Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
PLoS One ; 13(4): e0195909, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29649283

RESUMO

Eculizumab, a monoclonal antibody (mAb) directed against complement protein C5, is considered to be the current standard of care for patients with paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome. This study describes the generation and preclinical attributes of ALXN1210, a new long-acting anti-C5 mAb, obtained through select modifications to eculizumab to both largely abolish target-mediated drug disposition (TMDD) and increase recycling efficiency via the neonatal Fc receptor (FcRn). To attenuate the effect of TMDD on plasma terminal half-life (t1/2), histidine substitutions were engineered into the complementarity-determining regions of eculizumab to enhance the dissociation rate of the mAb:C5 complex in the acidic early endosome relative to the slightly basic pH of blood. Antibody variants with optimal pH-dependent binding to C5 exhibited little to no TMDD in mice in the presence of human C5. To further enhance the efficiency of FcRn-mediated recycling of the antibody, two additional substitutions were introduced to increase affinity for human FcRn. These substitutions yielded an additional doubling of the t½ of surrogate anti-mouse C5 antibodies with reduced TMDD in transgenic mice expressing the human FcRn. In conclusion, ALXN1210 is a promising new therapeutic candidate currently in clinical development for treatment of patients with PNH and atypical hemolytic uremic syndrome.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Complemento C5/antagonistas & inibidores , Desenho de Fármacos , Animais , Anticorpos Monoclonais/farmacocinética , Afinidade de Anticorpos , Avaliação Pré-Clínica de Medicamentos , Hemólise/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Cinética , Camundongos , Camundongos Transgênicos , Ligação Proteica , Receptores Fc/genética
3.
Neurosci Lett ; 617: 188-94, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-26892188

RESUMO

Intracerebral complement activation after severe traumatic brain injury (TBI) leads to a cascade of neuroinflammatory pathological sequelae that propagate host-mediated secondary brain injury and adverse outcomes. There are currently no specific pharmacological agents on the market to prevent or mitigate the development of secondary cerebral insults after TBI. A novel chimeric CR2-fH compound (mTT30) provides targeted inhibition of the alternative complement pathway at the site of tissue injury. This experimental study was designed to test the neuroprotective effects of mTT30 in a mouse model of closed head injury. The administration of 500 µg mTT30 i.v. at 1 h, 4 h and 24 h after head injury attenuated complement C3 deposition in injured brains, reduced the extent of neuronal cell death, and decreased post-injury microglial activation, compared to vehicle-injected placebo controls. These data imply that site-targeted alternative pathway complement inhibition may represent a new promising therapeutic avenue for the future management of severe TBI.


Assuntos
Inativadores do Complemento/uso terapêutico , Traumatismos Cranianos Fechados/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular , Complemento C3/metabolismo , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/patologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/patologia
4.
J Am Soc Nephrol ; 27(2): 405-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26047789

RESUMO

C3 glomerulopathy refers to renal disorders characterized by abnormal accumulation of C3 within the kidney, commonly along the glomerular basement membrane (GBM). C3 glomerulopathy is associated with complement alternative pathway dysregulation, which includes functional defects in complement regulator factor H (FH). There is no effective treatment for C3 glomerulopathy. We investigated the efficacy of a recombinant mouse protein composed of domains from complement receptor 2 (CR2) and FH (CR2-FH) in two models of C3 glomerulopathy with either preexisting or triggered C3 deposition along the GBM. FH-deficient mice spontaneously develop renal pathology associated with abnormal C3 accumulation along the GBM and secondary plasma C3 deficiency. CR2-FH partially restored plasma C3 levels in FH-deficient mice 2 hours after intravenous injection. CR2-FH specifically targeted glomerular C3 deposits, reduced the linear C3 reactivity assessed with anti-C3 and anti-C3b/iC3b/C3c antibodies, and prevented further spontaneous accumulation of C3 fragments along the GBM. Reduction in glomerular C3d and C9/C5b-9 reactivity was observed after daily administration of CR2-FH for 1 week. In a second mouse model with combined deficiency of FH and complement factor I, CR2-FH prevented de novo C3 deposition along the GBM. These data show that CR2-FH protects the GBM from both spontaneous and triggered C3 deposition in vivo and indicate that this approach should be tested in C3 glomerulopathy.


Assuntos
Complemento C3/antagonistas & inibidores , Membrana Basal Glomerular , Nefropatias/tratamento farmacológico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Complemento C3/metabolismo , Modelos Animais de Doenças , Membrana Basal Glomerular/metabolismo , Nefropatias/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/farmacologia
5.
Nat Commun ; 6: 10006, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26624227

RESUMO

Diseases of ectopic calcification of the vascular wall range from lethal orphan diseases such as generalized arterial calcification of infancy (GACI), to common diseases such as hardening of the arteries associated with aging and calciphylaxis of chronic kidney disease (CKD). GACI is a lethal orphan disease in which infants calcify the internal elastic lamina of their medium and large arteries and expire of cardiac failure as neonates, while calciphylaxis of CKD is a ubiquitous vascular calcification in patients with renal failure. Both disorders are characterized by vascular Mönckeburg's sclerosis accompanied by decreased concentrations of plasma inorganic pyrophosphate (PPi). Here we demonstrate that subcutaneous administration of an ENPP1-Fc fusion protein prevents the mortality, vascular calcifications and sequela of disease in animal models of GACI, and is accompanied by a complete clinical and biomarker response. Our findings have implications for the treatment of rare and common diseases of ectopic vascular calcification.


Assuntos
Doenças do Recém-Nascido/enzimologia , Doenças do Recém-Nascido/prevenção & controle , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Calcificação Vascular/enzimologia , Calcificação Vascular/prevenção & controle , Animais , Artérias/enzimologia , Artérias/patologia , Modelos Animais de Doenças , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Recém-Nascido , Doenças do Recém-Nascido/genética , Doenças do Recém-Nascido/mortalidade , Masculino , Camundongos Endogâmicos C57BL , Diester Fosfórico Hidrolases/administração & dosagem , Diester Fosfórico Hidrolases/genética , Pirofosfatases/administração & dosagem , Pirofosfatases/genética , Calcificação Vascular/genética , Calcificação Vascular/mortalidade
6.
Nat Med ; 12(5): 526-33, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16604089

RESUMO

The hypothalamus responds to circulating leptin and insulin in the control of food intake and body weight. A number of neurotransmitters in the hypothalamus, including gamma-aminobutyric acid (GABA), also have key roles in feeding. Huntingtin-associated protein 1 (Hap1) is expressed more abundantly in the hypothalamus than in other brain regions, and lack of Hap1 in mice leads to early postnatal death. Hap1 is also involved in intracellular trafficking of the GABA(A) receptor. Here, we report that fasting upregulates the expression of Hap1 in the rodent hypothalamus, whereas intracerebroventricular administration of insulin downregulates Hap1 by increasing its degradation through ubiquitination. Decreasing the expression of mouse hypothalamic Hap1 by siRNA reduces the level and activity of hypothalamic GABA(A) receptors and causes a decrease in food intake and body weight. These findings provide evidence linking hypothalamic Hap1 to GABA in the stimulation of feeding and suggest that this mechanism is involved in the feeding-inhibitory actions of insulin in the brain.


Assuntos
Ingestão de Alimentos , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de GABA-A/metabolismo , Animais , Peso Corporal , Eletrofisiologia , Jejum , Humanos , Hipotálamo/citologia , Insulina/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Ubiquitina/metabolismo
7.
J Neurosci ; 26(12): 3079-86, 2006 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-16554459

RESUMO

To investigate the role of erbB signaling in the interactions between peripheral axons and myelinating Schwann cells, we generated transgenic mice expressing a dominant-negative erbB receptor in these glial cells. Mutant mice have delayed onset of myelination, thinner myelin, shorter internodal length, and smaller axonal caliber in adulthood. Consistent with the morphological defects, transgenic mice also have slower nerve conduction velocity and defects in their responses to mechanical stimulation. Molecular analysis indicates that erbB signaling may contribute to myelin formation by regulating transcription of myelin genes. Analysis of sciatic nerves showed a reduction in the levels of expression of myelin genes in mutant mice. In vitro assays revealed that neuregulin-1 (NRG1) induces expression of myelin protein zero (P0). Furthermore, we found that the effects of NRG1 on P0 expression depend on the NRG1 isoform used. When NRG1 is presented to Schwann cells in the context of cell-cell contact, type III but not type I NRG1 regulates P0 gene expression. These results suggest that disruption of the NRG1-erbB signaling pathway could contribute to the pathogenesis of peripheral neuropathies with hypomyelination and neuropathic pain.


Assuntos
Fibras Nervosas Mielinizadas/metabolismo , Neuregulina-1/metabolismo , Proteínas Oncogênicas v-erbB/genética , Nervos Periféricos/crescimento & desenvolvimento , Células de Schwann/metabolismo , Sensação/genética , Animais , Axônios/metabolismo , Axônios/patologia , Comunicação Celular/genética , Diferenciação Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Camundongos Transgênicos , Proteína P0 da Mielina/biossíntese , Proteína P0 da Mielina/genética , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Fibras Nervosas Mielinizadas/patologia , Condução Nervosa/genética , Neuregulina-1/genética , Neuregulina-1/farmacologia , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/fisiopatologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Células de Schwann/patologia , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
8.
J Cell Biol ; 171(6): 1001-12, 2005 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-16365166

RESUMO

Huntington disease (HD) is characterized by the preferential loss of striatal medium-sized spiny neurons (MSNs) in the brain. Because MSNs receive abundant glutamatergic input, their vulnerability to excitotoxicity may be largely influenced by the capacity of glial cells to remove extracellular glutamate. However, little is known about the role of glia in HD neuropathology. Here, we report that mutant huntingtin accumulates in glial nuclei in HD brains and decreases the expression of glutamate transporters. As a result, mutant huntingtin (htt) reduces glutamate uptake in cultured astrocytes and HD mouse brains. In a neuron-glia coculture system, wild-type glial cells protected neurons against mutant htt-mediated neurotoxicity, whereas glial cells expressing mutant htt increased neuronal vulnerability. Mutant htt in cultured astrocytes decreased their protection of neurons against glutamate excitotoxicity. These findings suggest that decreased glutamate uptake caused by glial mutant htt may critically contribute to neuronal excitotoxicity in HD.


Assuntos
Neuroglia/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Adulto , Idoso , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Relação Dose-Resposta a Droga , Transportador 2 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Doença de Huntington/metabolismo , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Ratos , Proteínas da Membrana Plasmática de Transporte de Serotonina/toxicidade
9.
Am J Physiol Endocrinol Metab ; 289(4): E562-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16150954

RESUMO

It has been suggested that the developing brain is less vulnerable to the adverse effects of hypoglycemia than the mature brain; however, this issue remains controversial. We also do not know the magnitude or duration of hypoglycemia needed to trigger hypoglycemic brain injury during development. To address this issue a series of in vivo and in vitro studies were performed. First, we established an acute model of insulin-induced hypoglycemia in mice by administering 3 U/kg of neutral-protamine Hagadorn insulin subcutaneously. When we examined degenerating neurons in hippocampus and striatum by TUNEL labeling, injury was observed after 4 h of hypoglycemia in postnatal day (P)7 mice, and we observed more cell injury in animals rendered hypoglycemic at P7 than at P21. Studies of hippocampal slice cultures revealed that reduction in glucose concentration induced more neuronal injury in slices prepared from P3 and P7 than from P14 and P21 mice. Treatment of slices with an adenosine A(1) receptor (A(1)AR) antagonist reduced the hypoglycemic damage, whereas agonists increased damage, particularly in slices prepared from very young pups. This suggests a critically important role for A(1)ARs, which was further demonstrated by the reduction of hypoglycemic damage in hippocampal slices prepared from A(1)AR(-/-) mice. Furthermore, insulin-induced hypoglycemia in P7 A(1)AR(-/-) mice did not increase TUNEL-positive cells, but a major increase was seen in A(1)AR(+/-) mice. These observations show that the developing nervous system is indeed sensitive to acute hypoglycemic injury and that A(1)AR activation contributes to damage induced by hypoglycemia, particularly in immature mouse brain.


Assuntos
Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Receptor A1 de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Encefalopatias Metabólicas/etiologia , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Hipoglicemia/induzido quimicamente , Hipoglicemia/complicações , Insulina , Camundongos , Camundongos Endogâmicos C57BL
10.
J Cell Biol ; 163(1): 109-18, 2003 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-14557250

RESUMO

Although NH2-terminal mutant huntingtin (htt) fragments cause neurological disorders in Huntington's disease (HD), it is unclear how toxic htt fragments are generated and contribute to the disease process. Here, we report that complex NH2-terminal mutant htt fragments smaller than the first 508 amino acids were generated in htt-transfected cells and HD knockin mouse brains. These fragments constituted neuronal nuclear inclusions and appeared before neurological symptoms. The accumulation and aggregation of these htt fragments were associated with an age-dependent decrease in proteasome activity and were promoted by inhibition of proteasome activity. These results suggest that decreased proteasome activity contributes to late onset htt toxicity and that restoring the ability to remove NH2-terminal fragments will provide a more effective therapy for HD than inhibiting their production.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Animais , Anticorpos/imunologia , Encéfalo/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Complexo de Endopeptidases do Proteassoma
11.
J Neurosci ; 23(17): 6956-64, 2003 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-12890790

RESUMO

Huntington's disease (HD) is caused by a polyglutamine expansion in the disease protein huntingtin. The polyglutamine expansion causes huntingtin to interact abnormally with a number of proteins. However, it is unclear whether, and how, huntingtin-associated proteins are involved in the neurodegeneration in HD. Here, we show that huntingtin-associated protein-1 (HAP1), which is involved in intracellular trafficking of epidermal growth factor receptor (EGFR), is highly expressed in the hypothalamus. Mice lacking HAP1 die after birth because of depressed feeding activity. Terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling staining and electron microscopic examination revealed the degeneration in hypothalamic regions that control feeding behavior. Hypothalamic degeneration was also observed in HD transgenic mice that have a significant loss of body weight. Inhibition of HAP1 expression decreases EGFR signaling and cell viability, whereas overexpression of HAP1 enhances this signaling activity and inhibits mutant huntingtin-mediated cytotoxicity. These results suggest that the effect of mutant huntingtin on HAP1 and EGFR signaling may contribute to the hypothalamic neurodegeneration and loss of body weight in HD.


Assuntos
Doença de Huntington/genética , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/deficiência , Neurônios/metabolismo , Animais , Peso Corporal/genética , Morte Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Receptores ErbB/metabolismo , Comportamento Alimentar , Viabilidade Fetal/genética , Marcação de Genes , Homozigoto , Proteína Huntingtina , Doença de Huntington/patologia , Hipotálamo/patologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Ligação Proteica/genética , Ratos , Transdução de Sinais
12.
Nature ; 424(6948): 556-61, 2003 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12891359

RESUMO

The neuropathological hallmarks of Alzheimer's disease and other tauopathies include senile plaques and/or neurofibrillary tangles. Although mouse models have been created by overexpressing specific proteins including beta-amyloid precursor protein, presenilin and tau, no model has been generated by gene knockout. Phosphorylation of tau and other proteins on serine or threonine residues preceding proline seems to precede tangle formation and neurodegeneration in Alzheimer's disease. Notably, these phospho(Ser/Thr)-Pro motifs exist in two distinct conformations, whose conversion in some proteins is catalysed by the Pin1 prolyl isomerase. Pin1 activity can directly restore the conformation and function of phosphorylated tau or it can do so indirectly by promoting its dephosphorylation, which suggests that Pin1 is involved in neurodegeneration; however, genetic evidence is lacking. Here we show that Pin1 expression is inversely correlated with predicted neuronal vulnerability and actual neurofibrillary degeneration in Alzheimer's disease. Pin1 knockout in mice causes progressive age-dependent neuropathy characterized by motor and behavioural deficits, tau hyperphosphorylation, tau filament formation and neuronal degeneration. Thus, Pin1 is pivotal in protecting against age-dependent neurodegeneration, providing insight into the pathogenesis and treatment of Alzheimer's disease and other tauopathies.


Assuntos
Envelhecimento/fisiologia , Peptidilprolil Isomerase/metabolismo , Tauopatias/enzimologia , Tauopatias/prevenção & controle , Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/prevenção & controle , Motivos de Aminoácidos , Animais , Comportamento Animal/fisiologia , Deleção de Genes , Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Atividade Motora/fisiologia , Peptidilprolil Isomerase de Interação com NIMA , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/metabolismo , Peptidilprolil Isomerase/genética , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilação , Fosfosserina/metabolismo , Fosfotreonina/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Tauopatias/patologia , Tauopatias/fisiopatologia
13.
Hum Mol Genet ; 12(16): 2021-30, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12913073

RESUMO

In Huntington disease (HD), polyglutamine expansion causes the disease protein huntingtin to aggregate and accumulate in the nucleus and cytoplasm. The cytoplasmic huntingtin aggregates are found in axonal terminals and electrophysiological studies show that mutant huntingtin affects synaptic neurotransmission. However, the biochemical basis for huntingtin-mediated synaptic dysfunction is unclear. Using electron microscopy on sections of HD mouse brains, we found that axonal terminals containing huntingtin aggregates often had fewer synaptic vesicles than did normal axonal terminals. Subcellular fractionation and electron microscopy revealed that mutant huntingtin is co-localized with huntingtin-associated protein-1 (HAP1) in axonal terminals in the brains of HD transgenic mice. Mutant huntingtin binds more tightly to synaptic vesicles than does normal huntingtin, and it decreases the association of HAP1 with synaptic vesicles in HD mouse brains. Brain slices from HD transgenic mice that had axonal aggregates showed a significant decrease in [(3)H]glutamate release, suggesting that neurotransmitter release from synaptic vesicles was impaired. Taken together, these findings suggest that mutant huntingtin has an abnormal association with synaptic vesicles and this association impairs synaptic function.


Assuntos
Ácido Glutâmico/metabolismo , Doença de Huntington/genética , Proteínas do Tecido Nervoso/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Química Encefálica , Proteína Huntingtina , Doença de Huntington/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Imunoeletrônica , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Sinápticas/ultraestrutura
14.
J Neurosci ; 23(6): 2193-202, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12657678

RESUMO

Huntington's disease (HD) mouse models that express N-terminal huntingtin fragments show rapid disease progression and have been used for developing therapeutics. However, light microscopy reveals no significant neurodegeneration in these mice. It remains unclear how mutant huntingtin induces neurodegeneration. Using caspase staining, terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling, and electron microscopy, we observed that N171-82Q mice, which express the first 171 aa of mutant huntingtin, displayed more degenerated neurons than did other HD mouse models. The neurodegeneration was also evidenced by increased immunostaining for glial fibrillary acidic protein and ultrastructural features of apoptosis. R6/2 mice, which express exon 1 of mutant huntingtin, showed dark, nonapoptotic neurons and degenerated mitochondria associated with mutant huntingtin. In HD repeat knock-in mice (HdhCAG150), which express full-length mutant huntingtin, degenerated cytoplasmic organelles were found in both axons and neuronal cell bodies in association with mutant huntingtin that was not labeled by an antibody to huntingtin amino acids 342-456. Transfection of cultured cells with mutant huntingtin revealed that an N-terminal huntingtin fragment (amino acids 1-208 plus a 120 glutamine repeat) caused a greater increase in caspase activity than did exon 1 huntingtin and longer huntingtin fragments. These results suggest that context-dependent neurodegeneration in HD may be mediated by different N-terminal huntingtin fragments. In addition, this study has identified neurodegenerative markers for the evaluation of therapeutic treatments in HD mouse models.


Assuntos
Encéfalo/patologia , Doença de Huntington/genética , Doença de Huntington/patologia , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Nucleares/genética , Animais , Apoptose , Axônios/patologia , Axônios/ultraestrutura , Encéfalo/metabolismo , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Huntingtina , Doença de Huntington/metabolismo , Marcação In Situ das Extremidades Cortadas , Rim/citologia , Rim/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/farmacologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Proteínas Nucleares/biossíntese , Proteínas Nucleares/farmacologia , Organelas/patologia , Organelas/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Transfecção , Expansão das Repetições de Trinucleotídeos
15.
Hum Mol Genet ; 11(8): 905-14, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11971872

RESUMO

A pathological hallmark of polyglutamine diseases is the presence of inclusions or aggregates of the expanded polyglutamine protein. Polyglutamine inclusions are present in the neuronal nucleus in a number of inherited neurodegenerative disorders, including Huntington disease (HD). Recent studies suggest that polyglutamine inclusions may sequester polyglutamine-containing transcription factors and deplete their concentration in the nucleus, leading to altered gene expression. To test this hypothesis, we examined the expression and localization of the polyglutamine-containing or glutamine-rich transcription factors TBP, CBP and Sp1 in HD mouse models. All three transcription factors were diffusely distributed in the nucleus, despite the presence of abundant intranuclear inclusions. There were no differences in the nuclear staining of these transcription factors between HD and wild-type mouse brains. Although some CBP staining appeared as dots in the selective brain regions (e.g. hypothalamus and amygdala), double labeling showed that most CBP was not co-localized with huntingtin nuclear inclusions. Electron microscopy confirmed that CBP was diffusely distributed in the nucleus. Western blots showed that these transcription factors were not trapped in huntingtin inclusions. In the striatum of HD mice, which suffers a significant reduction in the expression of a number of genes, mutant huntingtin was present in both an aggregated and a diffuse form. These findings suggest that altered gene expression may result from the interactions of soluble mutant huntingtin with nuclear transcription factors, rather than from the depletion of transcription factors by nuclear inclusions.


Assuntos
Doença de Huntington/metabolismo , Corpos de Inclusão/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Proteína de Ligação a CREB , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Microscopia Eletrônica , Peptídeos/metabolismo , Fator de Transcrição Sp1/metabolismo , Proteína de Ligação a TATA-Box/metabolismo , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...