Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 13(6)2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38534372

ABSTRACT

Heat shock protein 47 (HSP47), also known as SERPINH1, functions as a collagen-specific molecular chaperone protein essential for the formation and stabilization of the collagen triple helix. Here, we delved into the regulatory pathways governed by HSP47, shedding light on collagen homeostasis. Our investigation revealed a significant reduction in HSP47 mRNA levels in the skin tissue of older mice as compared to their younger counterparts. The augmented expression of HSP47 employing lentivirus infection in fibroblasts resulted in an increased secretion of type I collagen. Intriguingly, the elevated expression of HSP47 in fibroblasts correlated with increased protein and mRNA levels of type I collagen. The exposure of fibroblasts to IRE1α RNase inhibitors resulted in the reduced manifestation of HSP47-induced type I collagen secretion and expression. Notably, HSP47-overexpressing fibroblasts exhibited increased XBP1 mRNA splicing. The overexpression of HSP47 or spliced XBP1 facilitated the nuclear translocation of ß-catenin and transactivated a reporter harboring TCF binding sites on the promoter. Furthermore, the overexpression of HSP47 or spliced XBP1 or the augmentation of nuclear ß-catenin through Wnt3a induced the expression of type I collagen. Our findings substantiate that HSP47 enhances type I collagen expression and secretion in fibroblasts by orchestrating a mechanism that involves an increase in nuclear ß-catenin through IRE1α activation and XBP1 splicing. This study therefore presents potential avenues for an anti-skin-aging strategy targeting HSP47-mediated processes.


Subject(s)
Collagen Type I , HSP47 Heat-Shock Proteins , Mice , Animals , Collagen Type I/metabolism , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Endoribonucleases/metabolism , beta Catenin/metabolism , Protein Serine-Threonine Kinases/metabolism , Collagen/metabolism , Fibroblasts/metabolism , RNA, Messenger/metabolism
2.
BMB Rep ; 56(11): 612-617, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37817442

ABSTRACT

Pleiotropic regulator 1 (PLRG1), a highly conserved element in the spliceosome, can form a NineTeen Complex (NTC) with Prp19, SPF27, and CDC5L. This complex plays crucial roles in both pre-mRNA splicing and DNA repair processes. Here, we provide evidence that PLRG1 has a multifaceted impact on cancer cell proliferation. Comparing its expression levels in cancer and normal cells, we observed that PLRG1 was upregulated in various tumor tissues and cell lines. Knockdown of PLRG1 resulted in tumor-specific cell death. Depletion of PLRG1 had notable effects, including mitotic arrest, microtubule instability, endoplasmic reticulum (ER) stress, and accumulation of autophagy, ultimately culminating in apoptosis. Our results also demonstrated that PLRG1 downregulation contributed to DNA damage in cancer cells, which we confirmed through experimental validation as DNA repair impairment. Interestingly, when PLRG1 was decreased in normal cells, it induced G1 arrest as a self-protective mechanism, distinguishing it from effects observed in cancer cells. These results highlight multifaceted impacts of PLRG1 in cancer and underscore its potential as a novel anti-cancer strategy by selectively targeting cancer cells. [BMB Reports 2023; 56(11): 612-617].


Subject(s)
Cell Cycle Proteins , RNA-Binding Proteins , Humans , RNA-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , HeLa Cells , Nuclear Proteins/metabolism , Cell Proliferation/genetics , Genomic Instability , Endoplasmic Reticulum Stress , Apoptosis/genetics , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 119(51): e2203711119, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36512497

ABSTRACT

The selenium-binding protein 1 (SELENBP1) has been reported to be up-regulated in the prefrontal cortex (PFC) of schizophrenia patients in postmortem reports. However, no causative link between SELENBP1 and schizophrenia has yet been established. Here, we provide evidence linking the upregulation of SELENBP1 in the PFC of mice with the negative symptoms of schizophrenia. We verified the levels of SELENBP1 transcripts in postmortem PFC brain tissues from patients with schizophrenia and matched healthy controls. We also generated transgenic mice expressing human SELENBP1 (hSELENBP1 Tg) and examined their neuropathological features, intrinsic firing properties of PFC 2/3-layer pyramidal neurons, and frontal cortex (FC) electroencephalographic (EEG) responses to auditory stimuli. Schizophrenia-like behaviors in hSELENBP1 Tg mice and mice expressing Selenbp1 in the FC were assessed. SELENBP1 transcript levels were higher in the brains of patients with schizophrenia than in those of matched healthy controls. The hSELENBP1 Tg mice displayed negative endophenotype behaviors, including heterotopias- and ectopias-like anatomical deformities in upper-layer cortical neurons and social withdrawal, deficits in nesting, and anhedonia-like behavior. Additionally, hSELENBP1 Tg mice exhibited reduced excitabilities of PFC 2/3-layer pyramidal neurons and abnormalities in EEG biomarkers observed in schizophrenia. Furthermore, mice overexpressing Selenbp1 in FC showed deficits in sociability. These results suggest that upregulation of SELENBP1 in the PFC causes asociality, a negative symptom of schizophrenia.


Subject(s)
Schizophrenia , Humans , Animals , Mice , Schizophrenia/genetics , Schizophrenia/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Brain/metabolism , Mice, Transgenic , Selenium-Binding Proteins/genetics , Selenium-Binding Proteins/metabolism
4.
BMB Rep ; 55(12): 615-620, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36195571

ABSTRACT

The murine leukemia virus-based semi-retroviral replicating vectors (MuLV-based sRRV) had been developed to improve safety and transgene capacity for cancer gene therapy. However, despite the apparent advantages of the sRRV, improvements in the in vivo transduction efficiency are still required to deliver therapeutic genes efficiently for clinical use. In this study, we established a gibbon ape leukemia virus (GaLV) envelopepseudotyped semi-replication-competent retrovirus vector system (spRRV) which is composed of two transcomplementing replication-defective retroviral vectors termed MuLV-Gag-Pol and GaLV-Env. We found that the spRRV shows considerable improvement in efficiencies of gene transfer and spreading in both human glioblastoma cells and pre-established human glioblastoma mouse model compared with an sRRV system. When treated with ganciclovir after intratumoral injection of each vector system into pre-established U-87 MG glioblastomas, the group of mice injected with spRRV expressing the herpes simplex virus type 1-thymidine kinase (HSV1-tk) gene showed a survival rate of 100% for more than 150 days, but all control groups of mice (HSV1-tk/PBS-treated and GFP/GCV-treated groups) died within 45 days after tumor injection. In conclusion, these findings sug-gest that intratumoral delivery of the HSV1-tk gene by the spRRV system is worthy of development in clinical trials for the treatment of malignant solid tumors. [BMB Reports 2022; 55(12): 615-620].


Subject(s)
Glioblastoma , Humans , Mice , Animals , Glioblastoma/genetics , Glioblastoma/therapy , Heterografts , Genetic Therapy , Gene Transfer Techniques , Ganciclovir/pharmacology , Ganciclovir/therapeutic use , Disease Models, Animal , Genetic Vectors/genetics
5.
Exp Mol Med ; 54(9): 1461-1471, 2022 09.
Article in English | MEDLINE | ID: mdl-36056186

ABSTRACT

Mitochondria in neural progenitors play a crucial role in adult hippocampal neurogenesis by being involved in fate decisions for differentiation. However, the molecular mechanisms by which mitochondria are related to the genetic regulation of neuronal differentiation in neural progenitors are poorly understood. Here, we show that mitochondrial dysfunction induced by amyloid-beta (Aß) in neural progenitors inhibits neuronal differentiation but has no effect on the neural progenitor stage. In line with the phenotypes shown in Alzheimer's disease (AD) model mice, Aß-induced mitochondrial damage in neural progenitors results in deficits in adult hippocampal neurogenesis and cognitive function. Based on hippocampal proteome changes after mitochondrial damage in neural progenitors identified through proteomic analysis, we found that lysine demethylase 5A (KDM5A) in neural progenitors epigenetically suppresses differentiation in response to mitochondrial damage. Mitochondrial damage characteristically causes KDM5A degradation in neural progenitors. Since KDM5A also binds to and activates neuronal genes involved in the early stage of differentiation, functional inhibition of KDM5A consequently inhibits adult hippocampal neurogenesis. We suggest that mitochondria in neural progenitors serve as the checkpoint for neuronal differentiation via KDM5A. Our findings not only reveal a cell-type-specific role of mitochondria but also suggest a new role of KDM5A in neural progenitors as a mediator of retrograde signaling from mitochondria to the nucleus, reflecting the mitochondrial status.


Subject(s)
Alzheimer Disease , Neurons , Proteome , Retinoblastoma-Binding Protein 2/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Differentiation , Lysine/metabolism , Mice , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Proteome/metabolism , Proteomics
6.
Cancers (Basel) ; 11(8)2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31370279

ABSTRACT

As glioblastomas are mostly localized infiltrative lesions, gene therapy based on the retroviral replicating vector (RRV) system is considered an attractive strategy. Combinations of multiple suicide genes can circumvent the limitations associated with each gene, achieving direct and synergistic cytotoxic effects, along with bystander cell killing. In this study, we constructed a semi-and pseudotyped-RRV (sp-RRV) system harboring two suicide genes-herpes simplex virus type 1 thymidine kinase (TK) and yeast cytosine deaminase (CD)-to verify the dissemination and antitumor efficacy of our sp-RRV system (spRRVe-sEF1α-TK/sRRVgp-sEF1α-CD) in seven patient-derived glioblastoma stem-like cells (GSCs). Flow cytometry and high-content analysis revealed a wide range of transduction efficiency and good correlation between the delivery of therapeutic genes and susceptibility to the prodrugs ganciclovir and 5-fluorocytosine in patient-derived GSCs in vitro. Intra-tumoral delivery of spRRVe-sEF1α-TK/sRRVgp-sEF1α-CD, combined with prodrug treatment, synergistically inhibited cell proliferation and angiogenesis while increasing apoptosis and the depletion of tumor-associated macrophages in orthotopic glioblastoma xenografts. Genomic profiling of patient-derived GSCs revealed that the key genes preventing sp-RRV infection and transmission were associated with cell adhesion, migration, development, differentiation, and proliferation. This is the first report demonstrating that a novel sp-RRV-mediated TK/CD double suicide gene transfer system has high oncolytic power against extremely heterogeneous and treatment-refractory glioblastomas.

7.
Exp Mol Med ; 50(3): e461, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29568074

ABSTRACT

Progressive cognitive declines are the main clinical symptoms of Alzheimer's disease (AD). Cognitive impairment in AD is directly correlated with amyloid beta (Aß)-mediated synaptic deficits. It is known that upregulation of neurogranin (Ng), a postsynaptic protein, contributes to the enhancement of synaptic plasticity and cognitive function. By contrast, downregulation of Ng expression results in learning and memory impairments. Interestingly, Ng expression is significantly reduced in the parenchyma of brains with AD. However, the pathological role that downregulated Ng plays in the cognitive dysfunctions observed in AD remains unclear. Therefore, the present study examined whether enhancing Ng expression affected cognitive functions in 5XFAD mice, an animal model of AD. We found that the Ng reductions and cognitive decline observed in 5XFAD mice were restored in mice that were intrahippocampally injected with an Ng-expressing lentiviral vector. Furthermore, overexpression of Ng upregulated expression of postsynaptic density protein-95 in the hippocampus of 5XFAD mice. These results suggest that the cause of cognitive decline in AD may be at least partially associated with reduced Ng levels, and thus, supplementation of Ng may be an appropriate therapeutic strategy for individuals with AD.


Subject(s)
Cognition/physiology , Hippocampus/metabolism , Lentivirus/genetics , Neurogranin/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Brain/metabolism , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Neurogranin/genetics
8.
Genome Res ; 2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29326299

ABSTRACT

KRAS is the most frequently mutated oncogene in human tumors, and its activating mutations represent important therapeutic targets. The combination of Cas9 and guide RNA from the CRISPR-Cas system recognizes a specific DNA sequence and makes a double-strand break, which enables editing of the relevant genes. Here, we harnessed CRISPR to specifically target mutant KRAS alleles in cancer cells. We screened guide RNAs using a reporter system and validated them in cancer cells after lentiviral delivery of Cas9 and guide RNA. The survival, proliferation, and tumorigenicity of cancer cells in vitro and the growth of tumors in vivo were determined after delivery of Cas9 and guide RNA. We identified guide RNAs that efficiently target mutant KRAS without significant alterations of the wild-type allele. Doxycycline-inducible expression of this guide RNA in KRAS-mutant cancer cells transduced with a lentiviral vector encoding Cas9 disrupted the mutant KRAS gene, leading to inhibition of cancer cell proliferation both in vitro and in vivo. Intra-tumoral injection of lentivirus and adeno-associated virus expressing Cas9 and sgRNA suppressed tumor growth in vivo, albeit incompletely, in immunodeficient mice. Expression of Cas9 and the guide RNA in cells containing wild-type KRAS did not alter cell survival or proliferation either in vitro and in vivo. Our study provides a proof-of-concept that CRISPR can be utilized to target driver mutations of cancers in vitro and in vivo.

9.
Aging (Albany NY) ; 8(9): 2062-2080, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27658230

ABSTRACT

Skeletal muscle mass and power decrease with age, leading to impairment of mobility and metabolism in the elderly. Ca2+ signaling is crucial for myoblast differentiation as well as muscle contraction through activation of transcription factors and Ca2+-dependent kinases and phosphatases. Ca2+ channels, such as dihydropyridine receptor (DHPR), two-pore channel (TPC) and inositol 1,4,5-triphosphate receptor (ITPR), function to maintain Ca2+ homeostasis in myoblasts. Here, we observed a significant decrease in expression of type 1 IP3 receptor (ITPR1), but not types 2 and 3, in aged mice skeletal muscle and isolated myoblasts, compared with those of young mice. ITPR1 knockdown using shRNA-expressing viruses in C2C12 myoblasts and tibialis anterior muscle of mice inhibited myotube formation and muscle regeneration after injury, respectively, a typical phenotype of aged muscle. This aging phenotype was associated with repression of muscle-specific genes and activation of the epidermal growth factor receptor (EGFR)-Ras-extracellular signal-regulated kinase (ERK) pathway. ERK inhibition by U0126 not only induced recovery of myotube formation in old myoblasts but also facilitated muscle regeneration after injury in aged muscle. The conserved decline in ITPR1 expression in aged human skeletal muscle suggests utility as a potential therapeutic target for sarcopenia, which can be treated using ERK inhibition strategies.


Subject(s)
Aging/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , Regeneration/physiology , Adult , Age Factors , Aged , Aging/genetics , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Middle Aged , Muscle Development/physiology , Muscle, Skeletal/metabolism , Phenotype , Signal Transduction
10.
EMBO Rep ; 16(10): 1318-33, 2015 10.
Article in English | MEDLINE | ID: mdl-26341627

ABSTRACT

The mitochondrial calcium uniporter (MCU) is responsible for mitochondrial calcium uptake and homeostasis. It is also a target for the regulation of cellular anti-/pro-apoptosis and necrosis by several oncogenes and tumour suppressors. Herein, we report the crystal structure of the MCU N-terminal domain (NTD) at a resolution of 1.50 Å in a novel fold and the S92A MCU mutant at 2.75 Å resolution; the residue S92 is a predicted CaMKII phosphorylation site. The assembly of the mitochondrial calcium uniporter complex (uniplex) and the interaction with the MCU regulators such as the mitochondrial calcium uptake-1 and mitochondrial calcium uptake-2 proteins (MICU1 and MICU2) are not affected by the deletion of MCU NTD. However, the expression of the S92A mutant or a NTD deletion mutant failed to restore mitochondrial Ca(2+) uptake in a stable MCU knockdown HeLa cell line and exerted dominant-negative effects in the wild-type MCU-expressing cell line. These results suggest that the NTD of MCU is essential for the modulation of MCU function, although it does not affect the uniplex formation.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium-Binding Proteins/metabolism , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Models, Molecular , Mutation , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Structure, Tertiary
11.
J Psychiatry Neurosci ; 40(2): 78-88, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25268789

ABSTRACT

BACKGROUND: Decreased expression of phospholipase C-ß1 (PLC-ß1) has been observed in the brains of patients with schizophrenia, but, to our knowledge, no studies have shown a possible association between this altered PLC-ß1 expression and the pathogenesis of schizophrenia. Although PLC-ß1-null (PLC-ß1(-/-)) mice exhibit multiple endophenotypes of schizophrenia, it remains unclear how regional decreases in PLC-ß1 expression in the brain contribute to specific behavioural defects. METHODS: We selectively knocked down PLC-ß1 in the medial prefrontal cortex (mPFC) using a small hairpin RNA strategy in mice. RESULTS: Silencing PLC-ß1 in the mPFC resulted in working memory deficits, as assayed using the delayed non-match-to-sample T-maze task. Notably, however, other schizophrenia-related behaviours observed in PLC-ß1-/- mice, including phenotypes related to locomotor activity, sociability and sensorimotor gating, were normal in PLC-ß1 knockdown mice. LIMITATIONS: Phenotypes of PLC-ß1 knockdown mice, such as locomotion, anxiety and sensorimotor gating, have already been published in our previous studies. Further, the neural mechanisms underlying the working memory deficit in mice may be different from those in human schizophrenia. CONCLUSION: These results indicate that PLC-ß1 signalling in the mPFC is required for working memory. Importantly, these results support the notion that the decrease in PLC-ß1 expression in the brains of patients with schizophrenia is a pathogenically relevant molecular marker of the disorder.


Subject(s)
Memory, Short-Term/physiology , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Prefrontal Cortex/physiopathology , Schizophrenia/physiopathology , Animals , Anxiety/physiopathology , Disease Models, Animal , Endophenotypes , Gene Knockdown Techniques , Locomotion/physiology , Male , Maze Learning/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phospholipase C beta/deficiency , Prepulse Inhibition/physiology , Reflex, Startle/physiology , Schizophrenic Psychology , Social Behavior
12.
Arch Pharm Res ; 38(8): 1552-60, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25269537

ABSTRACT

Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid (ω-3 PUFA) that protects against photodamage and photocarcinogenesis in mammals. Aquaporin-3 (AQP3) is a water/glycerol transport protein that is found in basal layer keratinocytes. In this study, we have investigated the protective effect of EPA against ultraviolet B (UVB)-induced AQP3 downregulation in human keratinocytes. EPA treatment was found to increase AQP3 gene and protein expression in human epidermal keratinocytes (HaCaT). Using a specific inhibitor, we observed that the effect of EPA on AQP3 expression was mediated by extracellular signal-regulated kinase (ERK) activation. UVB radiation induced AQP3 downregulation in HaCaT cells, and it was found that EPA treatment attenuated UVB-induced AQP3 reduction and the associated cell death. UVB-induced downregulation of AQP3 was blocked by EPA and p38 inhibitor SB203580. Collectively, the present results show that EPA increased AQP3 expression and that this led to a reduction UVB-induced photodamage.


Subject(s)
Aquaporin 3/drug effects , Aquaporin 3/radiation effects , Eicosapentaenoic Acid/pharmacology , Keratinocytes/drug effects , Keratinocytes/radiation effects , Ultraviolet Rays/adverse effects , Animals , Aquaporin 3/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Down-Regulation/radiation effects , Female , Humans , Keratinocytes/metabolism , Mice , Mice, Hairless
13.
Biochem J ; 447(3): 371-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22867515

ABSTRACT

The structural proximity and functional coupling between the SR (sarcoplasmic reticulum) and mitochondria have been suggested to occur in the heart. However, the molecular architecture involved in the SR-mitochondrial coupling remains unclear. In the present study, we performed various genetic and Ca2+-probing studies to resolve the proteins involved in the coupling process. By using the bacterial 2-hybrid, glutathione transferase pull-down, co-immunoprecipitation and immunocytochemistry assays, we found that RyR2 (ryanodine receptor type 2), which is physically associated with VDAC2 (voltage-dependent anion channel 2), was co-localized in SR-mitochondrial junctions. Furthermore, a fractionation study revealed that VDAC2 was co-localized with RyR2 only in the subsarcolemmal region. VDAC2 knockdown by targeted short hairpin RNA led to an increased diastolic [Ca2+] (calcium concentration) and abolishment of mitochondrial Ca2+ uptake. Collectively, the present study suggests that the coupling of VDAC2 with RyR2 is essential for Ca2+ transfer from the SR to mitochondria in the heart.


Subject(s)
Calcium/metabolism , Mitochondria, Heart/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Voltage-Dependent Anion Channel 2/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Humans , In Vitro Techniques , Ion Transport , Male , Mice , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Voltage-Dependent Anion Channel 2/genetics
14.
Cell Calcium ; 49(2): 136-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21241999

ABSTRACT

Voltage-dependent anion channels (VDACs) are pore forming proteins predominantly found in the outer mitochondrial membrane and are thought to transport Ca(2+). In this study, we have investigated the possible role of type 2 VDAC (VDAC2) in cardiac Ca(2+) signaling and Ca(2+) sparks using a lentiviral knock-down (KD) technique and two-dimensional confocal Ca(2+) imaging in immortalized autorhythmic adult atrial cells, HL-1. We confirmed high expression of VDAC2 protein in ventricular, atrial, and HL-1 cells using Western blot analysis. Infection of HL-1 cells with VDAC2-targeting lentivirus reduced the level of VDAC2 protein to ∼10%. Comparisons of autorhythmic Ca(2+) transients between wild-type (WT) and VDAC2 KD cells showed no significant change in the magnitude, decay, and beating rate of the Ca(2+) transients. Caffeine (10mM)-induced Ca(2+) release, which indicates sarcoplasmic reticulum (SR) Ca(2+) content, was not altered by VDAC2 KD. Interestingly, however, the intensity, width, and duration of the individual Ca(2+) sparks were significantly increased by VDAC2 KD in resting conditions, with no change in the frequency of sparks. VDAC2 KD significantly delayed mitochondrial Ca(2+) uptake during artificial Ca(2+) pulses in permeabilized HL-1 cells. These results suggest that VDAC2 may facilitate mitochondrial Ca(2+) uptake and restrict Ca(2+) spark expansion without regulating activations of sparks under resting conditions, thereby providing evidence on the functional role of VDAC2 in cardiac local Ca(2+) signaling.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Myocytes, Cardiac/metabolism , Voltage-Dependent Anion Channel 2/metabolism , Action Potentials , Animals , Caffeine/pharmacology , Cell Line, Transformed , Mice , Myocytes, Cardiac/cytology , RNA Interference , RNA, Small Interfering/metabolism , Sarcoplasmic Reticulum/metabolism , Voltage-Dependent Anion Channel 2/genetics
15.
Virus Genes ; 40(1): 37-43, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19859796

ABSTRACT

The Hepatitis B virus (HBV) is a causative agent of acute chronic hepatitis, cirrhosis, and hepatocarcinoma. The Hepatitis B virus X protein (HBx) has pleiotypic functions in the regulation of proliferation and apoptosis. It has been suggested that the anti-inflammatory drug sulfasalazine, which is commonly used to treat rheumatoid arthritis and inflammatory bowel disease, inhibits nuclear factor NF-kappaB and induces cell death in HBx-expressing liver cells. In this study, we demonstrate that sulfasalazine induces cell death via apoptosis in HBx-expressing liver cells, as evidenced by characteristic changes in nuclear morphology, cleavage of poly (ADP-ribose) polymerase (PARP), caspase-3 and caspase-9, and activation of caspase-3. We also demonstrate that inhibition of NF-kappaB by siRNA fails to induce apoptosis of HBx-expressing liver cells, indicating that sulfasalazine modulates apoptosis of HBx-expressing cells in an NF-kappaB-independent manner.


Subject(s)
Apoptosis/drug effects , Hepatitis B virus/metabolism , NF-kappa B/metabolism , Sulfasalazine/pharmacology , Trans-Activators/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Enzyme Activation/drug effects , Humans , Liver/cytology , Liver/drug effects , Liver/metabolism , NF-kappa B/genetics , Poly(ADP-ribose) Polymerases/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...