like kinases (Plks) have been identified as key regulators of this process. It is well established that Polo-like kinase 1 (Plk1) plays critical roles in mitosis but little is known about its functions at other stages of the cell cycle. Here we summarize the functions of Plk1 during DNA replication, focusing on the molecular events related to Origin Recognition Complex (ORC), the complex that is essential for the initiation of DNA replication. Within the context of Plk1 phosphorylation of Orc2, we also emphasize regulation of Orc2 in different organisms. This review is intended to provide some insight into how Plk1 coordinates DNA replication in S phase with chromosome segregation in mitosis, and orchestrates the cell cycle as a whole.

18.
Aging (Albany NY) ; 3(9): 846-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21931181

ABSTRACT

Alzheimer's disease (AD) is a progressive and fatal brain disease, but the pathogenesis of AD is still not understood. Aberrant cell-cycle re-entry of neuronal cells is emerging as a potential pathological mechanism in AD. Polo-like kinase 1 (Plk1) is an established regulator of many cell cycle-related events. Interestingly, Plk1 is present in susceptible hippocampal and cortical neurons of AD patients but not age-matched controls. However, whether Plk1 is involved in the pathogenesis of AD remains elusive. In this study, we showed that Plk1 activity is elevated in AD patient brain as indicated by the increased phosphorylation signal of p150Glued, a Plk1-specific substrate. Furthermore, we demonstrated that Plk1 is elevated during the cell-cycle re-entry of neuronal cells in an in vitro cell-culture model. Significantly, inhibition of Plk1 kinase activity or depletion of Plk1 by RNAi reduces ß-amyloid (Aß)-induced neuronal cell death. These results validate Plk1 as a possible target for AD therapy.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Death/drug effects , Neurons/metabolism , Neurons/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Replication , Dynactin Complex , Humans , Microtubule-Associated Proteins/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Polo-Like Kinase 1
19.
Mol Cell Biol ; 31(23): 4844-56, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947279

ABSTRACT

Polo-like kinase 1 (Plk1) plays pivotal roles in mitosis; however, little is known about its function in S phase. In this study, we show that inhibition of Plk1 impairs DNA replication and results in slow S-phase progression in cultured cancer cells. We have identified origin recognition complex 2 (Orc2), a member of the DNA replication machinery, as a Plk1 substrate and have shown that Plk1 phosphorylates Orc2 at Ser188 in vitro and in vivo. Furthermore, Orc2-S188 phosphorylation is enhanced when DNA replication is under challenge induced by ultraviolet, hydroxyurea, gemcitabine, or aphidicolin treatment. Cells expressing the unphosphorylatable mutant (S188A) of Orc2 had defects in DNA synthesis under stress, suggesting that this phosphorylation event is critical to maintain DNA replication under stress. To dissect the mechanism pertinent to this observation, we showed that Orc2-S188 phosphorylation associates with DNA replication origin and that cells expressing Orc2-S188A mutant fail to maintain the functional pre-replicative complex (pre-RC) under DNA replication stress. Furthermore, the intra-S-phase checkpoint is activated in Orc2-S188A-expressing cells to cause delay of S-phase progress. Our study suggests a novel role of Plk1 in facilitating DNA replication under conditions of stress to maintain genomic integrity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , Origin Recognition Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stress, Physiological , Amino Acid Motifs , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Line , Chromatin Immunoprecipitation , Genomic Instability , Humans , Origin Recognition Complex/chemistry , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , S Phase , S Phase Cell Cycle Checkpoints , Polo-Like Kinase 1
20.
J Biol Chem ; 286(41): 35795-35800, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21890624

ABSTRACT

Loss of the tumor suppressor Pten (phosphatase and tensin homolog deleted on chromosome 10) is thought to mediate the majority of prostate cancers, but the molecular mechanism remains elusive. In this study, we demonstrate that Pten-depleted cells suffer from mitotic stress and that nuclear function of Pten, but not its phosphatase activity, is required to reverse this stress phenotype. Further, depletion of Pten results in elevated expression of Polo-like kinase 1 (Plk1), a critical regulator of the cell cycle. We show that overexpression of Plk1 correlates with genetic inactivation of Pten during prostate neoplasia formation. Significantly, we find that elevated Plk1 is critical for Pten-depleted cells to adapt to mitotic stress for survival and that reintroduction of wild-type Pten into Pten-null prostate cancer cells reduces the survival dependence on Plk1. We further show that Plk1 confers the tumorigenic competence of Pten-deleted prostate cancer cells in a mouse xenograft model. These findings identify a role of Plk1 in facilitating loss of Pten-induced prostate cancer formation, which suggests that Plk1 might be a promising target for prostate cancer patients with inactivating Pten mutations.


Subject(s)
Cell Cycle Proteins/metabolism , Mitosis , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/genetics , Gene Deletion , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Stress, Physiological/genetics , Transplantation, Heterologous , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...