Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 30(2): 443-454, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38321220

RESUMO

Compromised vascular endothelial barrier function is a salient feature of diabetic complications such as sight-threatening diabetic macular edema (DME). Current standards of care for DME manage aspects of the disease, but require frequent intravitreal administration and are poorly effective in large subsets of patients. Here we provide evidence that an elevated burden of senescent cells in the retina triggers cardinal features of DME pathology and conduct an initial test of senolytic therapy in patients with DME. In cell culture models, sustained hyperglycemia provoked cellular senescence in subsets of vascular endothelial cells displaying perturbed transendothelial junctions associated with poor barrier function and leading to micro-inflammation. Pharmacological elimination of senescent cells in a mouse model of DME reduces diabetes-induced retinal vascular leakage and preserves retinal function. We then conducted a phase 1 single ascending dose safety study of UBX1325 (foselutoclax), a senolytic small-molecule inhibitor of BCL-xL, in patients with advanced DME for whom anti-vascular endothelial growth factor therapy was no longer considered beneficial. The primary objective of assessment of safety and tolerability of UBX1325 was achieved. Collectively, our data suggest that therapeutic targeting of senescent cells in the diabetic retina with a BCL-xL inhibitor may provide a long-lasting, disease-modifying intervention for DME. This hypothesis will need to be verified in larger clinical trials. ClinicalTrials.gov identifier: NCT04537884 .


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Edema Macular , Animais , Camundongos , Humanos , Edema Macular/tratamento farmacológico , Edema Macular/etiologia , Retinopatia Diabética/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Células Endoteliais , Senoterapia , Senescência Celular
2.
J Neuroinflammation ; 20(1): 145, 2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37344842

RESUMO

Cellular adaptation to low oxygen tension triggers primitive pathways that ensure proper cell function. Conditions of hypoxia and low glucose are characteristic of injured tissues and hence successive waves of inflammatory cells must be suited to function under low oxygen tension and metabolic stress. While Hypoxia-Inducible Factor (HIF)-1α has been shown to be essential for the inflammatory response of myeloid cells by regulating the metabolic switch to glycolysis, less is known about how HIF1α is triggered in inflammation. Here, we demonstrate that cells of the innate immune system require activity of the inositol-requiring enzyme 1α (IRE1α/XBP1) axis in order to initiate HIF1α-dependent production of cytokines such as IL1ß, IL6 and VEGF-A. Knockout of either HIF1α or IRE1α in myeloid cells ameliorates vascular phenotypes in a model of retinal pathological angiogenesis driven by sterile inflammation. Thus, pathways associated with ER stress, in partnership with HIF1α, may co-regulate immune adaptation to low oxygen.


Assuntos
Endorribonucleases , Proteínas Serina-Treonina Quinases , Humanos , Proteínas Serina-Treonina Quinases/genética , Hipóxia , Oxigênio/metabolismo , Células Mieloides/metabolismo , Inflamação/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia
3.
Proc Natl Acad Sci U S A ; 120(1): e2209973120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36574648

RESUMO

Obesity is a major risk factor for cancer. Conventional thought suggests that elevated adiposity predisposes to heightened inflammatory stress and potentiates tumor growth, yet underlying mechanisms remain ill-defined. Here, we show that tumors from patients with a body mass index >35 carry a high burden of senescent cells. In mouse syngeneic tumor models, we correlated a pronounced accretion of senescent cancer cells with poorly immunogenic tumors when mice were subjected to diet-induced obesity (DIO). Highly immunogenic tumors showed lesser senescence burden suggesting immune-mediated elimination of senescent cancer cells, likely targeted as a consequence of their senescence-associated secretory phenotype. Treatment with the senolytic BH3 mimetic small molecule inhibitor ABT-263 selectively stalled tumor growth in mice with DIO to rates comparable to regular diet-fed mice. Thus, consideration of body adiposity in the selection of cancer therapy may be a critical determinant for disease outcome in poorly immunogenic malignancies.


Assuntos
Senescência Celular , Neoplasias , Camundongos , Animais , Obesidade/complicações
4.
Sci Rep ; 11(1): 15767, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34344941

RESUMO

The beneficial effects of brown adipose tissue (BAT) on obesity and associated metabolic diseases are mediated through its capacity to dissipate energy as heat. While immune cells, such as tissue-resident macrophages, are known to influence adipose tissue homeostasis, relatively little is known about their contribution to BAT function. Here we report that neuropilin-1 (NRP1), a multiligand single-pass transmembrane receptor, is highly expressed in BAT-resident macrophages. During diet-induced obesity (DIO), myeloid-resident NRP1 influences interscapular BAT mass, and consequently vascular morphology, innervation density and ultimately core body temperature during cold exposure. Thus, NRP1-expressing myeloid cells contribute to the BAT homeostasis and potentially its thermogenic function in DIO.


Assuntos
Tecido Adiposo Marrom/fisiologia , Homeostase , Células Mieloides/metabolismo , Neuropilina-1/fisiologia , Obesidade/prevenção & controle , Termogênese , Animais , Dieta/efeitos adversos , Metabolismo Energético , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia
5.
EMBO Mol Med ; 13(5): e11754, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33876574

RESUMO

Age-related macular degeneration (AMD) in its various forms is a leading cause of blindness in industrialized countries. Here, we provide evidence that ligands for neuropilin-1 (NRP1), such as Semaphorin 3A and VEGF-A, are elevated in the vitreous of patients with AMD at times of active choroidal neovascularization (CNV). We further demonstrate that NRP1-expressing myeloid cells promote and maintain CNV. Expression of NRP1 on cells of myeloid lineage is critical for mitigating production of inflammatory factors such as IL6 and IL1ß. Therapeutically trapping ligands of NRP1 with an NRP1-derived trap reduces CNV. Collectively, our findings identify a role for NRP1-expressing myeloid cells in promoting pathological angiogenesis during CNV and introduce a therapeutic approach to counter neovascular AMD.


Assuntos
Neovascularização de Coroide , Degeneração Macular Exsudativa , Inibidores da Angiogênese , Humanos , Inflamação , Neuropilina-1/genética , Fator A de Crescimento do Endotélio Vascular , Acuidade Visual
6.
Cell Metab ; 33(4): 818-832.e7, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33548171

RESUMO

Attenuating pathological angiogenesis in diseases characterized by neovascularization such as diabetic retinopathy has transformed standards of care. Yet little is known about the molecular signatures discriminating physiological blood vessels from their diseased counterparts, leading to off-target effects of therapy. We demonstrate that in contrast to healthy blood vessels, pathological vessels engage pathways of cellular senescence. Senescent (p16INK4A-expressing) cells accumulate in retinas of patients with diabetic retinopathy and during peak destructive neovascularization in a mouse model of retinopathy. Using either genetic approaches that clear p16INK4A-expressing cells or small molecule inhibitors of the anti-apoptotic protein BCL-xL, we show that senolysis suppresses pathological angiogenesis. Single-cell analysis revealed that subsets of endothelial cells with senescence signatures and expressing Col1a1 are no longer detected in BCL-xL-inhibitor-treated retinas, yielding a retina conducive to physiological vascular repair. These findings provide mechanistic evidence supporting the development of BCL-xL inhibitors as potential treatments for neovascular retinal disease.


Assuntos
Senescência Celular , Doenças Retinianas/patologia , Proteína bcl-X/metabolismo , Animais , Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Cadeia alfa 1 do Colágeno Tipo I/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Flavonóis/química , Flavonóis/farmacologia , Flavonóis/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/metabolismo , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Proteína bcl-X/antagonistas & inibidores
7.
Aging (Albany NY) ; 12(24): 24836-24852, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33361521

RESUMO

MicroRNAs are small non-coding RNAs that post-transcriptionally regulate gene expression. We recently demonstrated that levels of miR-106b were significantly decreased in the vitreous and plasma of patients with neovascular age-related macular degeneration (AMD). Here we show that expression of the miR-106b-25 cluster is negatively regulated by the unfolded protein response pathway of protein kinase RNA-like ER kinase (PERK) in a mouse model of neovascular AMD. A reduction in levels of miR-106b triggers vascular growth both in vivo and in vitro by inducing production of pro-angiogenic factors. We demonstrate that therapeutic delivery of miR-106b to the retina with lentiviral vectors protects against aberrant retinal angiogenesis in two distinct mouse models of pathological retinal neovascularization. Results from this study suggest that miRNAs such as miR-106b have the potential to be used as multitarget therapeutics for conditions characterized by pathological retinal angiogenesis.


Assuntos
Neovascularização de Coroide/genética , Degeneração Macular/genética , MicroRNAs/genética , Neovascularização Retiniana/genética , Animais , Linhagem Celular , Movimento Celular/genética , Neovascularização de Coroide/patologia , Retinopatia Diabética , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/genética , Células Endoteliais , Queimaduras Oculares , Humanos , Terapia a Laser , Degeneração Macular/patologia , Camundongos , Oxigênio/toxicidade , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade , Resposta a Proteínas não Dobradas/genética , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
8.
EMBO Mol Med ; 8(12): 1366-1379, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27861126

RESUMO

Age-related macular degeneration in its neovascular form (NV AMD) is the leading cause of vision loss among adults above the age of 60. Epidemiological data suggest that in men, overall abdominal obesity is the second most important environmental risk factor after smoking for progression to late-stage NV AMD To date, the mechanisms that underscore this observation remain ill-defined. Given the impact of high-fat diets on gut microbiota, we investigated whether commensal microbes influence the evolution of AMD Using mouse models of NV AMD, microbiotal transplants, and other paradigms that modify the gut microbiome, we uncoupled weight gain from confounding factors and demonstrate that high-fat diets exacerbate choroidal neovascularization (CNV) by altering gut microbiota. Gut dysbiosis leads to heightened intestinal permeability and chronic low-grade inflammation characteristic of inflammaging with elevated production of IL-6, IL-1ß, TNF-α, and VEGF-A that ultimately aggravate pathological angiogenesis.


Assuntos
Neovascularização de Coroide/patologia , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Degeneração Macular/patologia , Neovascularização Patológica , Obesidade/complicações , Animais , Citocinas/sangue , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Disbiose/etiologia , Transplante de Microbiota Fecal , Inflamação/patologia , Degeneração Macular/epidemiologia , Camundongos , Obesidade/patologia
9.
PLoS One ; 9(4): e94175, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24714389

RESUMO

The transcription factor p53 mediates the apoptosis of post-mitotic neurons exposed to a wide range of stress stimuli. The apoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1, ASPP2 and iASPP. We previously showed that the pro-apoptotic members ASPP1 and ASPP2 contribute to p53-dependent death of retinal ganglion cells (RGCs). However, the role of the p53 inhibitor iASPP in the central nervous system (CNS) remains to be elucidated. To address this, we asked whether iASPP contributes to the survival of RGCs in an in vivo model of acute optic nerve damage. We demonstrate that iASPP is expressed by injured RGCs and that iASPP phosphorylation at serine residues, which increase iASPP affinity towards p53, is significantly reduced following axotomy. We show that short interference RNA (siRNA)-induced iASPP knockdown exacerbates RGC death, whereas adeno-associated virus (AAV)-mediated iASPP expression promotes RGC survival. Importantly, our data also demonstrate that increasing iASPP expression in RGCs downregulates p53 activity and blocks the expression of pro-apoptotic targets PUMA and Fas/CD95. This study demonstrates a novel role for iASPP in the survival of RGCs, and provides further evidence of the importance of the ASPP family in the regulation of neuronal loss after axonal injury.


Assuntos
Axônios/metabolismo , Sobrevivência Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Nervo Óptico/metabolismo , Proteínas Repressoras/metabolismo , Animais , Axotomia , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurônios/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética
10.
J Neurosci ; 33(5): 2205-16, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23365256

RESUMO

The transcription factor p53 mediates neuronal death in a variety of stress-related and neurodegenerative conditions. The proapoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1 and ASPP2. However, whether ASPP1/2 play a role in the regulation of p53-dependent neuronal death in the CNS is currently unknown. To address this, we asked whether ASPP1/2 contribute to the death of retinal ganglion cells (RGCs) using in vivo models of acute optic nerve damage in mice and rats. Here, we show that p53 is activated in RGCs soon after injury and that axotomy-induced RGC death is attenuated in p53 heterozygote and null mice. We demonstrate that ASPP1/2 proteins are abundantly expressed by injured RGCs, and that short interfering (si)RNA-based ASPP1 or ASPP2 knockdown promotes robust RGC survival. Comparative gene expression analysis revealed that siASPP-mediated downregulation of p53-upregulated-modulator-of-apoptosis (PUMA), Fas/CD95, and Noxa depends on p53 transcriptional activity. Furthermore, siRNA against PUMA or Fas/CD95 confers neuroprotection, demonstrating a functional role for these p53 targets in RGC death. Our study demonstrates a novel role for ASPP1 and ASPP2 in the death of RGCs and provides evidence that blockade of the ASPP-p53 pathway is beneficial for central neuron survival after axonal injury.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Morte Celular/fisiologia , Células Ganglionares da Retina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Receptor fas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Axônios/metabolismo , Regulação para Baixo , Feminino , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Receptor fas/genética
11.
Prog Retin Eye Res ; 31(2): 152-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155051

RESUMO

Glaucoma is a group of diseases characterized by progressive optic nerve degeneration that results in visual field loss and irreversible blindness. A crucial element in the pathophysiology of all forms of glaucoma is the death of retinal ganglion cells (RGCs), a population of CNS neurons with their soma in the inner retina and axons in the optic nerve. Strategies that delay or halt RGC loss have been recognized as potentially beneficial to preserve vision in glaucoma; however, the success of these approaches depends on an in-depth understanding of the mechanisms that lead to RGC dysfunction and death. In recent years, there has been an exponential increase in valuable information regarding the molecular basis of RGC death stemming from animal models of acute and chronic optic nerve injury as well as experimental glaucoma. The emerging landscape is complex and points at a variety of molecular signals - acting alone or in cooperation - to promote RGC death. These include: axonal transport failure, neurotrophic factor deprivation, toxic pro-neurotrophins, activation of intrinsic and extrinsic apoptotic signals, mitochondrial dysfunction, excitotoxic damage, oxidative stress, misbehaving reactive glia and loss of synaptic connectivity. Collectively, this body of work has considerably updated and expanded our view of how RGCs might die in glaucoma and has revealed novel, potential targets for neuroprotection.


Assuntos
Apoptose/fisiologia , Glaucoma/fisiopatologia , Células Ganglionares da Retina/patologia , Animais , Apoptose/efeitos dos fármacos , Transporte Axonal/efeitos dos fármacos , Progressão da Doença , Cães , Glaucoma/tratamento farmacológico , Glaucoma/metabolismo , Humanos , Camundongos , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Coelhos , Ratos , Receptor trkA/metabolismo , Retina/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo
12.
J Cell Physiol ; 225(3): 865-74, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20607800

RESUMO

Galpha(i)-coupled receptors comprise a diverse family of receptors that induce transformation by largely unknown mechanisms. We previously found that the Galpha(i)-coupled dopamine-D2short (D2S) receptor transforms Balb-D2S cells via Gαi3. To identify new Gαi effectors, a yeast two-hybrid screen was done using constitutively active Gαi3-Q204L as bait, and tumor necrosis factor-alpha (TNFα)-induced protein 8 (TNFAIP8, SCC-S2/NDED/GG2-1) was identified. In contrast, TNFAIP8-related TIPE1 and TIPE2 showed a very weak interaction with Gαi3. In yeast mating, in vitro pull-down, co-immunoprecipitation and bioluminescence resonance energy transfer (BRET) assays, TNFAIP8 preferentially interacted with activated Gαi proteins, consistent with direct Gαi-TNFAIP8 coupling. Over-expression or depletion of TNFAIP8 using antisense constructs in Balb-D2S cells did not affect D2S-induced signaling to Gαi-dependent inhibition of cAMP. In contrast, antisense depletion of TNFAIP8 completely inhibited spontaneous and D2S-induced foci formation, consistent with a role for TNFAIP8 in Gαi-dependent transformation. To address possible mechanisms, the effect of D2S signaling via TNFAIP8 on TNFα action was examined. D2S receptor activation inhibited TNFα-induced cell death in Balb-D2S cells, but not in cells depleted of TNFAIP8. However, depletion of TNFAIP8 did not prevent D2S-induced inhibition of TNFα-mediated caspase activation, suggesting that D2S/TNFAIP8-induced protection from TNFα-induced cell death is caspase-independent. The data suggest that Gαi-TNFAIP8-mediated rescue of pre-oncogenic cells enhances progression to oncogenic transformation, providing a selective target to inhibit cellular transformation.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Transformação Celular Neoplásica/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose/genética , Células 3T3 BALB , Caspases/metabolismo , Morte Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transferência Ressonante de Energia de Fluorescência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Imunoprecipitação , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Oligonucleotídeos Antissenso/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Técnicas do Sistema de Duplo-Híbrido
13.
J Neurochem ; 102(6): 1796-1804, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17767702

RESUMO

Although they have distinct functions, the signaling of dopamine-D(2) receptor short and long isoforms (D(2)S and D(2)L) is virtually identical. We compared inhibitory regulation of extracellular signal-regulated kinases (ERK1/2) in GH4 pituitary cells separately transfected with these isoforms. Activation of rat or human dopamine-D(2)S, muscarinic or somatostatin receptors inhibited thyrotropin-releasing hormone-induced ERK1/2 phosphorylation, while the D(2)L receptor failed to inhibit this response. In order to address the structural basis for the differential signaling of D(2)S and D(2)L receptors, we examined the D(2)L-SS mutant, in which a protein kinase C (PKC) pseudosubstrate site that is present in the D(2)L but not D(2)S receptor was converted to a consensus PKC site. In transfected GH4 cells, the D(2)L-SS mutant inhibited thyrotropin-releasing hormone-induced ERK1/2 phosphorylation almost as strongly as the D(2)S receptor. A D(2)S-triple mutant that eliminates PKC sites involved in D(2)S receptor desensitization also inhibited ERK1/2 activation. Similarly, in striatal cultures, the D(2)-selective agonist quinpirole inhibited potassium-stimulated ERK1/2 phosphorylation, indicating the presence of this pathway in neurons. In conclusion, the D(2)S and D(2)L receptors differ in inhibitory signaling to ERK1/2 due to specific residues in the D(2)L receptor alternatively spliced domain, which may account for differences in their function in vivo.


Assuntos
Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios/metabolismo , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/fisiologia , Processamento Alternativo/genética , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Células Cultivadas , Corpo Estriado/metabolismo , Agonistas de Dopamina/farmacologia , Humanos , Mutação/genética , Neurônios/efeitos dos fármacos , Fosforilação , Hipófise/metabolismo , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Quinase C/química , Estrutura Terciária de Proteína/genética , Ratos , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/genética , Transdução de Sinais/efeitos dos fármacos , Hormônio Liberador de Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA