Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Ann Hematol ; 101(10): 2209-2218, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36040481

ABSTRACT

The coincident downregulation of NR4A1 and NR4A3 has been implicated in myeloid leukemogenesis, but it remains unknown how these two genes function in myeloid cells and how their combined downregulation promotes myeloid leukemogenesis. Since NR4A1 abrogation is thought to confer a survival and proliferation advantage to myeloid cells, we hypothesized that downregulation of NR4A3 may have a complementary effect on myeloid cell differentiation. First, we tested the association between differentiation status of leukemic cells and NR4A3 expression using two large clinical datasets from patients with different acute myeloid leukemia (AML) subtypes. The analysis revealed a close association between differentiation status and different subtypes of AML Then, we probed the effects of differentiation-inducing treatments on NR4A3 expression and NR4A3 knockdown on cell differentiation using two myeloid leukemia cell lines. Differentiation-inducing treatments caused upregulation of NR4A3, while NR4A3 knockdown prevented differentiation in both cell lines. The cell culture findings were validated using samples from chronic myeloid leukemia (CML) patients at chronic, accelerated and blastic phases, and in acute promyelocytic leukemia (APL) patients before and after all trans-retinoic acid (ATRA)-based differentiation therapy. Progressive NR4A3 downregulation was coincident with impairments in differentiation in patients during progression to blastic phase of CML, and NR4A3 expression was increased in APL patients treated with ATRA-based differentiating therapy. Together, our findings demonstrate a tight association between impaired differentiation status and NR4A3 downregulation in myeloid leukemias, providing a plausible mechanistic explanation of how myeloid leukemogenesis might occur upon concurrent downregulation of NR4A1 and NR4A3.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Acute , Leukemia, Promyelocytic, Acute , Receptors, Steroid , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Promyelocytic, Acute/drug therapy , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Steroid/therapeutic use , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/therapeutic use , Tretinoin/pharmacology
2.
Biochem Biophys Res Commun ; 529(2): 263-269, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703421

ABSTRACT

The World Health Organization recently announced that pandemic status has been achieved for coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Exponential increases in patient numbers have been reported around the world, along with proportional increases in the number of COVID-19-related deaths. The SARS-CoV-2 infection rate in a population is expected to be influenced by social practices, availability of vaccines or prophylactics, and the prevalence of susceptibility genes in the population. Previous work revealed that cellular uptake of SARS-CoV-2 requires Angiotensin Converting Enzyme 2 (ACE-2) and a cellular protease. The spike (S) protein on SARS-CoV-2 binds ACE-2, which functions as an entry receptor. Following receptor binding, transmembrane protease serine 2 (encoded by TMPRSS2) primes the S protein to allow cellular uptake. Therefore, individual expression of TMPRSS2 may be a crucial determinant of SARS-CoV-2 infection susceptibility. Here, we utilized multiple large genome databases, including the GTEx portal, SNP nexus, and Ensembl genome project, to identify gene expression profiles for TMPRSS2 and its important expression quantitative trait loci. Our results show that four variants (rs464397, rs469390, rs2070788 and rs383510) affect expression of TMPRSS2 in lung tissue. The allele frequency of each variant was then assessed in regional populations, including African, American, European, and three Asian cohorts (China, Japan and Taiwan). Interestingly, our data shows that TMPRSS2-upregulating variants are at higher frequencies in European and American populations than in the Asian populations, which implies that these populations might be relatively susceptible to SARS-CoV-2 infection.


Subject(s)
Betacoronavirus/metabolism , Gene Expression Regulation/genetics , Internationality , Lung/metabolism , Receptors, Virus/genetics , Serine Endopeptidases/genetics , Asia/ethnology , Cohort Studies , Europe/ethnology , Gene Frequency , Genetics, Population , Geographic Mapping , Humans , Organ Specificity/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , SARS-CoV-2 , United States/ethnology , Up-Regulation/genetics
3.
Life Sci ; 254: 117755, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32437792

ABSTRACT

AIMS: Efficient memory formation in rodents depends on adult neurogenesis in the subgranular zone of the hippocampus, and mounting evidence suggests that deficiencies in initiating repair of oxidatively induced DNA damage may impair neurogenesis. Hence, we aimed to determine whether loss of the DNA glycosylase, endonuclease VIII-like 1 (Neil1), affects hippocampal neurogenesis and memory performance in young-adult mice. MAIN METHODS: Eight-week-old male wild-type and Neil1-deficient (Neil1-/-) mice were treated with bromodeoxyuridine to track neuronal proliferation and differentiation. A neurosphere formation assay was further used to measure neuroprogenitor proliferative capacity. Hippocampus-related memory functions were assessed with Y-maze spontaneous alternation and novel object recognition tests. KEY FINDINGS: Young-adult male Neil1-/- mice exhibited diminished adult hippocampal neurogenesis in the dentate gyrus, probably as a result of poor survival of newly proliferated neurons. Furthermore, the Y-maze spontaneous alternation and novel object recognition tests respectively revealed that Neil1 deficiency impairs spatial and non-spatial hippocampus-related memory functions. We also found that expression of p53, a central regulator of apoptosis, was upregulated in the dentate gyrus of Neil1-/- mice, while the level of ß-catenin, a key cell survival molecule, was downregulated. SIGNIFICANCE: The DNA glycosylase, Neil1, promotes successful hippocampal neurogenesis and learning and memory in young-adult mice.


Subject(s)
Cognition/physiology , DNA Glycosylases/deficiency , Hippocampus/enzymology , Memory/physiology , Neurons/enzymology , Animals , Cell Differentiation/physiology , Cell Survival/physiology , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/pathology , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Hippocampus/cytology , Hippocampus/metabolism , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/physiology , Neurons/cytology
4.
FASEB Bioadv ; 2(3): 166-181, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32161906

ABSTRACT

Dynamic fission and fusion events regulate mitochondrial shape, distribution, and rejuvenation, and proper control of these processes is essential for neuronal homeostasis. Here, we report that Gas7, a known cytoskeleton regulator, controls mitochondrial dynamics within neurons of the central nervous system. In this study, we generated an improved Gas7-knockout mouse and evaluated its mitochondrial phenotype. We first identified Gas7 in mitochondrial fractions from wild-type brain tissue, and observed Gas7 colocalization with mitochondria in primary cortical neurons. In Gas7-deficient brain tissue and neuronal cultures mitochondria were elongated with perinuclear clustering. These morphological abnormalities were associated with increased levels mitochondrial fusion proteins and increased PKA-dependent phosphorylation of Drp-1 in brain tissues, suggesting an imbalance of mitochondrial fusion and fission. Moreover, expression of mitochondrial quality control kinase, PINK1, and PINK1-specific phosphorylation of Mfn-2 (S442), Parkin (S65), and ubiquitin (S65) were all reduced in the knockout cells. Ectopic expression of Gas7 restored mitochondrial morphology and distribution, as well as PINK1 expression in Gas7-null cortical neurons. Collectively, our results introduce a novel role of mouse Gas7 in determining the dynamics, morphology, and intracellular distribution of neuronal mitochondria, which are expected to be required for normal neuronal function.

5.
Ther Adv Med Oncol ; 11: 1758835919875324, 2019.
Article in English | MEDLINE | ID: mdl-31632466

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is the major type of esophageal cancer in Asia and demonstrates poor survival rates following a therapeutic regimen. METHODS: Cancer stem cells (CSCs) are responsible for tumor initiation, progression, and treatment failure in cancers. Therefore, identification and characterization of CSCs may help to improve clinical outcomes for ESCC patients. Tumor sphere formation assay are performed to isolate cancer stem-like ESCC cells. QRT-PCR, tumor initiation, metastasis, CCRT treatment are used to evaluate ESCC cells' stemness properties in vitro and in vivo. RESULTS: The authors' data demonstrates that cancer stem-like ESCC cells harbored stemness characteristics including self-renewal, differentiation, and transdifferentiation, and possess tumor initiation, metastasis, and treatment inefficiency properties. For the identification of useful biomarkers of cancer stem-like ESCC cells, the authors further identified that CLDN4 was upregulated in cancer stem-like ESCC cells when compared with bulk cancer cells. High-CLDN4 cells harbored stemness and cisplatin/concurrent chemoradiation therapy (CCRT) resistance properties and a high level of CLDN4 was correlated with poor prognosis and poor CCRT response in ESCC patients. Importantly, thiamine tetrahydrofurfuryl disulfide (TTFD) decreased CLDN4 and attenuated stemness in ESCC cells, and TTFD combined with CCRT improved CCRT response in vivo. CONCLUSIONS: CLDN4 was suggested as prognostic and a CCRT response indicator for ESCC patients. TTFD combined with CCRT has potential to improve ESCC patient's clinical outcomes in the future.

6.
Sci Rep ; 8(1): 705, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335541

ABSTRACT

The molecular basis for ultraviolet (UV) light-induced nonmelanoma and melanoma skin cancers centers on cumulative genomic instability caused by inefficient DNA repair of dipyrimidine photoproducts. Inefficient DNA repair and subsequent translesion replication past these DNA lesions generate distinct molecular signatures of tandem CC to TT and C to T transitions at dipyrimidine sites. Since previous efforts to develop experimental strategies to enhance the repair capacity of basal keratinocytes have been limited, we have engineered the N-terminally truncated form (Δ228) UV endonuclease (UVDE) from Schizosaccharomyces pombe to include a TAT cell-penetrating peptide sequence with or without a nuclear localization signal (NLS): UVDE-TAT and UVDE-NLS-TAT. Further, a NLS was engineered onto a pyrimidine dimer glycosylase from Paramecium bursaria chlorella virus-1 (cv-pdg-NLS). Purified enzymes were encapsulated into liposomes and topically delivered to the dorsal surface of SKH1 hairless mice in a UVB-induced carcinogenesis study. Total tumor burden was significantly reduced in mice receiving either UVDE-TAT or UVDE-NLS-TAT versus control empty liposomes and time to death was significantly reduced with the UVDE-NLS-TAT. These data suggest that efficient delivery of exogenous enzymes for the initiation of repair of UVB-induced DNA damage may protect from UVB induction of squamous and basal cell carcinomas.


Subject(s)
Carcinogenesis/radiation effects , DNA Repair , Skin Neoplasms/prevention & control , Ultraviolet Rays , Animals , DNA Repair Enzymes/administration & dosage , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Mice, Hairless , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Breast Cancer Res ; 19(1): 133, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29258605

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) lacks both early detection biomarkers and viable targeted therapeutics. Moreover, chemotherapy only produces 20-30% pathologic complete response. Because miRNAs are frequently dysregulated in breast cancer and have broad tissue effects, individual or combinations of circulating miRNAs may serve as ideal diagnostic, predictive or prognostic biomarkers, as well as therapeutic targets. Understanding the role and mechanism of dysregulated miRNAs in TNBC may help to develop novel diagnostic and prognostic strategy for TNBC patients. METHODS: The miRNA array profiles of 1299 breast cancer patients were collected from the Metabric database and subjected to analysis of the altered miRNAs between TNBC and non-TNBC. In Student's t-test and Kaplan-Meier analysis, four upregulated miRNAs correlated with poor survival in TNBC but not in non-TNBC. Four miRNAs were manipulated in multiple cell lines to investigate their functional role in carcinogenesis. From these results, we studied miR-105 and miR-93-3p in greater detail. The level of miR-105 and miR-93-3p were evaluated in 25 breast cancer tumor tissues. In addition, the diagnostic utility of circulating miR-105 and miR-93-3p were examined in 12 normal and 118 breast cancer plasma samples by ROC curve construction. RESULTS: miR-105 and miR-93-3p were upregulated and correlated with poor survival in TNBC patients. Both miR-105 and miR-93-3p were found to activate Wnt/ß-catenin signaling by downregulation of SFPR1. By this action, stemness, chemoresistance, and metastasis were promoted. Importantly, the combination of circulating miR-105/93-3p may serve as a powerful biomarker for TNBC, even in early-stage disease. CONCLUSIONS: miR-105/93-3p activates Wnt/ß-catenin signaling by downregulating SFRP1 and thereby promotes stemness, chemoresistance, and metastasis in TNBC cells. Most importantly, combined circulating miR-105/93-3p levels represent a prime candidate for development into a diagnostic biomarker for both early- and late-stage TNBC.


Subject(s)
Biomarkers, Tumor , Circulating MicroRNA , Drug Resistance, Neoplasm/genetics , MicroRNAs/genetics , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents/pharmacology , Case-Control Studies , Female , Humans , Kaplan-Meier Estimate , MicroRNAs/blood , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , ROC Curve , Transcriptome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Wnt Signaling Pathway
8.
Oncotarget ; 8(18): 29699-29710, 2017 May 02.
Article in English | MEDLINE | ID: mdl-27447863

ABSTRACT

Trastuzumab is regarded as the primary therapy for patients with HER2-enriched breast cancer, but the pathological complete response for advanced cases is less than 30%. The underlying mechanism of trastuzumab resistance remains unclear and there are currently no conclusive biomarkers for patient response to trastuzumab. Identifying predictive biomarkers for trastuzumab response may allow treatments to be individually tailored and optimized multi-target therapies may be developed. CTMP activates AKT signaling in breast cancer and over-activation of AKT has been reported to contribute to trastuzumab resistance. In this study, we examined samples from 369 patients to investigate the correlation between CTMP expression level and patient outcome. Elevated CTMP expression was correlated with adverse outcomes in HER2-enriched patients including overall and disease-free survival as well as trastuzumab resistance. Ectopic expression of varying levels of CTMP in SkBR3 cells dose-dependently attenuated trastuzumab-mediated growth inhibition through AKT activation. In addition, inhibition of AKT signaling by AKT inhibitor IV and Rapamycin reversed CTMP-mediated trastuzumab resistance. In clinical samples, the high expression of CTMP was showed in trastuzumab non-responders and positively correlated with AKT activity. Taken together, we demonstrated that CTMP promotes AKT activation resulting in trastuzumab resistance in patients with HER2-enriched breast cancer. High CTMP expression not only predicted poor prognosis, but may also predict resistance to trastuzumab in HER2-enriched patients. Therefore, CTMP expression may be considered as a prognostic biomarker in HER2-enriched breast cancer and high expression may indicate a utility for AKT-inhibition in these patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Membrane Proteins/genetics , Receptor, ErbB-2/metabolism , Thiolester Hydrolases/genetics , Trastuzumab/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Female , Gene Expression , Humans , Membrane Proteins/metabolism , Middle Aged , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Recurrence , Thiolester Hydrolases/metabolism , Trastuzumab/therapeutic use
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 406-413, 2017 02.
Article in English | MEDLINE | ID: mdl-27840304

ABSTRACT

The cornerstone of current HIV treatment is a class of drugs called nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs). However, patients who receive long term treatment with NRTIs often develop severe side effects, which are related to mitochondrial toxicity. The potential contribution of NRTI-mediated toxicity to HIV-associated neurocognitive disorders (HAND) has not been fully explored. NRTI toxicity is thought to be mediated through mitochondrial DNA polymerase γ (pol γ) inhibition, which impairs mitochondrial DNA (mtDNA) synthesis and leads to various mitochondrial dysfunctions. To evaluate the relationship between NRTI-mediated pol γ inhibition and mitochondrial toxicity in neurons, we systematically investigated mitochondrial regulation in NRTI-treated primary cortical neurons by measuring parameters related to mtDNA content, retrograde signaling responses and mitochondrial homeostasis. The effects of four different NRTIs with variable pol γ inhibitory activity and mitochondrial toxicity were assessed. The strong pol γ inhibitor, ddI, abolished mtDNA synthesis and greatly reduced mtDNA content. However, mtDNA transcription was not as severely affected, and no defects in oxidative phosphorylation were observed. Detrimental effects on mitochondrial respiration and motility were observed after AZT treatment in the absence of mtDNA depletion or inhibition of mtDNA synthesis. The results suggest that individual NRTIs, such as ddI and AZT, have the potential to cause mitochondrial toxicity in neurons. This mitochondrial toxicity would be expected to contribute to neurotoxicity in the central nervous system, and therefore, HAND etiology may be affected by NRTI treatment.


Subject(s)
HIV Infections/drug therapy , Mitochondria/drug effects , Neurocognitive Disorders/chemically induced , Neurons/drug effects , Reverse Transcriptase Inhibitors/adverse effects , Zidovudine/adverse effects , Animals , Cells, Cultured , DNA, Mitochondrial/genetics , HIV Infections/pathology , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/pathology , Neurocognitive Disorders/pathology , Neurons/pathology
10.
DNA Repair (Amst) ; 48: 43-50, 2016 12.
Article in English | MEDLINE | ID: mdl-27818081

ABSTRACT

Oxidative stress and reactive oxygen species (ROS)-induced DNA base damage are thought to be central mediators of UV-induced carcinogenesis and skin aging. However, increased steady-state levels of ROS-induced DNA base damage have not been reported after chronic UV exposure. Accumulation of ROS-induced DNA base damage is governed by rates of lesion formation and repair. Repair is generally performed by Base Excision Repair (BER), which is initiated by DNA glycosylases, such as 8-oxoguanine glycosylase and Nei-Endonuclease VIII-Like 1 (NEIL1). In the current study, UV light (UVB) was used to elicit protracted low-level ROS challenge in wild-type (WT) and Neil1-/- mouse skin. Relative to WT controls, Neil1-/- mice showed an increased sensitivity to tissue destruction from the chronic UVB exposure, and corresponding enhanced chronic inflammatory responses as measured by cytokine message levels and profiling, as well as neutrophil infiltration. Additionally, levels of several ROS-induced DNA lesions were measured including 4,6-diamino-5-formamidopyrimidine (FapyGua), 2,6-diamino-4-hydroxy-5-formamidopyrimidine (FapyAde), 8-hydroxyguanine (8-OH-Gua), 5,6-dihydroxyuracil (5,6-diOH-Ura) and thymine glycol (ThyGly). In WT mice, chronic UVB exposure led to increased steady-state levels of FapyGua, FapyAde, and ThyGly with no significant increases in 8-OH-Gua or 5,6-diOH-Ura. Interestingly, the lesions that accumulated were all substrates of NEIL1. Collectively, these data suggest that NEIL1-initiated repair of a subset of ROS-induced DNA base lesions may be insufficient to prevent the initiation of inflammatory pathways during chronic UV exposure in mouse skin.


Subject(s)
DNA Glycosylases/genetics , DNA Repair , Reactive Oxygen Species/metabolism , Skin/radiation effects , Animals , Cytokines/biosynthesis , Cytokines/genetics , DNA Damage , DNA Glycosylases/deficiency , DNA Glycosylases/metabolism , Gene Expression Profiling , Gene Expression Regulation , Guanine/analogs & derivatives , Guanine/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Neutrophil Infiltration/radiation effects , Oxidative Stress , Pyrimidines/metabolism , Reactive Oxygen Species/agonists , Skin/metabolism , Skin/pathology , Thymine/analogs & derivatives , Thymine/metabolism , Ultraviolet Rays , Uracil/analogs & derivatives , Uracil/metabolism
11.
Mitochondrion ; 31: 9-19, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27581214

ABSTRACT

Neurodegeneration and mitochondrial dysfunction are closely linked across many clinical conditions. In genetic diseases that result from defects in mitochondrial DNA (mtDNA) synthesis or maintenance, neurodegeneration is a frequent and major component of the disease pathology. In sporadic neurodegenerative diseases such as Alzheimer's and Parkinson's disease, mtDNA defects have been observed clinically. Mitochondrial stress related to mtDNA dysregulation can produce neuronal dysfunction and death via impaired electron transport chain activity, which results in deficient ATP production and related increases in mitochondrial reactive oxygen species (ROS) production. However, mtDNA dysregulation in post-mitotic neurons may also produce disturbances in mitochondrial homeostasis that are known to impair neuronal function as well. In this study, we used sub-toxic doses of ethidium bromide (EtBr) to induce mtDNA-associated mitochondrial stress in primary cortical neurons and measured several aspects of mitochondrial homeostasis, mitochondrial function and cell death. We found that low-dose EtBr severely depletes mtDNA synthesis and mitochondrial mRNA levels. Furthermore, homeostatic processes are especially disrupted in toxin treated neurons while mitochondrial function is relatively preserved. Mitochondria become fragmented and motility is abolished, while respiration and mitochondrial polarization are partially maintained. Moreover at these doses, cells do not exhibit increased ROS production, clear neurite retraction or loss of viability. These results indicate that mitochondrial homeostasis is a sensitive marker of mtDNA associated stress compared to mitochondria-functional outputs or endpoints related to cellular toxicity. These homeostatic disruptions are expected to contribute to neuronal dysfunction and potentially drive neurodegenerative disease pathology.


Subject(s)
DNA, Mitochondrial/metabolism , Homeostasis , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Neurodegenerative Diseases/pathology , Neurons/pathology , Animals , Cerebral Cortex/pathology , Ethidium/toxicity , Mice, Inbred C57BL , Mitochondria/drug effects , Mutagens/toxicity
12.
Oncotarget ; 7(26): 39680-39693, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27203740

ABSTRACT

Concurrent chemoradiation therapy (CCRT) is the predominant treatment in esophageal cancer, however resistance to therapy and tumor recurrence are exceedingly common. Elevated ERBB2/Her2 may be at least partially responsible for both the high rates of recurrence and resistance to CCRT. This receptor tyrosine kinase is upregulated in 10-20% of esophageal squamous cell carcinoma (ESCC) tissues, and amplification of ERBB2 has been correlated with poor prognosis in esophageal cancer. Tissues from 131 ESCC patients, along with cell and animal models of the disease were used to probe the underlying mechanisms by which ERBB2 upregulation occurs and causes negative outcomes in ESCC. We found that overexpression of ERBB2 inhibited radiosensitivity in vitro. Furthermore, miR-193a-5p reduced ERBB2 expression by directly targeting the 3'UTR. Increased miR-193a-5p enhanced radiosensitivity and inhibited tumorigenesis in vitro and in vivo. Additionally, low miR-193a-5p expression correlated with poor prognosis in ESCC patients, and ESCC patients with good CCRT response exhibited higher miR-193a-5p expression. Our data suggest that patients with high miR-193a-5p will likely benefit from CCRT treatment alone, however a combination of CCRT with Herceptin may be beneficial for patients with low miR-193a-5p expression.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Chemoradiotherapy/methods , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Carcinogenesis , Case-Control Studies , Cell Line, Tumor , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local , Prognosis , Receptor, ErbB-2/metabolism , Risk Factors , Trastuzumab/metabolism
13.
Front Aging Neurosci ; 8: 57, 2016.
Article in English | MEDLINE | ID: mdl-27047373

ABSTRACT

Age-related cognitive impairment has become one of the most common health threats in many countries. The biological substrate of cognition is the interconnection of neurons to form complex information processing networks. Experience-based alterations in the activities of these information processing networks lead to neuroadaptation, which is physically represented at the cellular level as synaptic plasticity. Although synaptic plasticity is known to be affected by aging, the underlying molecular mechanisms are not well described. Astrocytes, a glial cell type that is infrequently investigated in cognitive science, have emerged as energy suppliers which are necessary for meeting the abundant energy demand resulting from glutamatergic synaptic activity. Moreover, the concerted action of an astrocyte-neuron metabolic shuttle is essential for cognitive function; whereas, energetic incoordination between astrocytes and neurons may contribute to cognitive impairment. Whether altered function of the astrocyte-neuron metabolic shuttle links aging to reduced synaptic plasticity is unexplored. However, accumulated evidence documents significant beneficial effects of long-term, regular exercise on cognition and synaptic plasticity. Furthermore, exercise increases the effectiveness of astrocyte-neuron metabolic shuttle by upregulation of astrocytic lactate transporter levels. This review summarizes previous findings related to the neuronal activity-dependent astrocyte-neuron metabolic shuttle. Moreover, we discuss how aging and exercise may shape the astrocyte-neuron metabolic shuttle in cognition-associated brain areas.

14.
Am J Respir Crit Care Med ; 193(8): 869-80, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26595459

ABSTRACT

RATIONALE: Non-small cell lung cancer (NSCLC) carries a poor survival rate mainly because of metastasis. However, the molecular mechanisms that govern NSCLC metastasis have not been described. Because huntingtin-interacting protein-1 (HIP1) is known to play a role in tumorigenesis, we tested the involvement of HIP1 in NSCLC progression and metastasis. OBJECTIVES: HIP1 expression was measured in human NSCLC tumors, and correlation with survival outcome was evaluated. Furthermore, we investigated the ability of HIP1 to suppress metastasis. The molecular mechanism by which HIP1 contributes to suppress metastasis was investigated. METHODS: We used tissue arrays containing samples from 121 patients with NSCLC to analyze HIP1 expression by immunohistochemistry. To investigate the role of HIP1 expression on metastasis, we evaluated cellular mobility, migration, and invasion using lung adenocarcinoma (AdCA) cells with modified HIP1 expression levels. The human disease mouse models with the same cells were applied to evaluate the HIP1 suppressing metastasis and its mechanism in vivo. MEASUREMENTS AND MAIN RESULTS: HIP1 expression in AdCA progression was found to be an early-stage prognostic biomarker, with low expression correlated to poor prognosis. We also found HIP1 to be a metastatic suppressor in AdCA. HIP1 significantly repressed the mobility of lung cancer cells in vitro and in vivo and regulated the epithelial-mesenchymal transition by repressing AKT/glycogen synthase kinase-3ß/ß-catenin signaling. CONCLUSIONS: HIP1 serves as an early-stage prognostic biomarker and a metastatic suppressor. Reduced expression during AdCA progression can relieve HIP1 suppression of Akt-mediated epithelial-mesenchymal transition and thereby lead to development of late metastases and poor prognosis.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Vesicular Transport Proteins/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma of Lung , Biomarkers, Tumor/genetics , Female , Humans , Male , Microfilament Proteins , Middle Aged , Survival Analysis
15.
Springerplus ; 4: 597, 2015.
Article in English | MEDLINE | ID: mdl-26543732

ABSTRACT

Ineffective therapeutic treatments and inadequate repair ability in the central nervous system are disturbing problems for several neurological diseases. Fortunately, the development of clinically applicable populations of stem cells has provided an avenue to overcome the failure of endogenous repair systems and substitute new cells into the damaged brain. However, there are still several existing obstacles to translating into clinical application. Here we review the stem-cell based therapies for Parkinson's disease and discuss the potential advantages and drawbacks. We hope this review may provide suggestions for viable strategies to overcome the current technical and biological issues associated with the application of stem cells in Parkinson's disease.

16.
Exp Biol Med (Maywood) ; 240(3): 403-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662955

ABSTRACT

Pin1 is a peptidyl-prolyl isomerase which plays a critical role in many diseases including cancer and Alzheimer's disease. The essential role of Pin1 is to affect stability, localization or function of phosphoproteins by catalyzing structural changes. Among the collection of Pin1 substrates, many have been shown to be involved in regulating cell cycle progression. The cell cycle disorder caused by dysregulation of these substrates is believed to be a common phenomenon in cancer. A number of recent studies have revealed possible functions of several important Pin1-binding cell cycle regulators. Investigating the involvement of Pin1 in the cell cycle may assist in the development of future cancer therapeutics. In this review, we summarize current knowledge regarding the network of Pin1 substrates and Pin1 regulators in cell cycle progression. In G1/S progression, cyclin D1, RB, p53, p27, and cyclin E are all well-known cell cycle regulators that are modulated by Pin1. During G2/M transition, our lab has shown that Aurora A suppresses Pin1 activity through phosphorylation at Ser16 and cooperates with hBora to modulate G2/M transition. We conclude that Pin1 may be thought of as a molecular timer which modulates cell cycle progression networks.


Subject(s)
Cell Cycle/physiology , Peptidylprolyl Isomerase/physiology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Phosphoproteins/physiology , Signal Transduction/physiology
17.
PLoS One ; 8(12): e81667, 2013.
Article in English | MEDLINE | ID: mdl-24349107

ABSTRACT

Following the formation of oxidatively-induced DNA damage, several DNA glycosylases are required to initiate repair of the base lesions that are formed. Recently, NEIL1 and other DNA glycosylases, including OGG1 and NTH1 were identified as potential targets in combination chemotherapeutic strategies. The potential therapeutic benefit for the inhibition of DNA glycosylases was validated by demonstrating synthetic lethality with drugs that are commonly used to limit DNA replication through dNTP pool depletion via inhibition of thymidylate synthetase and dihydrofolate reductase. Additionally, NEIL1-associated synthetic lethality has been achieved in combination with Fanconi anemia, group G. As a prelude to the development of strategies to exploit the potential benefits of DNA glycosylase inhibition, it was necessary to develop a reliable high-throughput screening protocol for this class of enzymes. Using NEIL1 as the proof-of-principle glycosylase, a fluorescence-based assay was developed that utilizes incision of site-specifically modified oligodeoxynucleotides to detect enzymatic activity. This assay was miniaturized to a 1536-well format and used to screen small molecule libraries for inhibitors of the combined glycosylase/AP lyase activities. Among the top hits of these screens were several purine analogs, whose postulated presence in the active site of NEIL1 was consistent with the paradigm of NEIL1 recognition and excision of damaged purines. Although a subset of these small molecules could inhibit other DNA glycosylases that excise oxidatively-induced DNA adducts, they could not inhibit a pyrimidine dimer-specific glycosylase.


Subject(s)
DNA Glycosylases/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Deoxyribonuclease (Pyrimidine Dimer)/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Purines/chemistry , Small Molecule Libraries/chemistry , Animals , DNA Glycosylases/chemistry , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Deoxyribonuclease (Pyrimidine Dimer)/chemistry , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Enzyme Assays , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Kinetics , Mice , Oxidation-Reduction , Protein Binding , Purines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Small Molecule Libraries/pharmacology , Substrate Specificity
18.
Pharmaceuticals (Basel) ; 5(10): 1103-19, 2012.
Article in English | MEDLINE | ID: mdl-23226091

ABSTRACT

In neuronal systems, the health and activity of mitochondria and synapses are tightly coupled. For this reason, it has been postulated that mitochondrial abnormalities may, at least in part, drive neurodegeneration in conditions such as Alzheimer's disease (AD). Mounting evidence from multiple Alzheimer's disease cell and mouse models and postmortem brains suggest that loss of mitochondrial integrity may be a key factor that mediates synaptic loss. Therefore, the prevention or rescue of mitochondrial dysfunction may help delay or altogether prevent AD-associated neurodegeneration. Since mitochondrial health is heavily dependent on antioxidant defenses, researchers have begun to explore the use of mitochondria-targeted antioxidants as therapeutic tools to prevent neurodegenerative diseases. This review will highlight advances made using a model mitochondria-targeted antioxidant peptide, SS31, as a potential treatment for AD.

19.
Hum Mol Genet ; 21(13): 2973-90, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22492996

ABSTRACT

The purpose of this study was to investigate the protective effects of the mitochondria-targeted antioxidant catalase (MCAT) and lifespan extension in mice that express amyloid beta (Aß). Using immunoblotting and immunostaining analyses, we measured the production of full-length amyloid precursor protein (APP), soluble APPα, C-terminal fragments CTF99 and CTF83, monomeric and oligomeric Aß, Aß deposits and beta site amyloid precursor protein cleaving enzyme 1 (BACE1), in different stages of disease progression in MCAT/AßPP and AßPP mice. Using quantitative reverse transcriptase polymerase chain reaction and immunostaining analyses, we studied the expression of catalase, BACE1, the Alzheimer's disease (AD) markers, synaptophysin, APP, neprilysin, insulin-degrading enzyme and transthyretin in MCAT, AßPP, MCAT/AßPP and wild-type (WT) mice. Using the high pressure liquid chromatography analysis of 8-hydroxy-2-deoxyguanosine, we measured oxidative DNA damage in the cerebral cortical tissues from MCAT, AßPP, MCAT/AßPP and WT mice. We found that the AßPP transgenic mice that carried the human MCAT gene lived 5 months longer than did the AßPP mice. We also found that the overexpression of MCAT in the brain sections from the MCAT/AßPP transgenic mice significantly correlated with a reduction in the levels of full-length APP, CTF99, BACE1, Aß levels (40 and 42), Aß deposits and oxidative DNA damage relative to the brain sections from the AßPP mice. Interestingly, we found significantly increased levels of soluble APPα and CTF83 in the MCAT/AßPP mice, relative to the AßPP mice. These data provide direct evidence that oxidative stress plays a primary role in AD etiopathology and that in MCAT mice express Aß, MCAT prevents abnormal APP processing, reduces Aß levels and enhances Aß-degrading enzymes in mice at different ages, corresponding to different stages of disease progression. These findings indicate that mitochondria-targeted molecules may be an effective therapeutic approach to treat patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Catalase/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/pathology , Catalase/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Damage/genetics , Disease Models, Animal , Female , Insulysin/biosynthesis , Insulysin/metabolism , Male , Mice , Mice, Transgenic , Neprilysin/biosynthesis , Neuroprotective Agents/metabolism , Oxidative Stress , Prealbumin/biosynthesis , RNA, Messenger/biosynthesis , Random Allocation , Synaptophysin/biosynthesis
20.
Hum Mol Genet ; 21(2): 406-20, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21997870

ABSTRACT

The purpose of this study was to investigate the link between mutant huntingtin (Htt) and neuronal damage in relation to mitochondria in Huntington's disease (HD). In an earlier study, we determined the relationship between mutant Htt and mitochondrial dynamics/synaptic viability in HD patients. We found mitochondrial loss, abnormal mitochondrial dynamics and mutant Htt association with mitochondria in HD patients. In the current study, we sought to expand on our previous findings and further elucidate the relationship between mutant Htt and mitochondrial and synaptic deficiencies. We hypothesized that mutant Htt, in association with mitochondria, alters mitochondrial dynamics, leading to mitochondrial fragmentation and defective axonal transport of mitochondria in HD neurons. In this study, using postmortem HD brains and primary neurons from transgenic BACHD mice, we identified mutant Htt interaction with the mitochondrial protein Drp1 and factors that cause abnormal mitochondrial dynamics, including GTPase Drp1 enzymatic activity. Further, using primary neurons from BACHD mice, for the first time, we studied axonal transport of mitochondria and synaptic degeneration. We also investigated the effect of mutant Htt aggregates and oligomers in synaptic and mitochondrial deficiencies in postmortem HD brains and primary neurons from BACHD mice. We found that mutant Htt interacts with Drp1, elevates GTPase Drp1 enzymatic activity, increases abnormal mitochondrial dynamics and results in defective anterograde mitochondrial movement and synaptic deficiencies. These observations support our hypothesis and provide data that can be utilized to develop therapeutic targets that are capable of inhibiting mutant Htt interaction with Drp1, decreasing mitochondrial fragmentation, enhancing axonal transport of mitochondria and protecting synapses from toxic insults caused by mutant Htt.


Subject(s)
Axons , GTP Phosphohydrolases/metabolism , Huntington Disease/pathology , Microtubule-Associated Proteins/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Synapses/pathology , Animals , Dynamins , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...