Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
Cereb Cortex ; 30(5): 2939-2947, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31813988

ABSTRACT

Reduced cortical thickness has been demonstrated in psychotic disorders, but its relationship to clinical symptoms has not been established. We aimed to identify the regions throughout neocortex where clinical psychosis manifestations correlate with cortical thickness. Rather than perform a traditional correlation analysis using total scores on psychiatric rating scales, we applied multidimensional item response theory to identify a profile of psychotic symptoms that was related to a region where cortical thickness was reduced. This analysis was performed using a large population of probands with psychotic disorders (N = 865), their family members (N = 678) and healthy volunteers (N = 347), from the 5-site Bipolar-Schizophrenia Network for Intermediate Phenotypes. Regional cortical thickness from structural magnetic resonance scans was measured using FreeSurfer; individual symptoms were rated using the Positive and Negative Syndrome Scale, Montgomery-Asberg Depression Rating Scale, and Young Mania Rating Scale. A cluster of cortical regions whose thickness was inversely related to severity of psychosis symptoms was identified. The regions turned out to be located contiguously in a large region of heteromodal association cortex including temporal, parietal and frontal lobe regions, suggesting a cluster of contiguous neocortical regions important to psychosis expression. When we tested the relationship between reduced cortical surface area and high psychotic symptoms we found no linked regions describing a related cortical set.


Subject(s)
Magnetic Resonance Imaging/methods , Multidimensional Scaling Analysis , Neocortex/diagnostic imaging , Psychometrics/methods , Psychotic Disorders/diagnostic imaging , Adult , Female , Humans , Male , Middle Aged , Neocortex/physiopathology , Psychotic Disorders/physiopathology , Young Adult
2.
Int J Mol Sci ; 20(10)2019 May 16.
Article in English | MEDLINE | ID: mdl-31100813

ABSTRACT

The L-type calcium channel blocker fendiline has been shown to interfere with Ras-dependent signaling in K-Ras mutant cancer cells. Earlier studies from our lab had shown that treatment of pancreatic cancer cells with fendiline causes significant cytotoxicity and interferes with proliferation, survival, migration, invasion and anchorage independent growth. Currently there are no effective therapies to manage PDACs. As fendiline has been approved for treatment of patients with angina, we hypothesized that, if proven effective, combinatorial therapies using this agent would be easily translatable to clinic for testing in PDAC patients. Here we tested combinations of fendiline with gemcitabine, visudyne (a YAP1 inhibitor) or tivantinib (ARQ197, a c-Met inhibitor) for their effectiveness in overcoming growth and oncogenic characteristics of PDAC cells. The Hippo pathway component YAP1 has been shown to bypass K-Ras addiction, and allow tumor growth, in a Ras-null mouse model. Similarly, c-Met expression has been associated with poor prognosis and metastasis in PDAC patients. Our results presented here show that combinations of fendiline with these inhibitors show enhanced anti-tumor activity in Panc1, MiaPaCa2 and CD18/HPAF PDAC cells, as evident from the reduced viability, migration, anchorage-independent growth and self-renewal. Biochemical analysis shows that these agents interfere with various signaling cascades such as the activation of Akt and ERK, as well as the expression of c-Myc and CD44 that are altered in PDACs. These results imply that inclusion of fendiline may improve the efficacy of various chemotherapeutic agents that could potentially benefit PDAC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Fendiline/pharmacology , Pyrrolidinones/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Verteporfin/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogens , Cell Cycle/drug effects , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phosphoproteins/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , YAP-Signaling Proteins , Gemcitabine
3.
J Biol Chem ; 290(9): 5267-79, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25568312

ABSTRACT

KCl withdrawal-induced apoptosis in cerebellar granule neurons is associated with aberrant cell cycle activation, and treatment with cyclin-dependent kinase (Cdk) inhibitors protects cells from undergoing apoptosis. Because the Cdk inhibitor flavopiridol is known to inhibit RNA polymerase II (Pol II)-dependent transcription elongation by inhibiting the positive transcription elongation factor b (P-TEFb, a complex of CDK9 and cyclin T), we examined whether inhibition of RNA Pol II protects neurons from apoptosis. Treatment of neurons with 5, 6-dichloro-1-ß-D-ribobenzimidazole (DRB), an RNA Pol II-dependent transcription elongation inhibitor, and flavopiridol inhibited phosphorylation and activation of Pol II and protected neurons from undergoing apoptosis. In addition to Pol II, neurons subjected to KCl withdrawal showed increased phosphorylation and activation of p70 S6 kinase, which was inhibited by both DRB and flavopiridol. Immunostaining analysis of the neurons deprived of KCl showed increased nuclear levels of phospho-p70 S6 kinase, and neurons protected with DRB and flavopiridol showed accumulation of the kinase into large spliceosome assembly factor-positive speckle domains within the nuclei. The formation of these foci corresponded with cell survival, and removal of the inhibitors resulted in dispersal of the speckles into smaller foci with subsequent apoptosis induction. Because p70 S6 kinase is known to induce translation of mRNAs containing a 5'-terminal oligopyrimidine tract, our data suggest that transcription and translation of this subset of mRNAs may contribute to KCl withdrawal-induced apoptosis in neurons.


Subject(s)
Apoptosis , Flavonoids , Neurons/metabolism , Piperidines , RNA Polymerase II/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Blotting, Western , Cells, Cultured , Cerebellum/cytology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Immunohistochemistry , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Piperidines/pharmacology , Potassium Chloride/metabolism , Potassium Chloride/pharmacology , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transcription Elongation, Genetic/drug effects
4.
Adv Exp Med Biol ; 929: 209-228, 2016.
Article in English | MEDLINE | ID: mdl-27771926

ABSTRACT

This chapter describes the potential use of flavopiridol, a CDK inhibitor with anti-inflammatory and anti-proliferative activities, in the treatment of various chronic diseases. Flavopiridol arrests cell cycle progression in the G1 or G2 phase by inhibiting the kinase activities of CDK1, CDK2, CDK4/6, and CDK7. Additionally, it binds tightly to CDK9, a component of the P-TEFb complex (CDK9/cyclin T), and interferes with RNA polymerase II activation and associated transcription. This in turn inhibits expression of several pro-survival and anti-apoptotic genes, and enhances cytotoxicity in transformed cells or differentiation in growth-arrested cells. Recent studies indicate that flavopiridol elicits anti-inflammatory activity via CDK9 and NFκB-dependent signaling. Overall, these effects of flavopiridol potentiate its ability to overcome aberrant cell cycle activation and/or inflammatory stimuli, which are mediators of various chronic diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Antiviral Agents/therapeutic use , Cardiovascular Agents/therapeutic use , Chronic Disease/drug therapy , Drug Discovery/methods , Flavonoids/therapeutic use , Piperidines/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antiviral Agents/chemistry , Apoptosis/drug effects , Cardiovascular Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Flavonoids/chemistry , Humans , Molecular Structure , Phytotherapy , Piperidines/chemistry , Plants, Medicinal , Signal Transduction/drug effects
5.
Dev Psychopathol ; 27(2): 615-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25997775

ABSTRACT

In this paper, we review the history of the concept of neuroplasticity as it relates to the understanding of neuropsychiatric disorders, using schizophrenia as a case in point. We briefly review the myriad meanings of the term neuroplasticity, and its neuroscientific basis. We then review the evidence for aberrant neuroplasticity and metaplasticity associated with schizophrenia as well as the risk for developing this illness, and discuss the implications of such understanding for prevention and therapeutic interventions. We argue that the failure and/or altered timing of plasticity of critical brain circuits might underlie cognitive and deficit symptoms, and may also lead to aberrant plastic reorganization in other circuits, leading to affective dysregulation and eventually psychosis. This "dysplastic" model of schizophrenia can suggest testable etiology and treatment-relevant questions for the future.


Subject(s)
Neuronal Plasticity/physiology , Psychotic Disorders/physiopathology , Schizophrenia/physiopathology , Humans , Psychotic Disorders/etiology , Psychotic Disorders/psychology , Risk Factors , Schizophrenia/diagnosis , Schizophrenia/etiology , Schizophrenic Psychology
6.
J Biol Chem ; 288(42): 30114-30124, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24022491

ABSTRACT

Pancreatic adenocarcinoma or pancreatic cancer is often diagnosed at a very late stage at which point treatment options are minimal. Current chemotherapeutic interventions prolong survival marginally, thereby emphasizing the acute need for better treatment options to effectively manage this disease. Studies from different laboratories have shown that the Alzheimer disease-associated amyloid precursor protein (APP) is overexpressed in various cancers but its significance is not known. Here we sought to determine the role of APP in pancreatic cancer cell survival and proliferation. Our results show that pancreatic cancer cells secrete high levels of sAPPα, the α-secretase cleaved ectodomain fragment of APP, as compared with normal non-cancerous cells. Treatment of cells with batimastat or GI254023X, inhibitors of the α-secretase ADAM10, prevented sAPPα generation and reduced cell survival. Additionally, inhibition of sAPPα significantly reduced anchorage independent growth of the cancer cells. The effect of batimastat on cell survival and colony formation was enhanced when sAPPα downregulation was combined with gemcitabine treatment. Moreover, treatment of batimastat-treated cells with recombinant sAPPα reversed the inhibitory effect of the drug thereby indicating that sAPPα can indeed induce proliferation of cancer cells. Down-regulation of APP and ADAM10 brought about similar results, as did batimastat treatment, thereby confirming that APP processing is important for growth and proliferation of these cells. These results suggest that inhibition of sAPPα generation might enhance the effectiveness of the existing chemotherapeutic regimen for a better outcome.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation/drug effects , Cytotoxins/metabolism , Deoxycytidine/analogs & derivatives , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Cell Line, Tumor , Cytotoxins/genetics , Deoxycytidine/pharmacology , Dipeptides/pharmacology , Humans , Hydroxamic Acids/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Thiophenes/pharmacology , Gemcitabine
7.
Neuro Oncol ; 26(1): 70-84, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37551745

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is refractory to current treatment modalities while side effects of treatments result in neurotoxicity and cognitive impairment. Here we test the hypothesis that inhibiting CDK7 or CDK9 would effectively combat GBM with reduced neurotoxicity. METHODS: We examined the effect of a CDK7 inhibitor, THZ1, and multiple CDK9 inhibitors (SNS032, AZD4573, NVP2, and JSH150) on GBM cell lines, patient-derived temozolomide (TMZ)-resistant and responsive primary tumor cells and glioma stem cells (GSCs). Biochemical changes were assessed by western blotting, immunofluorescence, multispectral imaging, and RT-PCR. In vivo, efficacy was assessed in orthotopic and subcutaneous xenograft models. RESULTS: CDK7 and CDK9 inhibitors suppressed the viability of TMZ-responsive and resistant GBM cells and GSCs at low nanomolar concentrations, with limited cytotoxic effects in vivo. The inhibitors abrogated RNA Pol II and p70S6K phosphorylation and nascent protein synthesis. Furthermore, the self-renewal of GSCs was significantly reduced with a corresponding reduction in Sox2 and Sox9 levels. Analysis of TCGA data showed increased expression of CDK7, CDK9, SOX2, SOX9, and RPS6KB1 in GBM; supporting this, multispectral imaging of a TMA revealed increased levels of CDK9, Sox2, Sox9, phospho-S6, and phospho-p70S6K in GBM compared to normal brains. RNA-Seq results suggested that inhibitors suppressed tumor-promoting genes while inducing tumor-suppressive genes. Furthermore, the studies conducted on subcutaneous and orthotopic GBM tumor xenograft models showed that administration of CDK9 inhibitors markedly suppressed tumor growth in vivo. CONCLUSIONS: Our results suggest that CDK7 and CDK9 targeted therapies may be effective against TMZ-sensitive and resistant GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/genetics , Ribosomal Protein S6 Kinases, 70-kDa/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/therapeutic use , Drug Resistance, Neoplasm , Cell Line, Tumor , Glioma/drug therapy , Brain Neoplasms/genetics , Xenograft Model Antitumor Assays , Cyclin-Dependent Kinase 9/metabolism
8.
Electrophoresis ; 33(24): 3728-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23161580

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly. Amyloid plaque formation through aggregation of the amyloid beta peptide derived from amyloid precursor protein (APP) is considered one of the hallmark processes leading to AD pathology; however, the precise role of APP in plaque formation and AD pathogenesis is yet to be determined. Using stable isotope labeling by amino acids in cell culture (SILAC) and MS, protein expression profiles of APP null, rat neuronal-like B103 cells were compared to B103-695 cells that express the APP isoform, APP-695. A total of 2979 unique protein groups were identified among three biological replicates and significant protein expression changes were identified in a total of 102 nonredundant proteins. Some of the top biological functions associated with the differentially expressed proteins identified include cellular assembly, organization and morphology, cell cycle, lipid metabolism, protein folding, and PTMs. We report several novel biological pathways influenced by APP-695 expression in neuronal-like cells and provide additional framework for investigating altered molecular mechanisms associated with APP expression and processing and contribution to AD pathology.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Isotope Labeling/methods , Neurons/metabolism , Proteome/analysis , Proteomics/methods , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Immunoblotting , Mass Spectrometry , Microscopy, Fluorescence , Neurons/chemistry , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Proteome/chemistry , Proteome/metabolism , Rats , Signal Transduction
9.
Adv Exp Med Biol ; 740: 1193-217, 2012.
Article in English | MEDLINE | ID: mdl-22453989

ABSTRACT

Calcium plays a major role in normal functioning of the cells. Deregulation of calcium-mediated signaling has been implicated in many neurodegenerative diseases including Alzheimer's disease. Studies in neurons and mice expressing Alzheimer's disease-associated transgenes have shown that the expression of familial Alzheimer's disease (FAD) mutants of presenilin (PS) and amyloid precursor protein (APP) alter calcium homeostasis and cause synaptic dysfunction and dendritic spine loss in neurons. Mechanistic studies have shown that FAD mutants of presenilin can affect the intracellular calcium levels by affecting the ER calcium stores. A function for presenilins as ER calcium leak channels has been established and studies show that presenilins affect ER calcium load through an effect on IP(3) receptors, ryanodine receptors, or SERCA pumps. Even in the absence of an active gamma-secretase complex, presenilins seem to affect calcium homeostasis suggesting that these two functions of presenilins are independent of each other. Studies using FAD mutants of APP have shown that unlike presenilins, FAD-APP do not affect calcium homeostasis in the absence of Aß. Both Aß and presenilins seem to affect calcium homeostasis at very early stages of disease development affecting the synaptic transmission and function prior to neuritic plaque development. Altered calcium signaling differentially regulates genes such as calcineurin, calmodulin kinase II, MAP kinase etc and induces protein modifications and neurite degeneration. Since functional synapses and synaptic transmission are fundamental processes in memory formation, alterations in these processes can lead to neuronal dysfunction and memory deficit as seen in Alzheimer's disease. This chapter gives an overview of calcium signaling in different systems, specifically neurons, the functioning of pre- and post-synaptic signaling, and how their deregulation influences pathology development in Alzheimer's disease.


Subject(s)
Alzheimer Disease/etiology , Calcium Signaling/physiology , Neurons/metabolism , Calcium/metabolism , Calcium Channels/physiology , Homeostasis , Humans , Mitochondria/metabolism , Neurotransmitter Agents/metabolism , Receptors, N-Methyl-D-Aspartate/physiology
10.
Cancer Res Commun ; 2(7): 624-638, 2022 07.
Article in English | MEDLINE | ID: mdl-35937460

ABSTRACT

The transcriptional co-activator YAP1 is the major oncogenic component of the Hippo signaling pathway and contributes to the genesis and progression of various tumors, including non-small cell lung cancer (NSCLC). YAP1 levels are regulated by the canonical Hippo kinases, MST1/2 and LATS1/2, which modulate its cytoplasmic retention and proteasomal degradation. While non-canonical regulation of YAP1 has been reported, its role in hypoxic response is not fully elucidated. The studies presented here show that YAP1 levels and function are modulated by VHL and PHD2. YAP1 could regulate multiple genes involved in angiogenesis through E2F1; it also associates with HIF1α in cancer cells under hypoxic conditions, inducing the VEGF-A promoter. Under normoxic conditions, PHD2 associates with and hydroxylates specific proline residues on YAP1, facilitating its interaction with VHL and promoting ubiquitination and subsequent proteasomal degradation. Exposure to hypoxia dissociates YAP1 from PHD2 and VHL, elevating YAP1 levels and enhancing its association with HIF1α. YAP1-HIF1α interaction was higher in NSCLC and RCC samples, indicating a role for this interaction in the genesis of these cancers. Our results thus reveal a novel mode of regulation of YAP1 by PHD2 and VHL in normoxic cells, suggesting that YAP1-mediated induction of VEGF and other genes contributes to hypoxic response in tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Vascular Endothelial Growth Factor A/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Transcription Factors/genetics , Hippo Signaling Pathway , Von Hippel-Lindau Tumor Suppressor Protein/genetics
11.
NPJ Parkinsons Dis ; 8(1): 143, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36302787

ABSTRACT

Parkinson's disease (PD) treatments modify disease symptoms but have not been shown to slow progression, characterized by gradual and varied motor and non-motor changes overtime. Variation in PD progression hampers clinical research, resulting in long and expensive clinical trials prone to failure. Development of models for short-term PD progression prediction could be useful for shortening the time required to detect disease-modifying drug effects in clinical studies. PD progressors were defined by an increase in MDS-UPDRS scores at 12-, 24-, and 36-months post-baseline. Using only baseline features, PD progression was separately predicted across all timepoints and MDS-UPDRS subparts in independent, optimized, XGBoost models. These predictions plus baseline features were combined into a meta-predictor for 12-month MDS UPDRS Total progression. Data from the Parkinson's Progression Markers Initiative (PPMI) were used for training with independent testing on the Parkinson's Disease Biomarkers Program (PDBP) cohort. 12-month PD total progression was predicted with an F-measure 0.77, ROC AUC of 0.77, and PR AUC of 0.76 when tested on a hold-out PPMI set. When tested on PDBP we achieve a F-measure 0.75, ROC AUC of 0.74, and PR AUC of 0.73. Exclusion of genetic predictors led to the greatest loss in predictive accuracy; ROC AUC of 0.66, PR AUC of 0.66-0.68 for both PPMI and PDBP testing. Short-term PD progression can be predicted with a combination of survey-based, neuroimaging, physician examination, and genetic predictors. Dissection of the interplay between genetic risk, motor symptoms, non-motor symptoms, and longer-term expected rates of progression enable generalizable predictions.

12.
Cancers (Basel) ; 13(15)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34359807

ABSTRACT

Non-small cell lung cancer has a 5-year survival rate of less than 12-15%, calling for the development of additional therapeutic strategies to combat this disease. Here we tested the efficacy of inhibiting cyclin-dependent kinase 9 (CDK9) on lung cancer cell lines with K-Ras and EGFR mutations and on lung cancer organoids. Three different CDK9 inhibitors reduced the viability and anchorage-independent growth of lung cancer cell lines at very low nanomolar to micromolar concentrations. CDK9 inhibition suppressed the expression of the anti-apoptotic protein, Mcl1, as well as the embryonic stem cell transcription factors, Sox2 and Sox9, which are pro-tumorigenic. In contrast, treatment with CDK9 inhibitors increased the levels of WT p53 and its downstream target p21 in K-Ras mutant cell lines. Furthermore, the CDK9 inhibitors could markedly reduce the viability of Osimertinib-resistant PC9 and AMG510-resistant H23 and H358 cells with comparable efficacy as the parental cells. CDK9 inhibitors could also significantly reduce the growth and viability of lung cancer organoids with high potency. Taken together, the data presented here strongly suggest that CDK9 inhibitors would be efficacious against K-Ras mutant and EGFR mutant NSCLCs, including those that develop resistance to targeted therapies.

13.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118929, 2021 03.
Article in English | MEDLINE | ID: mdl-33310066

ABSTRACT

Error-free progression through mitosis is critical for proper cell division and accurate distribution of the genetic material. The anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase regulates the progression from metaphase to anaphase and its activation is controlled by the cofactors Cdc20 and Cdh1. Additionally, genome stability is maintained by the spindle assembly checkpoint (SAC), which monitors proper attachment of chromosomes to spindle microtubules prior to cell division. We had shown a role for Tank Binding Kinase 1 (TBK1) in microtubule dynamics and mitosis and here we describe a novel role of TBK1 in regulating SAC in breast and lung cancer cells. TBK1 interacts with and phosphorylates Cdc20 and Cdh1 and depletion of TBK1 elevates SAC components. TBK1 inhibition increases the association of Cdc20 with APC/C and BubR1 indicating inactivation of APC/C; similarly, interaction of Cdh1 with APC/C is also enhanced. TBK1 and TTK inhibition reduces cell viability and enhances centrosome amplification and micronucleation. These results indicate that alterations in TBK1 will impede mitotic progression and combining TBK1 inhibitors with other regulators of mitosis might be effective in eliminating cancer cells.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cdc20 Proteins/metabolism , Cdh1 Proteins/metabolism , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , A549 Cells , Anaphase-Promoting Complex-Cyclosome/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , M Phase Cell Cycle Checkpoints , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/genetics
14.
Sci Rep ; 11(1): 9016, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907253

ABSTRACT

Nek2 (NIMA-related kinase 2) is a serine/threonine-protein kinase that localizes to centrosomes and kinetochores, controlling centrosome separation, chromosome attachments to kinetochores, and the spindle assembly checkpoint. These processes prevent centrosome amplification (CA), mitotic dysfunction, and chromosome instability (CIN). Our group and others have suggested that Nek2 maintains high levels of CA/CIN, tumor growth, and drug resistance. We identified that Nek2 overexpression correlates with poor survival of breast cancer. However, the mechanisms driving these phenotypes are unknown. We now report that overexpression of Nek2 in MCF10A cells drives CA/CIN and aneuploidy. Besides, enhanced levels of Nek2 results in larger 3D acinar structures, but could not initiate tumors in a p53+/+ or a p53-/- xenograft model. Nek2 overexpression induced the epithelial-to-mesenchymal transition (EMT) while its downregulation reduced the expression of the mesenchymal marker vimentin. Furthermore, either siRNA-mediated downregulation or INH6's chemical inhibition of Nek2 in MDA-MB-231 and Hs578t cells showed important EMT changes and decreased invasion and migration. We also showed that Slug and Zeb1 are involved in Nek2 mediated EMT, invasion, and migration. Besides its role in CA/CIN, Nek2 contributes to breast cancer progression through a novel EMT mediated mechanism.


Subject(s)
Centrosome/metabolism , Epithelial-Mesenchymal Transition , NIMA-Related Kinases/metabolism , Triple Negative Breast Neoplasms/enzymology , Acinar Cells/pathology , Aneuploidy , Animals , Carcinogenesis , Cell Line, Tumor , Cell Movement , Chromosomal Instability , Epithelial Cells/pathology , Female , Humans , Mice , Neoplasm Invasiveness , Snail Family Transcription Factors/metabolism , Survival Analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
15.
Nat Hum Behav ; 5(12): 1707-1716, 2021 12.
Article in English | MEDLINE | ID: mdl-34239076

ABSTRACT

Damage to specific brain circuits can cause specific neuropsychiatric symptoms. Therapeutic stimulation to these same circuits may modulate these symptoms. To determine whether these circuits converge, we studied depression severity after brain lesions (n = 461, five datasets), transcranial magnetic stimulation (n = 151, four datasets) and deep brain stimulation (n = 101, five datasets). Lesions and stimulation sites most associated with depression severity were connected to a similar brain circuit across all 14 datasets (P < 0.001). Circuits derived from lesions, deep brain stimulation and transcranial magnetic stimulation were similar (P < 0.0005), as were circuits derived from patients with major depression versus other diagnoses (P < 0.001). Connectivity to this circuit predicted out-of-sample antidepressant efficacy of transcranial magnetic stimulation and deep brain stimulation sites (P < 0.0001). In an independent analysis, 29 lesions and 95 stimulation sites converged on a distinct circuit for motor symptoms of Parkinson's disease (P < 0.05). We conclude that lesions, transcranial magnetic stimulation and DBS converge on common brain circuitry that may represent improved neurostimulation targets for depression and other disorders.


Subject(s)
Brain/diagnostic imaging , Deep Brain Stimulation/methods , Mental Disorders/therapy , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/therapy , Female , Humans , Magnetic Resonance Imaging , Male , Mental Disorders/diagnostic imaging , Neural Pathways/diagnostic imaging , Transcranial Magnetic Stimulation
16.
Methods Mol Biol ; 523: 311-22, 2009.
Article in English | MEDLINE | ID: mdl-19381929

ABSTRACT

Growth factor stimulation results in phosphorylation of histone H3 at ser 10 and this correlated with expression of immediate early genes suggesting that this phosphorylation is associated with transcriptional activation. Although Western immunoblot analysis allows the detection of protein modifications in histones, in order to determine the localization of histones during different phases of cell cycle or during treatment of cells with different drugs we have to use immunohistochemistry. The protocol described here allows the detection of phosphorylated histones in tissue-cultured cells and tissue sections by fluorescent or bright-field immunostaining analysis. Here we used a serine 10 specific P-histone H3 antibody to determine the localization of this phosphoprotein in an asynchronously growing H4 glioma cell line and brain sections. It has been shown that long-term potentiation (LTP) is associated with gene transcription, and histone acetylation plays a major role in LTP formation (Wood et al., Learn Mem 13:241-244, 2006; Wood et al., Hippocampus 15:610-621, 2005; Alarcon et al., Neuron 42:947-959, 2004; Korzus et al., Neuron 42:961-972, 2004). Stimulus-induced phosphorylation of histone H3 at serine 10 has also been implicated in hippocampal neurons and striatal neurons (Li et al., J Neurochem 90:1117-1131, 2004; Crosio et al., J Cell Sci 116:4905-4914, 2003). Co-staining with a cell-specific antibody will allow us to determine the type of cells that show activation of histone phosphorylation in the brain.


Subject(s)
Histones/metabolism , Immunohistochemistry/methods , Microtomy/methods , Animals , Antibodies, Phospho-Specific/immunology , Brain/cytology , Brain/metabolism , Cells, Cultured , Mice , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Paraffin Embedding , Phosphorylation , Phosphoserine/immunology
17.
Schizophr Res ; 207: 37-47, 2019 05.
Article in English | MEDLINE | ID: mdl-30409696

ABSTRACT

BACKGROUND: Several lines of investigations converge upon aberrant synaptic plasticity as a potential pathophysiological characteristic of schizophrenia. In vivo experiments using neuromodulatory perturbation techniques like Transcranial Magnetic and Direct Current Stimulation (TMS & tDCS) have been increasingly used to measure 'motor cortical plasticity' in schizophrenia. A systematic quantification of cortical plasticity and its moderators in schizophrenia is however lacking. METHOD: The PubMed/MEDLINE database was searched for studies up to December 31st, 2017 that examined case-control experiments comparing neuromodulation following single-session of TMS or tDCS. The primary outcome was the standardized mean difference for differential changes in motor evoked potential (MEP) amplitudes measured with single-pulse TMS (MEP Δ) between patients and healthy subjects following TMS or tDCS. After examining heterogeneity, meta-analyses were performed using fixed effects models. RESULTS: A total of 16 datasets comparing cortical plasticity (MEP Δ) between 189 schizophrenia patients and 187 healthy controls were included in the meta-analysis. Patients demonstrated diminished MEP Δ with effect sizes (Cohen's d) ranging from 0.66 (LTP-like plasticity) to 0.68 (LTD-like plasticity). Heterosynaptic plasticity studies demonstrated a greater effect size (0.79) compared to homosynaptic plasticity studies (0.62), though not significant (P = 0.43). Clinical, perturbation protocol- and measurement-related factors, and study quality did not significantly moderate the aberrant plasticity demonstrated in schizophrenia. CONCLUSIONS: Schizophrenia patients demonstrate diminished LTP- and LTD-like motor cortical plasticity, which is not influenced by the various clinical and experimental protocol related confounders. These consistent findings should encourage the use of perturbation-based biomarkers to characterize illness trajectories and treatment response.


Subject(s)
Electromyography , Evoked Potentials, Motor/physiology , Motor Cortex/physiopathology , Neuronal Plasticity/physiology , Schizophrenia/physiopathology , Transcranial Magnetic Stimulation , Humans
18.
Neurodegener Dis Manag ; 9(4): 217-239, 2019 08.
Article in English | MEDLINE | ID: mdl-31392920

ABSTRACT

Posterior cortical atrophy (PCA) is a progressive neurocognitive syndrome, most commonly associated with the loss of complex visuospatial functions. Diagnosis is challenging, and international consensus classification and nomenclature for PCA subtypes have only recently been reached. Presently, no established treatments exist. Efforts to develop treatments are hampered by the lack of standardized methods to monitor illness progression. Although measures developed from work with Alzheimer's disease and other dementias provide a foundation for diagnosing and monitoring progression, PCA presents unique challenges for clinicians counseling patients and families on clinical status and prognosis, and experts designing clinical trials of interventions. Here, we review issues facing PCA clinical research and care, summarize our approach to diagnosis and monitoring of disease progression, and outline ideas for developing tools for these purposes.


Subject(s)
Dementia/diagnosis , Occipital Lobe/pathology , Perceptual Disorders/etiology , Vision Disorders/etiology , Age of Onset , Aged , Alzheimer Disease/classification , Alzheimer Disease/diagnosis , Atrophy , Cognition Disorders/etiology , Cognitive Dysfunction/diagnosis , Dementia/complications , Dementia/pathology , Dementia/rehabilitation , Diagnosis, Differential , Disease Management , Disease Progression , Executive Function , Female , Genetic Predisposition to Disease , Humans , Language Disorders/etiology , Male , Memory Disorders/etiology , Mental Status and Dementia Tests , Middle Aged , Neuroimaging , Occipital Lobe/physiopathology , Psychomotor Disorders/etiology , Severity of Illness Index , Visual Perception
19.
Biol Psychiatry ; 86(10): 749-758, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31561861

ABSTRACT

BACKGROUND: Focal brain lesions can lend insight into the causal neuroanatomical substrate of depression in the human brain. However, studies of lesion location have led to inconsistent results. METHODS: Five independent datasets with different lesion etiologies and measures of postlesion depression were collated (N = 461). Each 3-dimensional lesion location was mapped to a common brain atlas. We used voxel lesion symptom mapping to test for associations between depression and lesion locations. Next, we computed the network of regions functionally connected to each lesion location using a large normative connectome dataset (N = 1000). We used these lesion network maps to test for associations between depression and connected brain circuits. Reproducibility was assessed using a rigorous leave-one-dataset-out validation. Finally, we tested whether lesion locations associated with depression fell within the same circuit as brain stimulation sites that were effective for improving poststroke depression. RESULTS: Lesion locations associated with depression were highly heterogeneous, and no single brain region was consistently implicated. However, these same lesion locations mapped to a connected brain circuit, centered on the left dorsolateral prefrontal cortex. Results were robust to leave-one-dataset-out cross-validation. Finally, our depression circuit derived from brain lesions aligned with brain stimulation sites that were effective for improving poststroke depression. CONCLUSIONS: Lesion locations associated with depression fail to map to a specific brain region but do map to a specific brain circuit. This circuit may have prognostic utility in identifying patients at risk for poststroke depression and therapeutic utility in refining brain stimulation targets.


Subject(s)
Brain/pathology , Depressive Disorder/physiopathology , Nerve Net/physiopathology , Neural Pathways/physiopathology , Adult , Aged , Brain/physiopathology , Brain Mapping , Case-Control Studies , Connectome , Depression , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Reproducibility of Results
20.
Transl Psychiatry ; 9(1): 230, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530798

ABSTRACT

Schizophrenia, Schizoaffective, and Bipolar disorders share behavioral and phenomenological traits, intermediate phenotypes, and some associated genetic loci with pleiotropic effects. Volumetric abnormalities in brain structures are among the intermediate phenotypes consistently reported associated with these disorders. In order to examine the genetic underpinnings of these structural brain modifications, we performed genome-wide association analyses (GWAS) on 60 quantitative structural brain MRI phenotypes in a sample of 777 subjects (483 cases and 294 controls pooled together). Genotyping was performed with the Illumina PsychChip microarray, followed by imputation to the 1000 genomes multiethnic reference panel. Enlargement of the Temporal Horns of Lateral Ventricles (THLV) is associated with an intronic SNP of the gene NRXN1 (rs12467877, P = 6.76E-10), which accounts for 4.5% of the variance in size. Enlarged THLV is associated with psychosis in this sample, and with reduction of the hippocampus and enlargement of the choroid plexus and caudate. Eight other suggestively significant associations (P < 5.5E-8) were identified with THLV and 5 other brain structures. Although rare deletions of NRXN1 have been previously associated with psychosis, this is the first report of a common SNP variant of NRXN1 associated with enlargement of the THLV in psychosis.


Subject(s)
Calcium-Binding Proteins/genetics , Lateral Ventricles/diagnostic imaging , Neural Cell Adhesion Molecules/genetics , Psychotic Disorders/genetics , Adult , Alleles , Female , Genome-Wide Association Study , Genotype , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Polymorphism, Single Nucleotide , Psychotic Disorders/diagnostic imaging , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL