Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Audiol Neurootol ; 25(3): 111-119, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31838466

RESUMO

BACKGROUND: Inner ear structures may be included in the radiation fields when irradiation is used to treat patients with head and neck cancers. These patients may also have concurrent infections that require gentamicin treatment. Radiation and gentamicin are both potentially ototoxic, and their combined use has been shown to result in synergistic ototoxicity in animals. OBJECTIVE: We aimed to confirm the synergistic ototoxicity of combined gentamicin and low-dose irradiation treatment and identify the underlying molecular mechanisms using an in vitro model. METHOD: We compared the ototoxic effects of gentamicin, low-dose irradiation, and their combination in the OC-k3 mouse cochlear cell line using cell viability assay, live/dead stain, apoptosis detection assay, oxidative stress detection, and studied the molecular mechanisms involved using immunoblot analysis. RESULTS: Combined treatment led to prolonged oxidative stress, reduced cell viability, and synergistic apoptosis. Gentamicin induced the concurrent accumulation of LC3b-II and SQSTM1/p62, suggesting an impairment of autophagic flux. Low-dose irradiation induced transient p53 phosphorylation and persistent Akt phosphorylation in response to DNA damage. In combined treatment, gentamicin attenuated irradiation-induced Akt activation. CONCLUSIONS: Besides increased oxidative stress, synergistic apoptosis observed in combined treatment could be attributed to gentamicin-induced perturbation of autophagic flux and attenuation of Akt phosphorylation, which led to an impairment of radiation-induced DNA repair response.


Assuntos
Apoptose/efeitos dos fármacos , Cóclea/efeitos dos fármacos , Gentamicinas/efeitos adversos , Ototoxicidade/etiologia , Radioterapia/efeitos adversos , Animais , Apoptose/efeitos da radiação , Linhagem Celular , Cóclea/efeitos da radiação , Camundongos , Fosforilação
2.
J Neurochem ; 136(3): 637-50, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26561212

RESUMO

Alzheimer's disease (AD) is the leading cause of dementia in old age and is characterized by the accumulation of ß-amyloid plaques and neurofibrillary tangles (NFT). Recent studies suggest that Fyn tyrosine kinase forms part of a toxic triad with ß-amyloid and tau in the disease process. However, it is not known whether Fyn is associated with the pathological features of AD in an isoform-specific manner. In this study, we identified selective up-regulation of the alternative-spliced FynT isoform with no change in FynB in the AD neocortex. Furthermore, gene ontology term enrichment analyses and cell type-specific localization of FynT immunoreactivity suggest that FynT up-regulation was associated with neurofibrillary degeneration and reactive astrogliosis. Interestingly, significantly increased FynT in NFT-bearing neurons was concomitant to decreased FynB immunoreactivity, suggesting an involvement of alternative splicing in NFT formation. Furthermore, cultured cells of astrocytic origin have higher FynT to FynB ratio compared to those of neuronal origin. Lastly, primary rat mixed neuron-astrocyte cultures treated with Aß25-35 showed selective up-regulation of FynT expression in activated astrocytes. Our findings point to an isoform-specific role of FynT in modulating neurofibrillary degeneration and reactive astrogliosis in AD. Fyn kinase is known to interact with ß-amyloid and tau, and contributes to Alzheimer's disease pathogenesis. In this study, it is shown that the alternatively spliced FynT isoform is specifically up-regulated in the AD neocortex, with no change in FynB isoform. The increased FynT correlated with markers of neurofibrillary degeneration and reactive astrogliosis. In primary mixed cultures, treatment with amyloid peptides specifically up-regulated FynT in activated astrocytes. This study points to altered alternative splicing as a potential pathogenic mechanism in AD.


Assuntos
Doença de Alzheimer/patologia , Córtex Pré-Frontal/enzimologia , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Córtex Pré-Frontal/patologia , Isoformas de Proteínas/genética , Proteínas Tirosina Quinases/genética , Ratos , Regulação para Cima/fisiologia
3.
J Neuroinflammation ; 13(1): 251, 2016 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-27663973

RESUMO

BACKGROUND: Andrographolide is the major labdane diterpenoid originally isolated from Andrographis paniculata and has been shown to have anti-inflammatory and antioxidative effects. However, there is a dearth of studies on the potential therapeutic utility of andrographolide in neuroinflammatory conditions. Here, we aimed to investigate the mechanisms underlying andrographolide's effect on the expression of anti-inflammatory and antioxidant heme oxygenase-1 (HO-1) in primary astrocytes. METHODS: Measurements of the effects of andrograholide on antioxidant HO-1 and its transcription factor, Nrf2, include gene expression, protein turnover, and activation of putative signaling regulators. RESULTS: Andrographolide potently activated Nrf2 and also upregulated HO-1 expression in primary astrocytes. Andrographolide's effects on Nrf2 seemed to be biphasic, with acute (within 1 h) reductions in Nrf2 ubiquitination efficiency and turnover rate, followed by upregulation of Nrf2 mRNA between 8 and 24 h. The acute regulation of Nrf2 by andrographolide seemed to be independent of Keap1 and partly mediated by p38 MAPK and ERK signaling. CONCLUSIONS: These data provide further insights into the mechanisms underlying andrographolide's effects on astrocyte-mediated antioxidant, and anti-inflammatory responses and support the further assessment of andrographolide as a potential therapeutic for neurological conditions in which oxidative stress and neuroinflammation are implicated.

4.
J Neuroinflammation ; 13: 34, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26860080

RESUMO

BACKGROUND: Andrographolide is the major bioactive compound isolated from Andrographis paniculata, a native South Asian herb used medicinally for its anti-inflammatory properties. In this study, we aimed to assess andrographolide's potential utility as an anti-neuroinflammatory therapeutic. METHODS: The effects of andrographolide on lipopolysaccharide (LPS)-induced chemokine up-regulation both in mouse cortex and in cultured primary astrocytes were measured, including cytokine profiling, gene expression, and, in cultured astrocytes, activation of putative signaling regulators. RESULTS: Orally administered andrographolide significantly attenuated mouse cortical chemokine levels from the C-C and C-X-C subfamilies. Similarly, andrographolide abrogated a range of LPS-induced chemokines as well as tumor necrosis factor (TNF)-α in astrocytes. In astrocytes, the inhibitory actions of andrographolide on chemokine and TNF-α up-regulation appeared to be mediated by nuclear factor-κB (NF-κB) or c-Jun N-terminal kinase (JNK) activation. CONCLUSIONS: These results suggest that andrographolide may be useful as a therapeutic for neuroinflammatory diseases, especially those characterized by chemokine dysregulation.


Assuntos
Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Córtex Cerebral/citologia , Quimiocinas CXC/metabolismo , Diterpenos/farmacologia , Regulação para Cima/efeitos dos fármacos , Análise de Variância , Animais , Sobrevivência Celular/efeitos dos fármacos , Quimiocinas CXC/genética , Relação Dose-Resposta a Droga , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase 4/metabolismo , Camundongos , NF-kappa B/metabolismo , Ratos
5.
Brain Pathol ; 31(2): 253-266, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33128789

RESUMO

Cumulative data suggest the involvement of Fyn tyrosine kinase in Alzheimer's disease (AD). Previously, our group has shown increased immunoreactivities of the FynT isoform in AD neocortex (with no change in the alternatively spliced FynB isoform) which associated with neurofibrillary degeneration and reactive astrogliosis. Since both the aforementioned neuropathological features are also variably found in Lewy Body dementias (LBD), we investigated potential perturbations of Fyn expression in the post-mortem neocortex of patients with AD, as well as those diagnosed as having one of the two main subgroups of LBD: Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB). We found selective upregulation of FynT expression in AD, PDD, and DLB which also correlated with cognitive impairment. Furthermore, increased FynT expression correlated with hallmark neuropathological lesions, soluble ß-amyloid, and phosphorylated tau, as well as markers of microglia and astrocyte activation. In line with the human post-mortem studies, cortical FynT expression in aged mice transgenic for human P301S tau was upregulated and further correlated with accumulation of aggregated phosphorylated tau as well as with microglial and astrocytic markers. Our findings provide further evidence for the involvement of FynT in neurodegenerative dementias, likely via effects on tauopathy and neuroinflammation.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Doença por Corpos de Lewy/enzimologia , Doença por Corpos de Lewy/patologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Feminino , Humanos , Isoenzimas , Masculino , Camundongos , Camundongos Transgênicos , Regulação para Cima
6.
Acta Otolaryngol ; 128(4): 440-5, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18368580

RESUMO

CONCLUSION: L-N-Acetylcysteine (L-NAC) significantly reduced reactive oxygen species (ROS) generation and cochlear cell apoptosis after irradiation. The safe and effective use of L-NAC in reducing radiation-induced sensorineural hearing loss (SNHL) should be verified by further in vivo studies. OBJECTIVES: Radiation-induced SNHL is a common complication after radiotherapy of head and neck tumours. There is growing evidence to suggest that ROS play an important role in apoptotic cochlear cell death from ototoxicity, resulting in SNHL. The aim of this study was to evaluate the effectiveness of L-NAC, an antioxidant, on radiation-induced apoptosis in cochlear cells. MATERIALS AND METHODS: The OC-k3 cochlear cell line was studied after 0 and 20 Gy of gamma-irradiation. Cell viability assay was performed using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide. Flow cytometry and TUNEL assay were done with and without the addition of 10 mmol/L of L-NAC. Intracellular generation of ROS was detected by 2',7'-dichlorofluorescein diacetate, with comparisons made using fluorescence intensity. RESULTS: L-NAC increased the viability of cells after irradiation. Generation of ROS was demonstrated at 1 h post-irradiation and was significantly reduced by L-NAC (p<0.0001). Flow cytometry and TUNEL assay showed cell apoptosis at 72 h post-irradiation, which was diminished by the addition of L-NAC.


Assuntos
Acetilcisteína/uso terapêutico , Apoptose/efeitos dos fármacos , Cóclea/patologia , Sequestradores de Radicais Livres/uso terapêutico , Perda Auditiva Neurossensorial/prevenção & controle , Lesões Experimentais por Radiação/prevenção & controle , Animais , Apoptose/efeitos da radiação , Linhagem Celular , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Citometria de Fluxo , Perda Auditiva Neurossensorial/etiologia , Perda Auditiva Neurossensorial/patologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Espécies Reativas de Oxigênio/metabolismo
7.
Sci Rep ; 7: 43651, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28266558

RESUMO

Fyn tyrosine kinase has been implicated in the pathogenesis of Alzheimer's disease (AD). We have previously reported that upregulation of the FynT isoform in AD brains was partly associated with astrocyte activation. In this study, we demonstrated selective FynT induction in murine cortex and primary astrocyte culture after prolonged exposure to inflammatory stimulants, suggesting that FynT may mediate persistent neuroinflammation. To delineate the functional role of astrocytic FynT in association with TNF-mediated inflammatory responses, immortalized normal human astrocytes (iNHA) stably expressing FynT kinase constitutively active (FynT-CA) or kinase dead (FynT-KD) mutants were treated with TNF and compared for inflammatory responses using high-throughput real-time RT-PCR and Luminex multi-analyte immunoassays. FynT-CA but not FynT-KD mutant exhibited drastic induction of proinflammatory cytokines and chemokines after prolonged exposure to TNF, which could be attenuated by treating with Fyn kinase inhibitor PP2 or silencing via FynT-specific DsiRNA. FynT kinase activity-dependent induction of PKCδ expression, PKCδ phosphorylation, as well as NFκB activation was detected at the late phase but not the early phase of TNF signaling. In conclusion, selective FynT induction by TNF may facilitate persistent inflammatory responses in astrocytes, which is highly relevant to chronic neuroinflammation in neurodegenerative diseases including but not limited to AD.


Assuntos
Processamento Alternativo , Inflamação/genética , Inflamação/metabolismo , Proteínas Proto-Oncogênicas c-fyn/genética , Fatores de Necrose Tumoral/metabolismo , Animais , Astrócitos/metabolismo , Biomarcadores , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , NF-kappa B/metabolismo , Fosforilação , Proteína Quinase C-delta/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Necrose Tumoral/farmacologia
8.
Genom Data ; 2: 290-2, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26484111

RESUMO

Alternative splicing is a highly regulated process which generates transcriptome and proteome diversity through the skipping or inclusion of exons within gene loci. Identification of aberrant alternative splicing associated with human diseases has become feasible with the development of new genomic technologies and powerful bioinformatics. We have previously reported genome-wide gene alterations in the neocortex of a well-characterized cohort of Alzheimer's disease (AD) patients and matched elderly controls using a commercial exon microarray platform [1]. Here, we provide detailed description of analyses aimed at identifying differential alternative splicing events associated with AD.

9.
Neurochem Int ; 64: 29-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24200817

RESUMO

Synaptic dysfunction, together with neuritic plaques, neurofibrillary tangles and cholinergic neuron loss is an established finding in the Alzheimer's disease (AD) neocortex. The synaptopathology of AD is known to involve both pre- and postsynaptic components. However, the status of rabphilin 3A (RPH3A), which interacts with the SNARE complex and regulates synaptic vesicle exocytosis and Ca(2+)-triggered neurotransmitter release, is at present unclear. In this study, we measured RPH3A and its ligand Rab3A as well as several SNARE proteins in postmortem neocortex of patients with AD, and found specific reductions of RPH3A immunoreactivity compared with aged controls. RPH3A loss correlated with dementia severity, cholinergic deafferentation, and increased ß-amyloid (Aß) concentrations. Furthermore, RPH3A expression is selectively downregulated in cultured neurons treated with Aß25-35 peptides. Our data suggest that presynaptic SNARE dysfunction forms part of the synaptopathology of AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Células Cultivadas , Feminino , Humanos , Masculino , Emaranhados Neurofibrilares/metabolismo , Fragmentos de Peptídeos/farmacologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Rabfilina-3A
10.
Int J Otolaryngol ; 2010: 523976, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21151649

RESUMO

Objective. Combined cisplatin (CDDP) and radiotherapy is increasingly being used to treat advanced head and neck cancers. As both CDDP and radiation can cause hearing loss, it is important to have a better understanding of the cellular and molecular ototoxic mechanisms involved in combined therapy. Procedure. The effects of CDDP, radiation, and combined CDDP-radiation on the OC-k3 cochlear cell line were studied using MTS assay, flow cytometry, Western blotting, and microarray analysis. Results. Compared to using CDDP or radiation alone, its combined use resulted in enhanced apoptotic cell death and apoptotic-related gene expression, including that of FAS. Phosphorylation of p53 at Ser15 (a marker for p53 pathway activation in response to DNA damage) was observed after treatment with either CDDP or radiation. However, posttreatment activation of p53 occurred earlier in radiation than in CDDP which corresponded to the timings of MDM2 and TP53INP1 expression. Conclusion. Enhanced apoptotic-related gene expressions leading to increased apoptotic cell deaths could explain the synergistic ototoxicity seen clinically in combined CDDP-radiation therapy. CDDP and radiation led to differential temporal activation of p53 which suggests that their activation is the result of different upstream processes. These have implications in future antiapoptotic treatments for ototoxicity.

11.
Ann Acad Med Singap ; 38(1): 91-4, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19221677

RESUMO

INTRODUCTION: Sensori-neural hearing loss (SNHL) is a frequent complication of conventional radiotherapy for head and neck tumours, especially nasopharyngeal carcinoma. To manage radiation-induced ototoxicity appropriately, an understanding of the cellular and molecular basis of this complication is necessary. MATERIALS AND METHODS: A medline search of relevant literature was done, focusing on the radiation-induced cellular and molecular processes that lead to hair cell death in the cochlea. RESULTS: Radiation-induced SNHL occurs in the cochlea, with the retro-cochlear pathways remaining functionally intact. By simulating radiotherapy regimes used clinically, radiation-induced cochlear cell degeneration in the absence of damage to the supporting structures and blood vessels has been demonstrated in animals. This could be due to apoptotic cochlear cell death, which has been shown to be associated with p53 upregulation and intra-cellular reactive oxygen species (ROS) generation. Oxidative stress may initiate the upstream processes that lead to apoptosis and other cell death mechanisms. CONCLUSIONS: A model of radiation-induced SNHL based on a dose and ROS-dependent cochlear cell apoptosis, is proposed. This model supports the feasibility of cochlear implantation, should one be clinically indicated. It can explain clinical observations such as radiation-induced SNHL being dose-dependent and affects the high frequencies more than the lower frequencies. It also opens up the possibility of preventive strategies targeted at different stages of the apoptotic process. Antioxidants look promising as effective agents to prevent radiation-induced ototoxicity; they target upstream processes leading to different cell death mechanisms that may co-exist in the population of damaged cells.


Assuntos
Perda Auditiva Neurossensorial/etiologia , Lesões por Radiação/complicações , Animais , Morte Celular , Linhagem Celular , Cóclea/efeitos da radiação , Genes p53 , Células Ciliadas Auditivas/efeitos da radiação , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Camundongos , Espécies Reativas de Oxigênio/metabolismo
12.
J Neural Transm (Vienna) ; 115(8): 1165-72, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18506388

RESUMO

The uridine nucleotide-activated P2Y2, P2Y4 and P2Y6 receptors are widely expressed in the brain and are involved in many CNS processes, including those which malfunction in Alzheimer's disease (AD). However, the status of these receptors in the AD neocortex, as well as their putative roles in the pathogenesis of neuritic plaques and neurofibrillary tangles, remain unclear. In this study, we used immunoblotting to measure P2Y2, P2Y4 and P2Y6 receptors in two regions of the postmortem neocortex of neuropathologically assessed AD patients and aged controls. P2Y2 immunoreactivity was found to be selectively reduced in the AD parietal cortex, while P2Y4 and P2Y6 levels were unchanged. In contrast, all three receptors were preserved in the occipital cortex, which is known to be minimally affected by AD neuropathology. Furthermore, reductions in parietal P2Y2 immunoreactivity correlated both with neuropathologic scores and markers of synapse loss. These results provide a basis for considering P2Y2 receptor changes as a neurochemical substrate of AD, and point towards uridine nucleotide-activated P2Y receptors as novel targets for disease-modifying AD pharmacotherapeutic strategies.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Idoso , Especificidade de Anticorpos , Estudos de Coortes , Feminino , Humanos , Imuno-Histoquímica , Estudos Longitudinais , Masculino , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Lobo Occipital/metabolismo , Lobo Occipital/patologia , Lobo Parietal/metabolismo , Lobo Parietal/patologia , Estudos Prospectivos , Receptores Purinérgicos P2Y2 , Sinaptofisina/metabolismo
13.
Apoptosis ; 11(12): 2127-36, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17051332

RESUMO

Cisplatin and gentamycin are both ototoxic and they have been shown to induce cochlear cell apoptosis. Although radiation is also ototoxic, radiation-induced apoptosis in cochlear cells has not been studied. This study aimed to investigate the biophysical changes of dose-related radiation-induced cochlear cell apoptosis in an experimental model. Post gamma-irradiation apoptosis was demonstrated in the cochlear cell-line OC-k3 by flow cytometry and TUNEL assay. This was dose-dependant with enhanced apoptosis resulting after 20 than 5 Gy, and occurred predominantly at 72 h post-irradiation. Microarray analysis showed associated dose-dependant apoptotic gene regulation changes. Western blotting revealed p53 up-regulation of at 72 h and phosphorylation at 3, 24, 48 and 72 h after irradiation. Early activation of c-jun occurred at 3 h, but was not sustained with time. Associated dose-dependant intracellular generation of reactive oxygen species (ROS) was also demonstrated using 2', 7'-dichlorofluorescein diacetate. In conclusion, this study demonstrated a dose-dependant cochlear cell apoptosis and associated ROS generation after irradiation, with p53 possibly playing a key role. Based on this ROS-linked apoptotic model, anti-oxidants and anti-apoptotic factors could potentially be used to prevent radiation-induced sensori-neural hearing loss. As these medications can be delivered topically through the middle ear, their systematic side effects could therefore be minimized.


Assuntos
Apoptose/efeitos da radiação , Cóclea/citologia , Cóclea/efeitos da radiação , Raios gama , Animais , Apoptose/genética , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Camundongos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo
14.
Nephron ; 91(3): 431-8, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12119473

RESUMO

BACKGROUND: The asialoglycoprotein receptor (ASGPR) is a C lectin which binds and endocytoses serum glycoproteins. In humans, the ASGPR is shown mainly to occur in hepatocytes, but does occur extrahepatically in thyroid, in small and large intestines, and in the testis. In the kidney, there has been evidence both for and against its existence in mesangial cells. METHODS: Standard light microscopy examination of renal tissue stained with an antibody against the ASGPR was performed. The mRNA expression for the ASGPR H1 and H2 subunits in primary human renal proximal tubular epithelial cells (RPTEC), in the human proximal tubular epithelial cell line HK2, and in human renal cortex was investigated using reverse-transcribed nested polymerase chain reaction. ASGPR protein expression as well as ligand binding and uptake were also examined using confocal microscopy and flow cytometry (fluorescence-activated cell sorting). RESULTS: Light microscopy of paraffin renal biopsy sections stained with a polyclonal antibody against the ASGPR showed proximal tubular epithelial cell staining of the cytoplasm and particularly in the basolateral region. Renal cortex and RPTEC specifically have mRNA for both H1 and H2 subunits of the ASGPR, but HK2 only expresses mRNA for H1. Using a monoclonal antibody, the presence of the ASGPR in RPTEC was shown by fluorescence-activated cell sorting and immunofluorescent staining. Specific binding and uptake of fluorescein isothiocyanate labelled asialofetuin which is a specific ASGPR ligand was also demonstrated in RPTEC. CONCLUSIONS: Primary renal proximal tubular epithelial cells have a functional ASGPR, consisting of the H1 and H2 subunits, that is capable of specific ligand binding and uptake.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Receptor de Asialoglicoproteína/genética , Assialoglicoproteínas/química , Assialoglicoproteínas/metabolismo , Linhagem Celular , Células Cultivadas , Fetuínas , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Túbulos Renais Proximais/citologia , Microscopia de Fluorescência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , alfa-Fetoproteínas/química , alfa-Fetoproteínas/metabolismo
15.
J Biol Chem ; 279(43): 45235-44, 2004 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-15322095

RESUMO

We report the identification of a novel cDNA fragment that shows significantly reduced expression in cancerous tissue compared with paired non-cancerous liver tissue in patients with hepatocellular carcinoma (HCC). The full-length transcript of 1733 bp encodes a protein of 308 amino acids that has all the hallmark features of mitochondrial carrier proteins. We designate the novel protein as HDMCP (HCC-down-regulated mitochondrial carrier protein). The HDMCP orthologs in human, mouse, and rat are found to exhibit close similarity in protein sequence and gene organization, as well as exclusive expression in the liver. Moreover, conserved syntenic regions have been demonstrated at the HDMCP gene locus in the human, mouse, and rat genome. Taken together, we suggest that HDMCP might have a conserved and unique biological function in the liver. Overexpression of HDMCP in transiently transfected cancer cells results in the loss of staining by MitoTracker dye, indicating that HDMCP could induce the dissipation of mitochondrial membrane potential (DeltaPsim). However, HDMCP-mediated disruption of DeltaPsim is not related to mitochondrial permeability transition or apoptosis. In addition, we further demonstrate that the dissipation of DeltaPsim is accompanied by significant reduction of cellular ATP in 293T cells overexpressing HDMCP or uncoupling protein 2 (UCP2). Our present findings suggest that HDMCP might be one of the long postulated uncoupling proteins that catalyze the physiological "proton leak" in the liver. The down-regulation of HDMCP in HCC cancer cells might result in the elevation of DeltaPsim, a common phenomenon found in cancer cells.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Transporte/metabolismo , Regulação para Baixo , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/metabolismo , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Northern Blotting , Linhagem Celular , Clonagem Molecular , Sequência Conservada , DNA Complementar/metabolismo , Biblioteca Gênica , Genoma , Humanos , Marcação In Situ das Extremidades Cortadas , Canais Iônicos , Potenciais da Membrana , Camundongos , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial , Modelos Genéticos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Prótons , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção , Proteína Desacopladora 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA