Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 42(10): 1961-1974, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35702047

RESUMO

We found that blood vitronectin (VTN) leaks into the brain and exacerbates tissue loss after stroke by increasing pro-inflammatory IL-6 expression in female, but not male, mice. VTN signals through integrins and downstream focal adhesion kinase (FAK). Here, a two day systemic treatment with a small molecule FAK inhibitor starting 6 h after middle cerebral artery occlusion reduced ipsilateral brain injury size by ∼40-45% at 7 and 14 d, as well as inflammation and motor dysfunction in wild-type female, but not male, mice. FAK inhibition also reduced IL-6 expression in the injured female striatum at 24 h by 62%. Inducible selective gene deletion of FAK in astrocytes also reduced acute IL-6 expression by 72% only in females, and mitigated infarct size by ∼80% and inflammation at 14 d after stroke. Lastly, VTN-/- females had better outcomes, but FAK inhibitor treatment had no additional protective or anti-inflammatory effects. Altogether, this suggests that VTN is detrimental in females primarily through FAK and that FAK inhibition provides neuroprotection (cerebroprotection) by reducing VTN-induced IL-6 expression in astrocytes. Thus, VTN signaling can be targeted to mitigate harmful inflammation with relevance to treatments for women with ischemic stroke, who often have worse outcomes than men.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Animais , Anti-Inflamatórios , Isquemia Encefálica/tratamento farmacológico , Feminino , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Integrinas/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Vitronectina/genética , Vitronectina/metabolismo
2.
Physiol Rep ; 10(9): e15301, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35531929

RESUMO

Vitronectin (VTN) is a glycoprotein enriched in the blood and activates integrin receptors. VTN blood levels increase only in female mice 24 h after an ischemic stroke and exacerbate brain injury through IL-6-driven inflammation, but the VTN induction mechanism is unknown. Here, a 30 min middle cerebral artery occlusion (MCAO) in female mice induced VTN protein in the liver (normally the main source) in concert with plasma VTN. Male mice were excluded as VTN is not induced after stroke. MCAO also increased plasma VTN levels after de novo expression of VTN in the liver of VTN-/- female mice, using a hepatocyte-specific (SERPINA1) promoter. MCAO did not affect SERPINA1 or VTN mRNA in the liver, brain, or several peripheral organs, or platelet VTN, compared to sham mice. Thus, hepatocytes are the source of stroke-induced increases in plasma VTN, which is independent of transcription. The cholinergic innervation by the parasympathetic vagus nerve is a potential source of brain-liver signaling after stroke. Right-sided vagotomy at the cervical level led to increased plasma VTN levels, suggesting that VTN release is inhibited by vagal tone. Co-culture of hepatocytes with cholinergic neurons or treatment with acetylcholine, but not noradrenaline (sympathetic transmitter), suppressed VTN expression. Hepatocytes have muscarinic receptors and the M1/M3 agonist bethanechol decreased VTN mRNA and protein release in vitro via M1 receptors. Finally, systemic bethanechol treatment blocked stroke-induced plasma VTN. Thus, VTN translation and release are inhibited by muscarinic signaling from the vagus nerve and presents a novel target for lessening detrimental VTN expression.


Assuntos
Acidente Vascular Cerebral , Vitronectina , Animais , Betanecol , Colinérgicos , Feminino , Infarto da Artéria Cerebral Média , Integrinas , Fígado/metabolismo , Camundongos , RNA Mensageiro , Acidente Vascular Cerebral/sangue , Nervo Vago/fisiologia , Vitronectina/sangue
3.
Stroke ; 51(5): 1587-1595, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32312218

RESUMO

Background and Purpose- Women have worse stroke outcomes than men, especially after menopause. Few studies have focused on female-specific mechanisms, other than hormones. We investigated the role of the blood protein VTN (vitronectin) after ischemic stroke in mice. Methods- Adult male and female VTN knockout and wild-type littermates and C57BL/6 mice received a middle cerebral artery occlusion and the injured brain tissue analyzed 24 hours to 3 weeks later for cell loss and inflammation, as well as neurological function. Blood VTN levels were measured before and after stroke. Results- Intravenously injected VTN leaked extensively from bloodstream into brain infarct and penumbra by 24 hours after stroke. Strikingly, VTN was detrimental in female, but not male, mice, as shown by reduced brain injury (26.2±2.6% versus 13.4±3.8%; P=0.018; n=6 and 5) and forelimb dysfunction in female VTN knockout mice. Stroke increased plasma VTN 2- to 8-fold at 24 hours in females (36±4 versus 145±24 µg/mL; P<0.0001; n=10 and 7), but not males (62±8 versus 68±6; P>0.99; n=10 and 7), and returned to control levels by 7 days. Individually variable VTN levels at 24 hours correlated with stroke-induced brain injury at 7 days only in females. VTN promoted stroke-induced microglia/macrophage activation and leukocyte infiltration in females. Proinflammatory IL (interleukin)-6 greatly increased in the striatum at 24 hours in wild-type mice but was increased ≈60% less in female (739±159 versus 268±111; P=0.02; n=7 and 6), but not male (889±178 versus 1179±295; P=0.73; n=10 and 11), knockout mice. In individual wild-type females, plasma VTN levels correlated with striatal IL-6 expression at 24 hours. The female-specific effect of VTN-induced IL-6 expression following stroke was not due to gonadal hormones, as shown by ovariectomy and castration. Lastly, intrastriatal injection of IL-6 in female mice immediately before stroke reversed the VTN knockout phenotypes of reduced brain injury and microglia/macrophage activation. Conclusions- VTN plays a novel sexually dimorphic detrimental pathophysiological role in females and might ultimately be a therapeutic target to improve stroke outcomes in women.


Assuntos
Barreira Hematoencefálica/metabolismo , Infarto da Artéria Cerebral Média/genética , Inflamação/genética , Interleucina-6/genética , Vitronectina/genética , Animais , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Feminino , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/metabolismo , Interleucina-6/metabolismo , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , RNA Mensageiro/metabolismo , Fatores Sexuais , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Vitronectina/sangue , Vitronectina/metabolismo
4.
Cell Commun Signal ; 18(1): 64, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32312272

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

5.
Exp Neurol ; 323: 113088, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31678139

RESUMO

Vitronectin (VTN) is a blood protein produced mainly by the liver. We show that VTN leaks from the bloodstream into the injury site and neighboring subventricular zone (SVZ) following ischemic stroke (middle cerebral artery occlusion, MCAO) in adult mice. MCAO is known to increase neurogenesis after stroke. VTN inhibits this response in females, but not in males, as shown by ~70% more stroke-induced SVZ neurogenesis in female VTN-/- mice at 14 d. In female VTN-/- mice, stroke-induced expression of interleukin-6 (IL-6) at 24 h was reduced in the SVZ. The closely related leukemia inhibitory factor (LIF) or pro-neurogenic ciliary neurotrophic factor (CNTF) were not affected. The female-specific effect of VTN on IL-6 expression was not due to sex hormones, as shown by ovariectomy and castration. IL-6 injection next to the SVZ reversed the MCAO-induced increase in neurogenesis seen in VTN-/- mice. Our in vitro and vivo data suggest that plasma VTN activates focal adhesion kinase (FAK) in the SVZ following MCAO, which reduces IL-6 expression in astrocytes but increases it in other cells such as microglia/macrophages. Inducible conditional astrocytic FAK deletion increased MCAO-induced IL-6 expression in females at 24 h and blocked MCAO-induced neurogenesis at 14 d, confirming a key detrimental role of IL-6. Collectively, these data suggest that leakage of VTN into the SVZ reduces the neurogenic response to stroke in female mice by promoting IL-6 expression. Reducing VTN or VTN signaling may be an approach to promote neurogenesis for neuroprotection and cell replacement after stroke in females.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Interleucina-6/metabolismo , Neurogênese/fisiologia , Acidente Vascular Cerebral/metabolismo , Vitronectina/metabolismo , Animais , Feminino , Ventrículos Laterais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Caracteres Sexuais , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/fisiopatologia
6.
Psychoneuroendocrinology ; 100: 96-105, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30299260

RESUMO

Ciliary neurotrophic factor (CNTF) is produced by astrocytes and promotes neurogenesis and neuroprotection. Little is known about the role of CNTF in affective behavior. We investigated whether CNTF affects depressive- and anxiety-like behavior in adult mice as tested in the forced swim, sucrose preference and elevated-T maze tests. Female wild type CNTF+/+ mice more readily developed behavioral despair with increased immobility time and decreased latency to immobility in the forced swim test than male CNTF+/+ littermates. The lack of CNTF in CNTF-/- mice had an opposite effect on depressive-like behavior in female mice (reduced immobility time and increased sucrose preference) vs. male mice (increased immobility time). Female wildtype mice expressed more CNTF in the amygdala than male mice. Ovariectomy increased CNTF expression, as well as immobility time, which was significantly reduced in CNTF-/- mice, suggesting that CNTF mediates overiectomy-induced immobility time, possibly in the amygdala. Progesterone but not 17-ß estradiol inhibited CNTF expression in cultured C6 astroglioma cells. Progesterone treatment also reduced CNTF expression in the amygdala and decreased immobility time in female CNTF+/+ but not in CNTF-/- mice. Castration did not alter CNTF expression in males nor their behavior. Lastly, there were no effects of CNTF on the elevated T-maze, a behavioral test of anxiety, suggesting that a different mechanism may underlie anxiety-like behavior. This study reveals a novel CNTF-mediated mechanism in stress-induced depressive-like behavior and points to opportunities for sex-specific treatments for depression, e.g. progesterone in females and CNTF-stimulating drugs in males.


Assuntos
Fator Neurotrófico Ciliar/fisiologia , Depressão/genética , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Comportamento Animal/fisiologia , Fator Neurotrófico Ciliar/genética , Depressão/patologia , Depressão/fisiopatologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/genética , Caracteres Sexuais , Células Tumorais Cultivadas
7.
Exp Neurol ; 312: 20-32, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30408465

RESUMO

Vitronectin (VTN) is a glycoprotein in the blood and affects hemostasis. VTN is also present in the extracellular matrix of various organs but little is known about its function in healthy adult tissues. We show, in adult mice, that VTN is uniquely expressed by approximately half of the pericytes of subventricular zone (SVZ) where neurogenesis continues throughout life. Intracerebral VTN antibody injection or VTN knockout reduced neurogenesis as well as expression of pro-neurogenic CNTF, and anti-neurogenic LIF and IL-6. Conversely, injections of VTN, or plasma from VTN+/+, but not VTN-/- mice, increased these cytokines. VTN promoted SVZ neurogenesis when LIF and IL-6 were suppressed by co-administration of a gp130 inhibitor. Unexpectedly, VTN inhibited FAK signaling and VTN-/- mice had increased FAK signaling in the SVZ. Further, an FAK inhibitor or VTN increased CNTF expression, but not in conditional astrocytic FAK knockout mice, suggesting that VTN increases CNTF through FAK inhibition in astrocytes. These results identify a novel role of pericyte-derived VTN in the brain, where it regulates SVZ neurogenesis through co-expression of CNTF, LIF and IL-6. VTN-integrin-FAK and gp130 signaling may provide novel targets to induce neurogenesis for cell replacement therapies.


Assuntos
Fator Neurotrófico Ciliar/biossíntese , Neurogênese/fisiologia , Pericitos/metabolismo , Prosencéfalo/metabolismo , Vitronectina/biossíntese , Animais , Anticorpos/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Vitronectina/antagonistas & inibidores
8.
Glia ; 66(11): 2456-2469, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30500112

RESUMO

Astrocyte-derived ciliary neurotrophic factor (CNTF) promotes adult subventricular zone (SVZ) neurogenesis. We found that focal adhesion kinase (FAK) and JNK, but not ERK or P38, repress CNTF in vitro. Here, we defined the FAK-JNK pathway and its regulation of CNTF in mice, and the related leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), which promote stem cell renewal at the expense of neurogenesis. Intrastriatal injection of FAK inhibitor, FAK14, in adult male C57BL/6 mice reduced pJNK and increased CNTF expression in the SVZ-containing periventricular region. Injection of a JNK inhibitor increased CNTF without affecting LIF and IL-6, and increased SVZ proliferation and neuroblast formation. The JNK inhibitor had no effect in CNTF-/- mice, suggesting that JNK inhibits SVZ neurogenesis by repressing CNTF. Inducible deletion of FAK in astrocytes increased SVZ CNTF and neurogenesis, but not LIF and IL-6. Intrastriatal injection of inhibitors suggested that P38 reduces LIF and IL-6 expression, whereas ERK induces CNTF and LIF. Intrastriatal FAK inhibition increased LIF, possibly through ERK, and IL-6 through another pathway that does not involve P38. Systemic injection of FAK14 also inhibited JNK while increasing CNTF, but did not affect P38 and ERK activation, or LIF and IL-6 expression. Importantly, systemic FAK14 increased SVZ neurogenesis in wild-type C57BL/6 and CNTF+/+ mice, but not in CNTF-/- littermates, indicating that it acts by upregulating CNTF. These data show a surprising differential regulation of related cytokines and identify the FAK-JNK-CNTF pathway as a specific target in astrocytes to promote neurogenesis and possibly neuroprotection in neurological disorders.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Ciliar/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Ventrículos Laterais/citologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neurogênese/fisiologia , Animais , Antracenos/farmacologia , Astrócitos/efeitos dos fármacos , Linhagem Celular Tumoral , Fator Neurotrófico Ciliar/genética , Citocinas/genética , Citocinas/metabolismo , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Antígeno Ki-67/metabolismo , Ventrículos Laterais/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fatores de Tempo
9.
J Cell Sci ; 131(3)2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29222114

RESUMO

We defined how blood-derived vitronectin (VTN) rapidly and potently activates leukemia inhibitory factor (LIF) and pro-inflammatory interleukin 6 (IL-6) in vitro and after vascular injury in the brain. Treatment with VTN (but not fibrinogen, fibronectin, laminin-111 or collagen-I) substantially increased LIF and IL-6 within 4 h in C6-astroglioma cells, while VTN-/- mouse plasma was less effective than that from wild-type mice. LIF and IL-6 were induced by intracerebral injection of recombinant human (rh)VTN in mice, but induction seen upon intracerebral hemorrhage was less in VTN-/- mice than in wild-type littermates. In vitro, VTN effects were inhibited by RGD, αvß3 and αvß5 integrin-blocking peptides and antibodies. VTN activated focal adhesion kinase (FAK; also known as PTK2), whereas pharmacological- or siRNA-mediated inhibition of FAK, but not PYK2, reduced the expression of LIF and IL-6 in C6 and endothelial cells and after traumatic cell injury. Dominant-negative FAK (Y397F) reduced the amount of injury-induced LIF and IL-6. Pharmacological inhibition or knockdown of uPAR (also known as PLAUR), which binds VTN, also reduced cytokine expression, possibly through a common target of uPAR and integrins. We propose that VTN leakage into tissues promotes inflammation. Integrin-FAK signaling is therefore a novel IL-6 and LIF regulation mechanism relevant to the inflammation and stem cell fields.


Assuntos
Encéfalo/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Interleucina-6/metabolismo , Fator Inibidor de Leucemia/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais , Vitronectina/sangue , Animais , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Humanos , Integrinas/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Regulação para Cima
11.
Cell Commun Signal ; 14(1): 32, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27978828

RESUMO

BACKGROUND: STAT3 is increasingly becoming known for its non-transcriptional regulation of mitochondrial bioenergetic function upon activation of its S727 residue (S727-STAT3). Lengthy mitochondrial dysfunction can lead to cell death. We tested whether an integrin-FAK-STAT3 signaling pathway we recently discovered regulates mitochondrial function and cell survival, and treatments thereof. METHODS: Cultured mouse brain bEnd5 endothelial cells were treated with integrin, FAK or STAT3 inhibitors, FAK siRNA, as well as integrin and STAT3 activators. STAT3 null cells were transfected with mutant STAT3 plasmids. Outcome measures included oxygen consumption rate for mitochondrial bioenergetics, Western blotting for protein phosphorylation, mitochondrial membrane potential for mitochondrial integrity, ROS production, and cell counts. RESULTS: Vitronectin-dependent mitochondrial basal respiration, ATP production, and maximum reserve and respiratory capacities were suppressed within 4 h by RGD and αvß3 integrin antagonist peptides. Conversely, integrin ligands vitronectin, laminin and fibronectin stimulated mitochondrial function. Pharmacological inhibition of FAK completely abolished mitochondrial function within 4 h while FAK siRNA treatments confirmed the specificity of FAK signaling. WT, but not S727A functionally dead mutant STAT3, rescued bioenergetics in cells made null for STAT3 using CRISPR-Cas9. STAT3 inhibition with stattic in whole cells rapidly reduced mitochondrial function and mitochondrial pS727-STAT3. Stattic treatment of isolated mitochondria did not reduce pS727 whereas more was detected upon phosphatase inhibition. This suggests that S727-STAT3 is activated in the cytoplasm and is short-lived upon translocation to the mitochondria. FAK inhibition reduced pS727-STAT3 within mitochondria and reduced mitochondrial function in a non-transcriptional manner, as shown by co-treatment with actinomycin. Treatment with the small molecule bryostatin-1 or hepatocyte growth factor (HGF), which indirectly activate S727-STAT3, preserved mitochondrial function during FAK inhibition, but failed in the presence of the STAT3 inhibitor. FAK inhibition induced loss of mitochondrial membrane potential, which was counteracted by bryostatin, and increased superoxide and hydrogen peroxide production. Bryostatin and HGF reduced the substantial cell death caused by FAK inhibition over a 24 h period. CONCLUSION: These data suggest that extracellular matrix molecules promote STAT3-dependent mitochondrial function and cell survival through integrin-FAK signaling. We furthermore show a new treatment strategy for cell survival using S727-STAT3 activators.


Assuntos
Células Endoteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrinas/metabolismo , Mitocôndrias/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Trifosfato de Adenosina/metabolismo , Animais , Morte Celular , Linhagem Celular Tumoral , Células Endoteliais/citologia , Metabolismo Energético , Camundongos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo
12.
J Mol Neurosci ; 59(3): 404-10, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27074745

RESUMO

We explored the response of a panel of selected microRNAs (miRNAs) in neuroprotection produced by ischemic preconditioning. Hippocampal neuronal cultures were exposed to a 30-min oxygen-glucose deprivation (OGD). In our hands, this duration of OGD does not result in neuronal loss in vitro but significantly reduces neuronal death from a subsequent 'lethal' OGD insult. RT-qPCR was used to determine the expression of 16 miRNAs of interest at 1 and 24-h post-OGD. One miRNA (miR-98) was significantly decreased at 1-h post-OGD. Ten miRNAs (miR-9, miR-21, miR-29b, miR-30e, miR-101a, miR-101b, miR-124a, miR-132, miR-153, miR-204) were increased significantly at 24-h post-OGD. No miRNAs were decreased at 24-h. The increases observed in the 24-h group suggested that these miRNAs might play a role in preconditioning-induced neuroprotection. We selected the widely studied miR-132, a brain enriched, CREB regulated miRNA, to explore its role in simulated ischemic insults. We found that hippocampal neurons transduced with lentiviral vectors expressing miR-132 were protected from OGD and NMDA treatment, but not hydrogen peroxide. These findings add to the growing literature that targeting neuroprotective pathways controlled by miRNAs may represent a therapeutic strategy for the treatment of ischemic brain injury.


Assuntos
Glucose/deficiência , MicroRNAs/genética , Neurônios/metabolismo , Oxigênio/metabolismo , Regulação para Cima , Animais , Hipóxia Celular , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Glucose/metabolismo , Hipocampo/irrigação sanguínea , Hipocampo/citologia , Peróxido de Hidrogênio/toxicidade , Precondicionamento Isquêmico , MicroRNAs/metabolismo , N-Metilaspartato/toxicidade , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar
13.
Int Rev Neurobiol ; 110: 349-71, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24209445

RESUMO

Patients with primary familial brain calcifications (PFBC) present bilateral calcifications, often affecting basal ganglia, thalamus, and cerebellum, inherited in an autosomal dominant pattern of segregation. Affected individuals display a wide variety of motor and cognitive impairments such as parkinsonism, dystonia, migraine, dementia, psychosis, and mood symptoms. Worldwide growth in the availability of neuroimaging procedures, combined with careful screening of patients and their relatives, has increased detection of PFBC. Recently, mutations in the SLC20A2 gene coding for the inorganic phosphate transporter PiT2 were linked to PFBC, thereby implicating impaired phosphate transport as an underlying disease mechanism. To date, around 20 families of various ethnicities carry different mutations in SLC20A2 correlate with ~40% of PFBC cases. More recently, two French families were recently reported with mutations in PDGFRB: c.1973T>C, p.L658P and c.2959C>T, p.R987W, a class III tyrosine kinase receptor. Six other families were found with mutations in PDGFB, and, in general, mutations at the PDGF pathway add a new dimension to the physiopathology of PFBC so far explained by a disturbance in phosphate homeostasis with SLC20A2. The identification of SLC20A2, PDGFRB, and PDGFB provides a new avenue for potential treatments based on compounds such as bisphosphonates and those modulating the PDGFB pathway.


Assuntos
Encefalopatias/genética , Encefalopatias/patologia , Calcinose/genética , Calcinose/patologia , Humanos
14.
Cell Commun Signal ; 11: 35, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23693126

RESUMO

BACKGROUND: Ciliary neurotrophic factor (CNTF) expression is repressed in astrocytes by neuronal contact in the CNS and is rapidly induced by injury. Here, we defined an inhibitory integrin signaling pathway. RESULTS: The integrin substrates laminin, fibronectin and vitronectin, but not collagen, thrombospondin or fibrinogen, reduced CNTF expression in C6 astroglioma cells. Antibodies against αv and ß5, but not α6 or ß1, integrin induced CNTF. Together, the ligand and antibody specificity suggests that CNTF is repressed by αvß5 integrin. Antibodies against Thy1, an abundant neuronal surface protein whose function is unclear, induced CNTF in neuron-astrocyte co-cultures indicating that it is a neuroglial CNTF repressor. Inhibition of the integrin signaling molecule Focal Adhesion Kinase (FAK) or the downstream c-Jun N-terminal kinase (JNK), but not extracellular regulated kinase (ERK) or p38 MAPK, greatly induced CNTF mRNA and protein expression within 4 hours. This selective inhibitory pathway phosphorylated STAT3 on its inhibitory ser-727 residue interfering with activity of the pro-transcription Tyr-705 residue. STAT3 can activate CNTF transcription because it bound to its promoter and FAK antagonist-induced CNTF was reduced by blocking STAT3. Microinjection of FAK inhibitor directly into the brain or spinal cord in adult mice rapidly induced CNTF mRNA and protein expression. Importantly, systemic treatment with FAK inhibitors over 3 days induced CNTF in the subventricular zone and increased neurogenesis. CONCLUSIONS: Neuron-astroglia contact mediated by integrins serves as a sensor to enable rapid neurotrophic responses and provides a new pharmacological avenue to exploit the neuroprotective properties of endogenous CNTF.


Assuntos
Fator Neurotrófico Ciliar/metabolismo , Neuroglia/metabolismo , Receptores de Vitronectina/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Fator Neurotrófico Ciliar/genética , Técnicas de Cocultura , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Camundongos Endogâmicos C57BL , Neurogênese , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Quinolonas/farmacologia , Ratos , Serina/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Antígenos Thy-1/metabolismo , Tirosina/metabolismo
15.
Neurobiol Dis ; 49: 68-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22960105

RESUMO

Focal brain ischemia in adult rats rapidly and robustly induces neurogenesis in the subventricular zone (SVZ) but there are few and inconsistent reports in mice, presenting a hurdle to genetically investigate the endogenous neurogenic regulators such as ciliary neurotrophic factor (CNTF). Here, we first provide a platform for further studies by showing that middle cerebral artery occlusion in adult male C57BL/6 mice robustly enhances neurogenesis in the SVZ only under very specific conditions, i.e., 14days after a 30min occlusion. CNTF expression paralleled changes in the number of proliferated, BrdU-positive, SVZ cells. Stroke-induced proliferation was absent in CNTF-/- mice, suggesting that it is mediated by CNTF. MCAO-increased CNTF appears to act on C cell proliferation and by inducing FGF2 expression but not via EGF expression or Notch1 signaling of neural stem cells in the SVZ. CNTF is unique, as expression of other gp130 ligands, IL-6 and LIF, did not predict SVZ proliferation or showed no or only small compensatory increases in CNTF-/- mice. Expression of tumor necrosis factor-α, which can inhibit neurogenesis, and the presence of leukocytes in the SVZ were inversely correlated with neurogenesis, but pro-inflammatory cytokines did not affect CNTF expression in cultured astrocytes. These results suggest that slowly up-regulated CNTF in the SVZ mediates stroke-induced neurogenesis and is counteracted by inflammation. Further pharmacological stimulation of endogenous CNTF might be a good therapeutic strategy for cell replacement after stroke as CNTF regulates normal patterns of neurogenesis and is expressed almost exclusively in the nervous system.


Assuntos
Encéfalo/fisiopatologia , Fator Neurotrófico Ciliar/metabolismo , Neurogênese/fisiologia , Nicho de Células-Tronco/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Astrócitos/fisiologia , Células Cultivadas , Fator Neurotrófico Ciliar/genética , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média , Leucócitos/fisiologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/patologia , Células-Tronco Neurais/fisiologia , Neuroimunomodulação/fisiologia , Acidente Vascular Cerebral/patologia
16.
J Neurosci ; 32(27): 9277-87, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22764235

RESUMO

Ciliary neurotrophic factor (CNTF) is a potent neural cytokine with very low expression in the CNS, predominantly by astrocytes. CNTF increases rapidly and greatly following traumatic or ischemic injury. Understanding the underlying mechanisms would help to design pharmacological treatments to increase endogenous CNTF levels for neuroprotection. Here, we show that astroglial CNTF expression in the adult mouse striatum is increased twofold within 1 h and increases up to >30-fold over 2 weeks following a focal stroke caused by a transient middle cerebral artery occlusion (MCAO). Selective neuronal loss caused by intrastriatal injection of quinolinic acid resulted in a comparable increase. Cocultured neurons reduced CNTF expression in astrocytes, which was prevented by light trypsinization. RGD (arginine-glycine-aspartic acid) blocking peptides induced CNTF expression, which was dependent on transcription. Astroglial CNTF expression was not affected by diffusible neuronal molecules or by neurotransmitters. The transient ischemia does not seem to directly increase CNTF, as intrastriatal injection of an ischemic solution or exposure of naive mice or cultured cells to severe hypoxia had minimal effects. Inflammatory mechanisms were probably also not involved, as intrastriatal injection of proinflammatory cytokines (IFNγ, IL6) in naive mice had no or small effects, and anti-inflammatory treatments did not diminish the increase in CNTF after MCAO. CNTF-/- mice had more extensive tissue loss and similar astrocyte activation after MCAO than their wild-type littermates. These data suggest that contact-mediated integrin signaling between neurons and astrocytes normally represses CNTF expression and that neuronal dysfunction causes a rapid protective response by the CNS.


Assuntos
Astrócitos/patologia , Comunicação Celular/genética , Fator Neurotrófico Ciliar/genética , Hipóxia Encefálica/patologia , Infarto da Artéria Cerebral Média/patologia , Degeneração Neural/patologia , Neurônios/patologia , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Linhagem Celular Tumoral , Fator Neurotrófico Ciliar/biossíntese , Técnicas de Cocultura , Hipóxia Encefálica/genética , Hipóxia Encefálica/fisiopatologia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Neurônios/metabolismo , Neurônios/fisiologia , Cultura Primária de Células
17.
Methods Mol Biol ; 549: 217-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378206

RESUMO

There is incontrovertible evidence that neural progenitor cells (NPC) are found in the adult brain. The ability to identify and track NPC in the adult brain is of considerable importance if the properties of these cells are to be harnessed as potential therapies for degenerative brain disorders. The most commonly used approach of identifying these NPC in experimental studies, bromodeoxyuridine (BrdU) labelling, is outlined in this chapter. Immunohistochemical protocols for detecting endogenous and exogenous (introduced via transplantation) NPC in fresh-frozen and paraffin wax embedded brain tissue are described. Advice on how to label these NPC is also offered and multi-label fluorescence immunochemical staining approaches to determine the differentiation fate of NPC are described.


Assuntos
Encéfalo/citologia , Imuno-Histoquímica/métodos , Neurônios , Células-Tronco , Adulto , Animais , Anticorpos/metabolismo , Antimetabólitos/metabolismo , Bromodesoxiuridina/metabolismo , Humanos , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Fenótipo , Ratos , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/fisiologia , Inclusão do Tecido/métodos , Preservação de Tecido/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...