Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Glia ; 71(10): 2343-2355, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37272718

RESUMO

Oligodendrocytes produce lipid-rich myelin sheaths that provide metabolic support to the underlying axon and facilitate saltatory conduction. Oligodendrocyte mitochondria supply the bulk of energy and carbon-chain backbones required for lipid synthesis. The sparsity of mitochondria in the myelin sheath suggests that tight regulation of mitochondrial trafficking is crucial for their efficient distribution in the cell. In particular, retention of mitochondria at axoglial junctions would support local lipid synthesis and membrane remodeling during myelination. How mitochondrial docking in oligodendrocytes is regulated is not known. Our findings indicate that syntaphilin (SNPH), a mitochondrial docking protein that has been characterized in neurons, is expressed by oligodendrocyte precursor cells (OPCs) and mature oligodendrocytes in vitro and present in the myelin sheath in vivo. We have previously reported that bath application of netrin-1 promotes the elaboration of myelin basic protein-positive membranes, and that localized presentation of a netrin-1 coated microbead results in rapid accumulation of mitochondria at the site of oligodendrocyte-bead adhesion. Here we show that netrin-1 increases the redistribution of SNPH to oligodendrocyte processes during the expansion of myelin basic protein-positive membranes and that SNPH clusters at the oligodendrocyte plasma membrane at sites of adhesion with netrin-1-coated beads where mitochondria are retained. These findings suggest roles for SNPH in oligodendrocytes regulating netrin-1-mediated mitochondrial docking and myelin membrane expansion.


Assuntos
Proteína Básica da Mielina , Bainha de Mielina , Bainha de Mielina/metabolismo , Proteína Básica da Mielina/metabolismo , Netrina-1/metabolismo , Oligodendroglia/metabolismo , Mitocôndrias/metabolismo , Lipídeos
2.
Glia ; 69(2): 392-412, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32910475

RESUMO

Mitochondria are dynamic organelles that produce energy and molecular precursors that are essential for myelin synthesis. Unlike in neurons, mitochondria in oligodendrocytes increase intracellular movement in response to glutamatergic activation and are more susceptible to oxidative stress than in astrocytes or microglia. The signaling pathways that regulate these cell type-specific mitochondrial responses in oligodendrocytes are not understood. Here, we visualized mitochondria migrating through thin cytoplasmic channels crossing myelin basic protein-positive compacted membranes and localized within paranodal loop cytoplasm. We hypothesized that local extracellular enrichment of netrin-1 might regulate the recruitment and function of paranodal proteins and organelles, including mitochondria. We identified rapid recruitment of mitochondria and paranodal proteins, including neurofascin 155 (NF155) and the netrin receptor deleted in colorectal carcinoma (DCC), to sites of contact between oligodendrocytes and netrin-1-coated microbeads in vitro. We provide evidence that Src-family kinase activation and Rho-associated protein kinase (ROCK) inhibition downstream of netrin-1 induces mitochondrial elongation, hyperpolarization of the mitochondrial inner membrane, and increases glycolysis. Our findings identify a signaling mechanism in oligodendrocytes that is sufficient to locally recruit paranodal proteins and regulate the subcellular localization, morphology, and function of mitochondria.


Assuntos
Dinâmica Mitocondrial , Receptor DCC , Metabolismo Energético , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Oligodendroglia/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Quinases Associadas a rho/metabolismo
3.
Reprod Biol Endocrinol ; 14(1): 56, 2016 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-27604347

RESUMO

BACKGROUND: Angiogenesis is an essential process in endometriosis disease progression. Earlier, we demonstrated that anti-angiogenic peptide, ABT-898 prevents neoangiogenesis of human endometriotic lesions in a xenograft mouse model. Since angiogenesis is essential for normal ovarian and uterine function, we evaluated effects of ABT-898 on normal female reproductive processes in mice. METHODS: Cycling female C57BL/6N mice were dosed with ABT-898 (100 mg/kg) or 5 % dextrose control for 21 consecutive days to cover multiple estrous cycles (average estrous cycle 4 to 5 days in mice). Pregnant female mice were dosed with ABT-898 (100 mg/kg) or control on alternate days over the course of gestation, beginning at gestation day 7.5 to 17.5 (gestation length 21 days). Histological analysis along with CD31 and Vimentin immunohistochemistry were performed on ovaries and uteri obtained from treated and control mice. To understand the influence of ABT-898 on systemic angiogenic factors, a Pro Mouse Cytokine 9-plex assay was performed on plasma samples obtained from mice prior to treatment, during the second week of ABAT-898 or control treatment and on the last day of treatment. RESULTS: ABT-898 did not affect the number of estrous cycles over the 21 day treatment compared to control. Histological analysis of ovaries found no difference in the number of primordial, primary, secondary, and antral follicles between ABT-898 treated and control groups. Similarly, no difference was observed in the microvessel density between ABT-898 treated and control uteri, ovarian follicles or corpus luteum when assessed using CD31 or vimentin immunohistochemistry. Electron microscopy revealed similar capillary structure and appearance in both ABT-898 treated and control uteri. Although peripheral blood angiogenic cytokine profiles (IL-15, IL-18, M-CSF, b-FGF, PDGF-bb, MIG, MIP-2, LIF and VEGF) changed over the course of the intervention, there was no significant difference between ABT-898 and control groups at any of the studied time points. Treatment with ABT-898 during pregnancy had no effect on litter size at birth, pup weight at birth or pup weight at weaning. CONCLUSION: Our findings suggest that ABT-898 may not alter angiogenesis dependent reproductive processes in female mice. However, an extensive reproductive toxicology screening is required to substantiate use of ABT-898 in future.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Materiais Biomiméticos/administração & dosagem , Oligopeptídeos/administração & dosagem , Reprodução/efeitos dos fármacos , Trombospondina 1/administração & dosagem , Animais , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Gravidez , Reprodução/fisiologia , Resultado do Tratamento
4.
Am J Pathol ; 181(2): 570-82, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22727957

RESUMO

Endometriosis is a common cause of pelvic pain and infertility in women, and a common indication for hysterectomy, yet the disease remains poorly diagnosed and ineffectively treated. Because endometriotic lesions require new blood supply for survival, inhibiting angiogenesis could provide a novel therapeutic strategy. ABT-898 mimics the antiangiogenic properties of thrombospondin-1, so we hypothesized that ABT-898 will prevent neovascularization of human endometriotic lesions and that ABT-898 treatment will not affect reproductive outcomes in a mouse model. Endometriosis was induced in BALB/c-Rag2(-/-)Il2rg(-/-) mice by surgical implantation of human endometrial fragments in the peritoneal cavity. Mice received daily injections of ABT-898 for 21 days. Flow cytometry was performed to measure circulating endothelial progenitor cells in peripheral blood. Cytokines were measured in plasma samples. Half of the ABT-898-treated and control mice were euthanized to assess neovascularization of endometriotic lesions, using CD31(+) immunofluorescence. The remaining mice were mated and euthanized at gestation day 12. Endometriotic lesions increased circulating endothelial progenitor cells 13 days after engraftment, relative to baseline. Endometriotic lesions from ABT-898-treated mice exhibited reduced neovascularization, compared with controls, and lesions had fewer CD31(+) microvessels. Chronic treatment with ABT-898 did not lead to any fetal anomalies or affect litter size at gestation day 12, compared with controls. Our results suggest that ABT-898 inhibits neovascularization of human endometriotic lesions without affecting mouse fecundity.


Assuntos
Endometriose/tratamento farmacológico , Endometriose/patologia , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Peptidomiméticos/uso terapêutico , Trombospondina 1/uso terapêutico , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/sangue , Modelos Animais de Doenças , Endometriose/sangue , Endometriose/diagnóstico por imagem , Endométrio/patologia , Ciclo Estral/efeitos dos fármacos , Feminino , Fertilidade/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Camundongos , Neovascularização Patológica/complicações , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Oligopeptídeos/farmacologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Reprodução/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia , Trombospondina 1/farmacologia , Ultrassonografia
5.
Macromol Biosci ; 19(5): e1900036, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30938926

RESUMO

Poly-d-lysine (PDL) and poly-l-lysine are standard surfaces for culturing neural cells; however, both are relatively unstable, costly, and the coated surface typically must be prepared immediately before use. Here, polyelectrolyte multilayers (PEMs) are employed as highly stable, relatively inexpensive, alternative substrates to support primary neural cell culture. Initial findings identify specific silk-based PEMs that significantly outperform the capacity of PDL to promote neuronal survival and process extension. Based on these results, a library of PEM variants, including commercial and bio-sourced polyelectrolytes, is generated and three silk-based PEMs that substantially outperform PDL as a substrate for primary neurons in cell culture are identified. Further, testing these PEM variants as substrates for primary oligodendrocyte progenitors demonstrates that one silk-based PEM functions significantly better than PDL. These findings reveal specificity of cellular responses, indicating that PEMs may be tuned to optimally support different neural cell types.


Assuntos
Proliferação de Células , Matriz Extracelular/química , Neurônios/metabolismo , Polieletrólitos , Polilisina , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Neurônios/citologia , Polieletrólitos/química , Polieletrólitos/farmacologia , Polilisina/química , Polilisina/farmacologia , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície
7.
Sci Rep ; 6: 30020, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27451946

RESUMO

Myelin injury in multiple sclerosis (MS) has been attributed both to "outside-in" primary immune mediated and "inside-out" metabolic stress of oligodendrocyte (OL) related mechanisms. Subsequent remyelination is dependent on recruitment and differentiation of oligodendrocyte progenitor cells (OPCs). RNS60 is a physically-modified saline containing charge-stabilized nanobubbles generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. Administration of RNS60 has been shown to reduce the severity of EAE by dampening the immune response and myelin loss. Additionally, RNS60 has been demonstrated to enhance mitochondrial ATP synthesis in neurons. Here, we used post-natal rat derived OLs and OPCs to assess the impact of RNS60 on the response of OLs to metabolic stress in vitro (glucose-nutrient deprivation, referred to as 'NG') and on OPC differentiation capacity. Under the NG condition, our findings indicate that RNS60 decreases caspases 3/7 activation. Respirometric analyses revealed that RNS60 increased spare glycolytic capacity (SGC) under normal culture conditions. However, RNS60 enhanced OL spare respiratory capacity (SRC) when a metabolic stress was present. Furthermore, we show that RNS60 promotes OPC differentiation under physiological conditions. Our findings provide evidence for the potential therapeutic efficacy of RNS60 through the promotion of OL survival and OPC differentiation.


Assuntos
Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Células-Tronco Neurais/citologia , Fármacos Neuroprotetores/farmacologia , Oxigênio/farmacologia , Remielinização/fisiologia , Cloreto de Sódio/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Glicólise/efeitos dos fármacos , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio/química , Estresse Fisiológico/efeitos dos fármacos
8.
PLoS One ; 10(3): e0121545, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25811892

RESUMO

Endometriosis is a gynecological disease defined by the growth of endometrium outside of the uterus. Although endometriosis contributes to 50% of female infertility cases, medical treatments are incompatible with pregnancy. Angiogenesis, the growth of blood vessels from existing vasculature, plays a crucial role in endometriotic lesion growth and survival. Previously, we demonstrated the effectiveness of thrombospondin-1 analog, ABT-898 (Abbott Laboratories) to inhibit endometriotic lesion vascularization in mice. We have now evaluated the trans-generational implications of ABT-898 treatment before and during mouse pregnancy. We hypothesized that ABT-898 would target lesion vasculature without affecting pregnancy, offspring development, or ovarian and uterine vascularity in mice. Endometriosis was induced using human endometrium in ß-estradiol-primed BALB/c-Rag-2-/-Il2rγ-/- mice receiving intraperitoneal injections of ABT-898 (25 mg/kg) or 5% dextrose control for 21 days. Ultrasound assessment of lesion vascularization revealed a reduction in blood flow supplying treated lesions. Excised ABT-898 treated lesions stained for CD31+ endothelial cells exhibited a decrease in microvessel density. Following confirmation of estrous cycling, mice were bred and treated with ABT-898 on gestation days 7, 9, 11, 13, 15, 17, and 19. ABT-898 did not affect estrous cycling or pregnancy parameters including litter size across generations and offspring weight gain. Quantification of angiogenic cytokine plasma levels revealed no significant differences between treatment groups. Vimentin staining of the uterus and ovary revealed no observable effects of ABT-898. Similarly, no obvious histological anomalies were observed in the kidney, liver, ovary, or uterus following ABT-898 treatment. These results suggest that ABT-898 effectively inhibit endometriotic lesion vascularization without affecting trans-generational pregnancy outcomes in mice.


Assuntos
Endometriose/metabolismo , Endometriose/fisiopatologia , Trombospondina 1/metabolismo , Animais , Citocinas/biossíntese , Citocinas/sangue , Modelos Animais de Doenças , Endometriose/diagnóstico por imagem , Endometriose/tratamento farmacológico , Endometriose/patologia , Células Endoteliais/metabolismo , Ciclo Estral , Feminino , Fertilidade/efeitos dos fármacos , Xenoenxertos , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Fígado/citologia , Fígado/efeitos dos fármacos , Camundongos , Camundongos Knockout , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Oligopeptídeos/farmacologia , Ovário/citologia , Ovário/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Resultado da Gravidez , Trombospondina 1/farmacologia , Ultrassonografia , Útero/citologia , Útero/efeitos dos fármacos
9.
J Reprod Immunol ; 97(1): 85-94, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23432875

RESUMO

Endometriosis is a gynecological disease characterized by the growth of endometrium outside of the uterine cavity. It is often associated with dysmenorrhea, dyspareunia, pelvic pain and infertility. One of the key requirements for endometriotic lesions to survive is development of a blood supply to support their growth. Indeed, dense vascularization is characteristic feature of endometriotic lesions. This has led to the idea that suppression of blood vessel growth (anti-angiogenic therapy) may be a successful therapeutic approach for endometriosis. Potential effectiveness of anti-angiogenic therapies has been assessed in some animal models but there are no reports of human clinical trials. Without understanding the specific mechanism by which endometriosis lesions establish a new blood supply, short-term animal experiments will have limited value for translation into human medicine. Further, it is crucial to use appropriate animal models to assess efficacy of anti-angiogenic compounds. Syngeneic and autologous rodent models, where endometrial fragments are auto-transplanted into the peritoneal cavity are commonly used in anti-angiogenic therapy studies. Another approach is xenograft models where human endometrium is engrafted into immunodeficient mice. Here we review the animal models and experimental techniques used to evaluate anti-angiogenic therapies for endometriosis. We also review our own work on the role of stromal cell derived factor-1 in the recruitment of endothelial progenitor cells in endometriotic lesion angiogenesis, and the effects of the anti-angiogenic peptide ABT-898, a thrombospondin-1 mimetic, on endometriotic lesion growth and vascular development.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endometriose/tratamento farmacológico , Endométrio/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Animais , Quimiocina CXCL12/antagonistas & inibidores , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Endométrio/patologia , Endométrio/transplante , Feminino , Humanos , Camundongos , Oligopeptídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa