Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Hippocampus ; 34(9): 491-502, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39091158

RESUMO

Hippocampal area CA2 has garnered attention in recent times owing to its significant involvement in social memory and distinctive plasticity characteristics. Research has revealed that the CA2 region demonstrates a remarkable resistance to plasticity, particularly in the Schaffer Collateral (SC)-CA2 pathway. In this study we investigated the role of Nogo-A, a well-known axon growth inhibitor and more recently discovered plasticity regulator, in modulating plasticity within the CA2 region. The findings demonstrate that blocking Nogo-A in male rat hippocampal slices facilitates the establishment of both short-term and long-term plasticity in the SC-CA2 pathway, while having no impact on the Entorhinal Cortical (EC)-CA2 pathway. Additionally, the study reveals that inhibiting Nogo-A enables association between the SC and EC pathways. Mechanistically, we confirm that Nogo-A operates through its well-known co-receptor, p75 neurotrophin receptor (p75NTR), and its downstream signaling factor such as Rho-associated protein kinase (ROCK), as their inhibition also allows plasticity induction in the SC-CA2 pathway. Additionally, the induction of long-term depression (LTD) in both the EC and SC-CA2 pathways led to persistent LTD, which was not affected by Nogo-A inhibition. Our study demonstrates the involvement of Nogo-A mediated signaling mechanisms in limiting synaptic plasticity within the CA2 region.


Assuntos
Região CA2 Hipocampal , Plasticidade Neuronal , Proteínas Nogo , Sinapses , Animais , Proteínas Nogo/metabolismo , Masculino , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Região CA2 Hipocampal/fisiologia , Região CA2 Hipocampal/metabolismo , Região CA2 Hipocampal/efeitos dos fármacos , Ratos Sprague-Dawley , Ratos , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Córtex Entorrinal/fisiologia , Córtex Entorrinal/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Vias Neurais/fisiologia , Proteínas da Mielina/metabolismo , Proteínas da Mielina/genética , Proteínas do Tecido Nervoso , Receptores de Fatores de Crescimento
2.
Tissue Eng Regen Med ; 21(5): 777-789, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38630369

RESUMO

BACKGROUND: Hepatic fibrosis (HF) is a histopathological change in the process of long-term liver injury caused by cytokine secretion and internal environment disturbance, resulting in excessive liver repair and fiber scar. Nogo-B protein is widely distributed in peripheral tissues and organs and can regulate the migration of endothelial cells by activating TGF-ß1 in vascular remodeling after injury. Nogo-B has been shown to promote organ fibrosis. This study was to determine the role of Nogo-B in HF. METHODS: An HF model was built by intraperitoneal injections with 20% carbon tetrachloride. Localization of Nogo-B was detected by FISH. The interaction between Nogo-B and BACE1 was confirmed by Co-IP. Autophagy flux was analyzed using tandem mRFP-GFP-LC3 fluorescence microscopy, electron microscopy, and western blotting. Detection of serum AST and ALT and H&E staining were utilized to detect the degree of liver injury. The HF was evaluated by Masson trichromatic staining. RT-qPCR, western blotting, and immunofluorescence were employed to detect relevant indicators. RESULTS: Reducing Nogo-B suppressed AST and ALT levels, the accumulation of collagen I and α-SMA, and expressions of pro-fibrotic genes in mouse liver. BACE1 was a potential downstream target of Nogo-B. Nogo-B was upregulated in TGF-ß1-activated hepatic stellate cells (HSCs). Knocking down Nogo-B caused the downregulation of pro-fibrotic genes and inhibited viability of HSCs. Nogo-B knockdown prevented CCL4-induced fibrosis, accompanied by downregulation of extracellular matrix. Nogo-B inhibited HSC autophagy and increased lipid accumulation. BACE1 knockdown inhibited HSC autophagy and activation in LX-2 cells. CONCLUSION: Nogo-B knockdown prevents HF by directly inhibiting BACe1-mediated autophagy.


Assuntos
Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases , Autofagia , Cirrose Hepática , Proteínas Nogo , Animais , Humanos , Masculino , Camundongos , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Tetracloreto de Carbono , Células Estreladas do Fígado/metabolismo , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos Endogâmicos C57BL , Proteínas Nogo/metabolismo
3.
Cell Rep ; 43(7): 114357, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38955182

RESUMO

Cell functions rely on intracellular transport systems distributing bioactive molecules with high spatiotemporal accuracy. The endoplasmic reticulum (ER) tubular network constitutes a system for delivering luminal solutes, including Ca2+, across the cell periphery. How the ER structure enables this nanofluidic transport system is unclear. Here, we show that ER membrane-localized reticulon 4 (RTN4/Nogo) is sufficient to impose neurite outgrowth inhibition in human cortical neurons while acting as an ER morphoregulator. Improving ER transport visualization methodologies combined with optogenetic Ca2+ dynamics imaging and in silico modeling, we observed that ER luminal transport is modulated by ER tubule narrowing and dilation, proportional to the amount of RTN4. Excess RTN4 limited ER luminal transport and Ca2+ release, while RTN4 elimination reversed the effects. The described morphoregulatory effect of RTN4 defines the capacity of the ER for peripheral Ca2+ delivery for physiological releases and thus may constitute a mechanism for controlling the (re)generation of neurites.


Assuntos
Cálcio , Retículo Endoplasmático , Neurônios , Proteínas Nogo , Retículo Endoplasmático/metabolismo , Proteínas Nogo/metabolismo , Humanos , Cálcio/metabolismo , Neurônios/metabolismo , Neuritos/metabolismo , Transporte Biológico , Crescimento Neuronal/efeitos dos fármacos
4.
Neurosci Biobehav Rev ; 163: 105767, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38885889

RESUMO

Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.


Assuntos
Mitocôndrias , Esclerose Múltipla , Proteínas Nogo , Humanos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Nogo/metabolismo , Animais , Transdução de Sinais/fisiologia , Axônios/metabolismo , Axônios/patologia
5.
Nat Commun ; 15(1): 6845, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39122737

RESUMO

Glucagon-like peptide 1 (GLP1), which is mainly processed and cleaved from proglucagon in enteroendocrine cells (EECs) of the intestinal tract, acts on the GLP1 receptor in pancreatic cells to stimulate insulin secretion and to inhibit glucagon secretion. However, GLP1 processing is not fully understood. Here, we show that reticulon 4B (Nogo-B), an endoplasmic reticulum (ER)-resident protein, interacts with the major proglucagon fragment of proglucagon to retain proglucagon on the ER, thereby inhibiting PCSK1-mediated cleavage of proglucagon in the Golgi. Intestinal Nogo-B knockout in male type 2 diabetes mellitus (T2DM) mice increases GLP1 and insulin levels and decreases glucagon levels, thereby alleviating pancreatic injury and insulin resistance. Finally, we identify aberrantly elevated Nogo-B expression and inhibited proglucagon cleavage in EECs from diabetic patients. Our study reveals the subcellular regulatory processes involving Nogo-B during GLP1 production and suggests intestinal Nogo-B as a potential therapeutic target for T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Retículo Endoplasmático , Peptídeo 1 Semelhante ao Glucagon , Proteínas Nogo , Proglucagon , Pró-Proteína Convertase 1 , Animais , Humanos , Masculino , Camundongos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Retículo Endoplasmático/metabolismo , Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Complexo de Golgi/metabolismo , Células HEK293 , Insulina/metabolismo , Resistência à Insulina , Intestinos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nogo/metabolismo , Proteínas Nogo/genética , Proglucagon/metabolismo , Proglucagon/genética , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 1/genética , Ligação Proteica , Proteólise
6.
Acta Physiologica Sinica ; (6): 301-308, 2022.
Artigo em Chinês | WPRIM | ID: wpr-927606

RESUMO

Nogo-B receptor (NgBR) is a specific receptor of Nogo-B, a member of reticulon 4 protein family. It is widely expressed in many tissues and mainly located in cell membrane and endoplasmic reticulum. Previous studies have revealed that NgBR is involved in a variety of physiological and pathophysiological processes, such as dolichol synthesis, lipid metabolism, cholesterol trafficking, insulin resistance, vascular remodeling and angiogenesis, tumorigenesis and nervous system diseases. Further studies on the molecular characteristics and biological function of NgBR might be of great significance to understand its role in diverse diseases and provide possible clinical strategies for the treatment of diseases.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Metabolismo dos Lipídeos , Proteínas Nogo/metabolismo , Receptores de Superfície Celular/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa