Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
PLoS One ; 18(11): e0294312, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38033125

RESUMO

Lysosomes play important roles in catabolism, nutrient sensing, metabolic signaling, and homeostasis. NPC1 deficiency disrupts lysosomal function by inducing cholesterol accumulation that leads to early neurodegeneration in Niemann-Pick type C (NPC) disease. Mitochondria pathology and deficits in NPC1 deficient cells are associated with impaired lysosomal proteolysis and metabolic signaling. It is thought that activation of the transcription factor TFEB, an inducer of lysosome biogenesis, restores lysosomal-autophagy activity in lysosomal storage disorders. Here, we investigated the effect of trehalose, a TFEB activator, in the mitochondria pathology of NPC1 mutant fibroblasts in vitro and in mouse developmental Purkinje cells ex vivo. We found that in NPC1 mutant fibroblasts, serum starvation or/and trehalose treatment, both activators of TFEB, reversed mitochondria fragmentation to a more tubular mitochondrion. Trehalose treatment also decreased the accumulation of Filipin+ cholesterol in NPC1 mutant fibroblasts. However, trehalose treatment in cerebellar organotypic slices (COSCs) from wild-type and Npc1nmf164 mice caused mitochondria fragmentation and lack of dendritic growth and degeneration in developmental Purkinje cells. Our data suggest, that although trehalose successfully restores mitochondria length and decreases cholesterol accumulation in NPC1 mutant fibroblasts, in COSCs, Purkinje cells mitochondria and dendritic growth are negatively affected possibly through the overactivation of the TFEB-lysosomal-autophagy pathway.


Assuntos
Mitocôndrias , Doença de Niemann-Pick Tipo C , Trealose , Animais , Humanos , Camundongos , Colesterol/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Células de Purkinje/patologia , Trealose/farmacologia
2.
Sci Rep ; 13(1): 5665, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37024714

RESUMO

Purkinje cell (PC) loss occurs at an early age in patients and animal models of Niemann-Pick Type C (NPC), a lysosomal storage disease caused by mutations in the Npc1 or Npc2 genes. Although degeneration of PCs occurs early in NPC, little is known about how NPC1 deficiency affects the postnatal development of PCs. Using the Npc1nmf164 mouse model, we found that NPC1 deficiency significantly affected the postnatal development of PC dendrites and synapses. The developing dendrites of Npc1nmf164 PCs were significantly deficient in mitochondria and lysosomes. Furthermore, anabolic (mTORC1) and catabolic (TFEB) signaling pathways were not only perturbed but simultaneously activated in NPC1-deficient PCs, suggesting a loss of metabolic balance. We also found that mice with conditional heterozygous deletion of the Phosphatase and Tensin Homolog Deleted on Chromosome 10 gene (Pten-cHet), an inhibitor of mTORC1, showed similar early dendritic alterations in PCs to those found in Npc1-deficient mice. However, in contrast to Npc1nmf164 mice, Pten-cHet mice exhibited the overactivation of the mTORC1 pathway but with a strong inhibition of TFEB signaling, along with no dendritic mitochondrial reductions by the end of their postnatal development. Our data suggest that disruption of the lysosomal-metabolic signaling in PCs causes dendritic and synaptic developmental deficits that precede and promote their early degeneration in NPC.


Assuntos
Doença de Niemann-Pick Tipo C , Células de Purkinje , Camundongos , Animais , Células de Purkinje/metabolismo , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Animais de Doenças , Lisossomos/metabolismo
3.
Development ; 147(21)2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32611604

RESUMO

Little is known about the effects of NPC1 deficiency in brain development and whether these effects contribute to neurodegeneration in Niemann-Pick disease type C (NPC). Degeneration of cerebellar Purkinje cells occurs at an earlier stage and to a greater extent in NPC; therefore, we analyzed the effect of NPC1 deficiency on microglia and on climbing fiber synaptic refinement during cerebellar postnatal development using the Npc1nmf164 mouse. Our analysis revealed that NPC1 deficiency leads to early phenotypic changes in microglia that are not associated with an innate immune response. However, the lack of NPC1 in Npc1nmf164 mice significantly affected the early development of microglia by delaying the radial migration, increasing the proliferation and impairing the differentiation of microglia precursor cells during postnatal development. Additionally, increased phagocytic activity of differentiating microglia was observed at the end of the second postnatal week in Npc1nmf164 mice. Moreover, significant climbing fiber synaptic refinement deficits along with an increased engulfment of climbing fiber synaptic elements by microglia were found in Npc1nmf164 mice, suggesting that profound developmental defects in microglia and synaptic connectivity might precede and predispose Purkinje cells to early neurodegeneration in NPC.


Assuntos
Cerebelo/crescimento & desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Microglia/metabolismo , Microglia/patologia , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Movimento Celular , Proliferação de Células , Cerebelo/imunologia , Modelos Animais de Doenças , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos Endogâmicos C57BL , Proteína C1 de Niemann-Pick , Fagocitose , Sinapses/metabolismo , Desmame
4.
eNeuro ; 7(3)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32273396

RESUMO

Age-related cognitive decline and many dementias involve complex interactions of both genetic and environmental risk factors. Recent evidence has demonstrated a strong association of obesity with the development of dementia. Furthermore, white matter damage is found in obese subjects and mouse models of obesity. Here, we found that components of the complement cascade, including complement component 1qa (C1QA) and C3 are increased in the brain of Western diet (WD)-fed obese mice, particularly in white matter regions. To functionally test the role of the complement cascade in obesity-induced brain pathology, female and male mice deficient in C1QA, an essential molecule in the activation of the classical pathway of the complement cascade, were fed a WD and compared with WD-fed wild type (WT) mice, and to C1qa knock-out (KO) and WT mice fed a control diet (CD). C1qa KO mice fed a WD became obese but did not show pericyte loss or a decrease in laminin density in the cortex and hippocampus that was observed in obese WT controls. Furthermore, obesity-induced microglia phagocytosis and breakdown of myelin in the corpus callosum were also prevented by deficiency of C1QA. Collectively, these data show that C1QA is necessary for damage to the cerebrovasculature and white matter damage in diet-induced obesity.


Assuntos
Complemento C1q , Substância Branca , Animais , Complemento C1q/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade , Substância Branca/metabolismo
5.
Sci Rep ; 9(1): 14722, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31605022

RESUMO

Niemann Pick Type-C disease (NPC) is an inherited lysosomal storage disease (LSD) caused by pathogenic variants in the Npc1 or Npc2 genes that lead to the accumulation of cholesterol and lipids in lysosomes. NPC1 deficiency causes neurodegeneration, dementia and early death. Cerebellar Purkinje cells (PCs) are particularly hypersensitive to NPC1 deficiency and degenerate earlier than other neurons in the brain. Activation of microglia is an important contributor to PCs degeneration in NPC. However, the mechanisms by which activated microglia promote PCs degeneration in NPC are not completely understood. Here, we are demonstrating that in the Npc1nmf164 mouse cerebellum, microglia in the molecular layer (ML) are activated and contacting dendrites at early stages of NPC, when no loss of PCs is detected. During the progression of PCs degeneration in Npc1nmf164 mice, accumulation of phagosomes and autofluorescent material in microglia at the ML coincided with the degeneration of dendrites and PCs. Feeding Npc1nmf164 mice a western diet (WD) increased microglia activation and corresponded with a more extensive degeneration of dendrites but not PC somata. Together our data suggest that microglia contribute to the degeneration of PCs by interacting, engulfing and phagocytosing their dendrites while the cell somata are still present.


Assuntos
Dendritos/metabolismo , Microglia/metabolismo , Degeneração Neural/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Células de Purkinje/metabolismo , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Dieta Ocidental , Modelos Animais de Doenças , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/genética , Proteína C1 de Niemann-Pick , Fagocitose/genética , Fagossomos/metabolismo
6.
Invest Ophthalmol Vis Sci ; 60(10): 3283-3296, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369031

RESUMO

Purpose: Glaucoma is a complex disease with major risk factors including advancing age and increased intraocular pressure (IOP). Dissecting these earliest events will likely identify new avenues for therapeutics. Previously, we performed transcriptional profiling in DBA/2J (D2) mice, a widely used mouse model relevant to glaucoma. Here, we use these data to identify and test regulators of early gene expression changes in DBA/2J glaucoma. Methods: Upstream regulator analysis (URA) in Ingenuity Pathway Analysis was performed to identify potential master regulators of differentially expressed genes. The function of one putative regulator, mesenchyme homeobox 2 (Meox2), was tested using a combination of genetic, biochemical, and immunofluorescence approaches. Results: URA identified Meox2 as a potential regulator of early gene expression changes in the optic nerve head (ONH) of DBA/2J mice. Meox2 haploinsufficiency did not affect the characteristic diseases of the iris or IOP elevation seen in DBA/2J mice but did cause a significant increase in the numbers of eyes with axon damage compared to controls. While young mice appeared normal, aged Meox2 haploinsufficient DBA/2J mice showed a 44% reduction in MEOX2 protein levels. This correlated with modulation of age- and disease-specific vascular and myeloid alterations. Conclusions: Our data support a model whereby Meox2 controls IOP-dependent vascular remodeling and neuroinflammation to promote axon survival. Promoting these earliest responses prior to IOP elevation may be a viable neuroprotective strategy to delay or prevent human glaucoma.


Assuntos
Axônios/patologia , Glaucoma/genética , Haploinsuficiência/genética , Proteínas de Homeodomínio/genética , Degeneração Neural/genética , Disco Óptico/patologia , Células Ganglionares da Retina/patologia , Animais , Pressão Sanguínea/fisiologia , Western Blotting , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/fisiologia , Glaucoma/patologia , Pressão Intraocular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Degeneração Neural/patologia , Microscopia com Lâmpada de Fenda
7.
PLoS Genet ; 15(5): e1008155, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31150388

RESUMO

Classical laboratory strains show limited genetic diversity and do not harness natural genetic variation. Mouse models relevant to Alzheimer's disease (AD) have largely been developed using these classical laboratory strains, such as C57BL/6J (B6), and this has likely contributed to the failure of translation of findings from mice to the clinic. Therefore, here we test the potential for natural genetic variation to enhance the translatability of AD mouse models. Two widely used AD-relevant transgenes, APPswe and PS1de9 (APP/PS1), were backcrossed from B6 to three wild-derived strains CAST/EiJ, WSB/EiJ, PWK/PhJ, representative of three Mus musculus subspecies. These new AD strains were characterized using metabolic, functional, neuropathological and transcriptional assays. Strain-, sex- and genotype-specific differences were observed in cognitive ability, neurodegeneration, plaque load, cerebrovascular health and cerebral amyloid angiopathy. Analyses of brain transcriptional data showed strain was the greatest driver of variation. We identified significant variation in myeloid cell numbers in wild type mice of different strains as well as significant differences in plaque-associated myeloid responses in APP/PS1 mice between the strains. Collectively, these data support the use of wild-derived strains to better model the complexity of human AD.


Assuntos
Doença de Alzheimer/genética , Modelos Animais de Doenças , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Selvagens/genética , Encéfalo/metabolismo , Variação Genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide , Presenilina-1/genética , Reprodutibilidade dos Testes
8.
Annu Rev Vis Sci ; 5: 99-122, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31226014

RESUMO

Inflammation of the blood vessels that serve the central nervous system has been increasingly identified as an early and possibly initiating event among neurodegenerative conditions such as Alzheimer's disease and related dementias. However, the causal relevance of vascular inflammation to major retinal degenerative diseases is unresolved. Here, we describe how genetics, aging-associated changes, and environmental factors contribute to vascular inflammation in age-related macular degeneration, diabetic retinopathy, and glaucoma. We highlight the importance of mouse models in studying the underlying mechanisms and possible treatments for these diseases. We conclude that data support vascular inflammation playing a central if not primary role in retinal degenerative diseases, and this association should be a focus of future research.


Assuntos
Retinopatia Diabética/fisiopatologia , Glaucoma/fisiopatologia , Degeneração Macular/fisiopatologia , Vasculite Retiniana/fisiopatologia , Envelhecimento/fisiologia , Animais , Modelos Animais de Doenças , Interação Gene-Ambiente , Humanos , Inflamação/fisiopatologia , Fatores de Risco
9.
Cell Death Dis ; 9(6): 705, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29899326

RESUMO

The cJun N-terminal kinases (JNKs; JNK1, JNK2, and JNK3) promote degenerative processes after neuronal injury and in disease. JNK2 and JNK3 have been shown to promote retinal ganglion cell (RGC) death after optic nerve injury. In their absence, long-term survival of RGC somas is significantly increased after mechanical optic nerve injury. In glaucoma, because optic nerve damage is thought to be a major cause of RGC death, JNKs are an important potential target for therapeutic intervention. To assess the role of JNK2 and JNK3 in an ocular hypertensive model of glaucoma, null alleles of Jnk2 and Jnk3 were backcrossed into the DBA/2J (D2) mouse. JNK activation occurred in RGCs following increased intraocular pressure in D2 mice. However, deficiency of both Jnk2 and Jnk3 together did not lessen optic nerve damage or RGC death. These results differentiate the molecular pathways controlling cell death in ocular hypertensive glaucoma compared with mechanical optic nerve injury. It is further shown that JUN, a pro-death component of the JNK pathway in RGCs, can be activated in glaucoma in the absence of JNK2 and JNK3. This implicates JNK1 in glaucomatous RGC death. Unexpectedly, at younger ages, Jnk2-deficient mice were more likely to develop features of glaucomatous neurodegeneration than D2 mice expressing Jnk2. This appears to be due to a neuroprotective effect of JNK2 and not due to a change in intraocular pressure. The Jnk2-deficient context also unmasked a lesser role for Jnk3 in glaucoma. Jnk2 and Jnk3 double knockout mice had a modestly increased risk of neurodegeneration compared with mice only deficient in Jnk2. Overall, these findings are consistent with pleiotropic effects of JNK isoforms in glaucoma and suggest caution is warranted when using JNK inhibitors to treat chronic neurodegenerative conditions.


Assuntos
Glaucoma/enzimologia , Glaucoma/patologia , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Hipertensão Ocular/enzimologia , Hipertensão Ocular/patologia , Animais , Axônios/metabolismo , Morte Celular , Ativação Enzimática , Regulação da Expressão Gênica , Glaucoma/fisiopatologia , Pressão Intraocular , Camundongos Endogâmicos DBA , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Degeneração Neural/fisiopatologia , Hipertensão Ocular/fisiopatologia , Nervo Óptico/enzimologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Retina/enzimologia , Retina/patologia , Retina/fisiopatologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
10.
Neurobiol Aging ; 42: 50-60, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27143421

RESUMO

Evidence suggests that multiple genetic and environmental factors conspire together to increase susceptibility to Alzheimer's disease (AD). The amyloid cascade hypothesis states that deposition of the amyloid-ß (Aß) peptide is central to AD; however, evidence in humans and animals suggests that Aß buildup alone is not sufficient to cause neuronal cell loss and cognitive decline. Mouse models that express high levels of mutant forms of amyloid precursor protein and/or cleaving enzymes deposit amyloid but do not show neuron loss. Therefore, a double-hit hypothesis for AD has been proposed whereby vascular dysfunction precedes and promotes Aß toxicity. In support of this, copy number variations in mesenchyme homeobox 2 (MEOX2), a gene involved in vascular development, are associated with severe forms of AD. However, the role of MEOX2 in AD has not been studied. Here, we tested Meox2 haploinsufficiency in B6.APP/PS1 (B6.APB(Tg)) mice, a mouse model of AD. Despite no overt differences in plaque deposition or glial activation, B6.APB(Tg) mice that carry only one copy of Meox2 (B6.APB(Tg).Mx(-/+)) show increased neuronal cell loss, particularly in regions containing plaques, compared with B6.APB(Tg) mice. Neuronal cell loss corresponds with a significant decrease in plaque-associated microvessels, further supporting a synergistic effect of vascular compromise and amyloid deposition on neuronal cell dysfunction in AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Haploinsuficiência/genética , Haploinsuficiência/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Neurônios/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Memória Espacial
11.
Sci Rep ; 6: 21568, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26888450

RESUMO

Studies have assessed individual components of a western diet, but no study has assessed the long-term, cumulative effects of a western diet on aging and Alzheimer's disease (AD). Therefore, we have formulated the first western-style diet that mimics the fat, carbohydrate, protein, vitamin and mineral levels of western diets. This diet was fed to aging C57BL/6J (B6) mice to identify phenotypes that may increase susceptibility to AD, and to APP/PS1 mice, a mouse model of AD, to determine the effects of the diet in AD. Astrocytosis and microglia/monocyte activation were dramatically increased in response to diet and was further increased in APP/PS1 mice fed the western diet. This increase in glial responses was associated with increased plaque burden in the hippocampus. Interestingly, given recent studies highlighting the importance of TREM2 in microglia/monocytes in AD susceptibility and progression, B6 and APP/PS1 mice fed the western diet showed significant increases TREM2+ microglia/monocytes. Therefore, an increase in TREM2+ microglia/monocytes may underlie the increased risk from a western diet to age-related neurodegenerative diseases such as Alzheimer's disease. This study lays the foundation to fully investigate the impact of a western diet on glial responses in aging and Alzheimer's disease.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Dieta Ocidental/efeitos adversos , Gliose/metabolismo , Microglia/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Gliose/genética , Gliose/patologia , Camundongos , Camundongos Transgênicos , Microglia/patologia
12.
PLoS Biol ; 13(10): e1002279, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26512759

RESUMO

Aging is the major risk factor for neurodegenerative diseases such as Alzheimer's disease, but little is known about the processes that lead to age-related decline of brain structures and function. Here we use RNA-seq in combination with high resolution histological analyses to show that aging leads to a significant deterioration of neurovascular structures including basement membrane reduction, pericyte loss, and astrocyte dysfunction. Neurovascular decline was sufficient to cause vascular leakage and correlated strongly with an increase in neuroinflammation including up-regulation of complement component C1QA in microglia/monocytes. Importantly, long-term aerobic exercise from midlife to old age prevented this age-related neurovascular decline, reduced C1QA+ microglia/monocytes, and increased synaptic plasticity and overall behavioral capabilities of aged mice. Concomitant with age-related neurovascular decline and complement activation, astrocytic Apoe dramatically decreased in aged mice, a decrease that was prevented by exercise. Given the role of APOE in maintaining the neurovascular unit and as an anti-inflammatory molecule, this suggests a possible link between astrocytic Apoe, age-related neurovascular dysfunction and microglia/monocyte activation. To test this, Apoe-deficient mice were exercised from midlife to old age and in contrast to wild-type (Apoe-sufficient) mice, exercise had little to no effect on age-related neurovascular decline or microglia/monocyte activation in the absence of APOE. Collectively, our data shows that neurovascular structures decline with age, a process that we propose to be intimately linked to complement activation in microglia/monocytes. Exercise prevents these changes, but not in the absence of APOE, opening up new avenues for understanding the complex interactions between neurovascular and neuroinflammatory responses in aging and neurodegenerative diseases such as Alzheimer's disease.


Assuntos
Envelhecimento , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Complemento C1q/metabolismo , Atividade Motora , Doenças Neurodegenerativas/prevenção & controle , Doenças Vasculares/prevenção & controle , Animais , Apolipoproteínas E/sangue , Apolipoproteínas E/química , Apolipoproteínas E/genética , Astrócitos/imunologia , Astrócitos/patologia , Astrócitos/ultraestrutura , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/ultraestrutura , Ativação do Complemento , Complemento C1q/genética , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/ultraestrutura , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Homozigoto , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Monócitos/ultraestrutura , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Plasticidade Neuronal , Acoplamento Neurovascular , Estabilidade Proteica , Doenças Vasculares/imunologia , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
13.
PLoS One ; 9(9): e107291, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25207540

RESUMO

Some pathological manifestations of diabetes in the eye include retinopathy, cataracts and elevated intraocular pressure (IOP). Loss of retinal ganglion cells (RGCs) in non-proliferative stages of diabetic retinopathy and small increases in IOP in diabetic patients has raised the possibility that diabetes affects the development and progression of ocular hypertension and glaucoma. The Ins2Akita mutation is known to cause diabetes and retinopathy on a C57BL/6J (B6) background by as early as 3 months of age. Here, the impact of the Akita mutation on glaucoma was assessed using DBA/2J (D2) mice, a widely used mouse model of ocular hypertension induced glaucoma. In D2.Ins2Akita/+ mice, the contribution of diabetes to vascular permeability, IOP elevation, RGC loss, and glaucoma development was assessed. D2.Ins2Akita/+ mice developed a severe diabetic nephropathy and early mortality between 6-8 months of age. This agrees with previous reports showing that the D2 background is more susceptible to diabetes than the B6 background. In addition, D2.Ins2Akita/+ mice had vascular leakage, astrocyte reactivity and a significant increase in IOP. However no RGC loss and no anterograde axonal transport dysfunction were found at 8.5 months of age. Therefore, our data show that despite severe diabetes and an increased IOP compared to controls, RGCs do not lose axon transport or degenerate. This may be due to a DBA/2J-specific genetic modifier(s) that could provide novel and important avenues for developing new therapies for diabetic retinopathy and possibly glaucoma.


Assuntos
Nefropatias Diabéticas/genética , Predisposição Genética para Doença , Glaucoma/genética , Insulina/genética , Hipertensão Ocular/genética , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Axônios/metabolismo , Axônios/patologia , Permeabilidade Capilar , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/mortalidade , Modelos Animais de Doenças , Expressão Gênica , Glaucoma/etiologia , Glaucoma/metabolismo , Glaucoma/mortalidade , Humanos , Insulina/metabolismo , Pressão Intraocular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Mutação , Hipertensão Ocular/complicações , Hipertensão Ocular/metabolismo , Hipertensão Ocular/mortalidade , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Especificidade da Espécie , Análise de Sobrevida
14.
Neurobiol Dis ; 71: 44-52, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25132557

RESUMO

The endothelin system is implicated in various human and animal glaucomas. Targeting the endothelin system has great promise as a treatment for human glaucoma, but the cell types involved and the exact mechanisms of action are not clearly elucidated. Here, we report a detailed characterization of the endothelin system in specific cell types of the optic nerve head (ONH) during glaucoma in DBA/2J mice. First, we show that key components of the endothelin system are expressed in multiple cell types. We discover that endothelin 2 (EDN2) is expressed in astrocytes as well as microglia/monocytes in the ONH. The endothelin receptor type A (Ednra) is expressed in vascular endothelial cells, while the endothelin receptor type B (Ednrb) receptor is expressed in ONH astrocytes. Second, we show that Macitentan treatment protects from glaucoma. Macitentan is a novel, orally administered, dual endothelin receptor antagonist with greater affinity, efficacy and safety than previous antagonists. Finally, we test the combinatorial effect of targeting both the endothelin and complement systems as a treatment for glaucoma. Similar to endothelin, the complement system is implicated in a variety of human and animal glaucomas, and has great promise as a treatment target. We discovered that combined targeting of the endothelin (Bosentan) and complement (C1qa mutation) systems is profoundly protective. Remarkably, 80% of DBA/2J eyes subjected to this combined inhibition developed no detectable glaucoma. This opens an exciting new avenue for neuroprotection in glaucoma.


Assuntos
Complemento C1q/metabolismo , Endotelina-2/metabolismo , Glaucoma/complicações , Degeneração Neural/etiologia , Degeneração Neural/terapia , Receptor de Endotelina A/metabolismo , Animais , Astrócitos/metabolismo , Bosentana , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Antagonistas do Receptor de Endotelina A/uso terapêutico , Glaucoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos DBA , Degeneração Neural/patologia , Pirimidinas/uso terapêutico , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Sulfonamidas/metabolismo , Sulfonamidas/uso terapêutico
15.
Artigo em Inglês | MEDLINE | ID: mdl-24993677

RESUMO

Glaucoma is a multifactorial neurodegenerative disorder affecting 80 million people worldwide. Loss of retinal ganglion cells and degeneration of their axons in the optic nerve are the major pathological hallmarks. Neuroinflammatory processes, inflammatory processes in the central nervous system, have been identified in human glaucoma and in experimental models of the disease. Furthermore, neuroinflammatory responses occur at early stages of experimental glaucoma, and inhibition of certain proinflammatory pathways appears neuroprotective. Here, we summarize the current understanding of neuroinflammation in the central nervous system, with emphasis on events at the optic nerve head during early stages of glaucoma.


Assuntos
Glaucoma/etiologia , Nervo Óptico/imunologia , Neurite Óptica/complicações , Células Ganglionares da Retina/imunologia , Astrócitos/fisiologia , Movimento Celular/fisiologia , Ativação do Complemento/fisiologia , Glaucoma/imunologia , Glaucoma/patologia , Humanos , Tolerância Imunológica/fisiologia , Leucócitos/fisiologia , Microglia/fisiologia , Neurite Óptica/imunologia , Neurite Óptica/fisiopatologia , Transdução de Sinais/fisiologia
16.
PLoS Genet ; 10(3): e1004089, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603532

RESUMO

Anterior chamber depth (ACD) is a key anatomical risk factor for primary angle closure glaucoma (PACG). We conducted a genome-wide association study (GWAS) on ACD to discover novel genes for PACG on a total of 5,308 population-based individuals of Asian descent. Genome-wide significant association was observed at a sequence variant within ABCC5 (rs1401999; per-allele effect size =  -0.045 mm, P = 8.17 × 10(-9)). This locus was associated with an increase in risk of PACG in a separate case-control study of 4,276 PACG cases and 18,801 controls (per-allele OR = 1.13 [95% CI: 1.06-1.22], P = 0.00046). The association was strengthened when a sub-group of controls with open angles were included in the analysis (per-allele OR = 1.30, P = 7.45 × 10(-9); 3,458 cases vs. 3,831 controls). Our findings suggest that the increase in PACG risk could in part be mediated by genetic sequence variants influencing anterior chamber dimensions.


Assuntos
Câmara Anterior/patologia , Estudo de Associação Genômica Ampla , Glaucoma de Ângulo Fechado/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Câmara Anterior/metabolismo , Povo Asiático , Glaucoma de Ângulo Fechado/patologia , Humanos , Polimorfismo de Nucleotídeo Único , Fatores de Risco
17.
BMC Genomics ; 14: 831, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24274089

RESUMO

BACKGROUND: Alzheimer's disease affects more than 35 million people worldwide but there is no known cure. Age is the strongest risk factor for Alzheimer's disease but it is not clear how age-related changes impact the disease. Here, we used a mouse model of Alzheimer's disease to identify age-specific changes that occur prior to and at the onset of traditional Alzheimer-related phenotypes including amyloid plaque formation. To identify these early events we used transcriptional profiling of mouse brains combined with computational approaches including singular value decomposition and hierarchical clustering. RESULTS: Our study identifies three key events in early stages of Alzheimer's disease. First, the most important drivers of Alzheimer's disease onset in these mice are age-specific changes. These include perturbations of the ribosome and oxidative phosphorylation pathways. Second, the earliest detectable disease-specific changes occur to genes commonly associated with the hypothalamic-adrenal-pituitary (HPA) axis. These include the down-regulation of genes relating to metabolism, depression and appetite. Finally, insulin signaling, in particular the down-regulation of the insulin receptor substrate 4 (Irs4) gene, may be an important event in the transition from age-related changes to Alzheimer's disease specific-changes. CONCLUSION: A combination of transcriptional profiling combined with computational analyses has uncovered novel features relevant to Alzheimer's disease in a widely used mouse model and offers avenues for further exploration into early stages of AD.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Transcriptoma , Doença de Alzheimer/genética , Animais , Córtex Cerebral/metabolismo , Análise por Conglomerados , Modelos Animais de Doenças , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Insulina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ocitocina/metabolismo , Fenótipo , Sistema Hipófise-Suprarrenal/metabolismo , Transdução de Sinais , Transcrição Gênica
18.
J Neuroinflammation ; 10: 76, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23806181

RESUMO

BACKGROUND: Glaucoma is an age-related neurodegenerative disorder involving the loss of retinal ganglion cells (RGCs), which results in blindness. Studies in animal models have shown that activation of inflammatory processes occurs early in the disease. In particular, the complement cascade is activated very early in DBA/2J mice, a widely used mouse model of glaucoma. A comprehensive analysis of the role of the complement cascade in DBA/2J glaucoma has not been possible because DBA/2J mice are naturally deficient in complement component 5 (C5, also known as hemolytic complement, Hc), a key mediator of the downstream processes of the complement cascade, including the formation of the membrane attack complex. METHODS: To assess the role of C5 in DBA/2J glaucoma, we backcrossed a functional C5 gene from strain C57BL/6J to strain DBA/2J for at least 10 generations. The prevalence and severity of glaucoma was evaluated using ocular examinations, IOP measurements, and assessments of optic nerve damage and RGC degeneration. To understand how C5 affects glaucoma, C5 expression was assessed in the retinas and optic nerves of C5-sufficient DBA/2J mice, using immunofluorescence. RESULTS: C5-sufficient DBA/2J mice developed a more severe glaucoma at an earlier age than standard DBA/2J mice, which are therefore protected by C5 deficiency. Components of the membrane attack complex were found to be deposited at sites of axonal injury in the optic nerve head and associated with RGC soma in the retina. CONCLUSION: C5 plays an important role in glaucoma, with its deficiency lessening disease severity. These results highlight the importance of fully understanding the role of the complement cascade in neurodegenerative diseases. Inhibiting C5 may be beneficial as a therapy for human glaucoma.


Assuntos
Complemento C5/deficiência , Glaucoma/metabolismo , Glaucoma/prevenção & controle , Animais , Glaucoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Índice de Gravidade de Doença
19.
Exp Neurol ; 246: 54-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22285251

RESUMO

Glaucoma is a neurodegenerative disease affecting 70million people worldwide. For some time, analysis of human glaucoma and animal models suggested that RGC axonal injury in the optic nerve head (where RGC axons exit the eye) is an important early event in glaucomatous neurodegeneration. During the last decade advances in molecular biology and genome manipulation have allowed this hypothesis to be tested more critically, at least in animal models. Data indicate that RGC axon degeneration precedes soma death. Preventing soma death using mouse models that are mutant for BAX, a proapoptotic gene, is not sufficient to prevent the degeneration of RGC axons. This indicates that different degeneration processes occur in different compartments of the RGC during glaucoma. Furthermore, the Wallerian degeneration slow allele (Wld(s)) slows or prevents RGC axon degeneration in rodent models of glaucoma. These experiments and many others, now strongly support the hypothesis that axon degeneration is a critical pathological event in glaucomatous neurodegeneration. However, the events that lead from a glaucomatous insult (e.g. elevated intraocular pressure) to axon damage in glaucoma are not well defined. For developing new therapies, it will be necessary to clearly define and order the molecular events that lead from glaucomatous insults to axon degeneration.


Assuntos
Axônios/metabolismo , Axônios/patologia , Glaucoma/metabolismo , Glaucoma/patologia , Transdução de Sinais/fisiologia , Animais , Humanos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Degeneração Walleriana/metabolismo , Degeneração Walleriana/patologia
20.
Neurobiol Dis ; 48(1): 124-31, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22750530

RESUMO

Amyotrophic lateral sclerosis (ALS) is characterised by substantial loss of both upper and lower motor neuron function, with sensory and cognitive systems less affected. Though heritable forms of the disease have been described, the vast majority of cases are sporadic with poorly defined underlying pathogenic mechanisms. Here we demonstrate that the neurological pathology induced in transgenic mice by overexpression of γ-synuclein, a protein not previously associated with ALS, recapitulates key features of the disease, namely selective damage and loss of discrete populations of upper and lower motor neurons and their axons, contrasted by limited effects upon the sensory system.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Axônios/patologia , Neurônios Motores/patologia , Medula Espinal/patologia , gama-Sinucleína/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Percepção do Tato/fisiologia , gama-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA