Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
Nat Commun ; 15(1): 6845, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39122737

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2 , Retículo Endoplásmico , Péptido 1 Similar al Glucagón , Proteínas Nogo , Proglucagón , Proproteína Convertasa 1 , Animales , Humanos , Masculino , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Retículo Endoplásmico/metabolismo , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Aparato de Golgi/metabolismo , Células HEK293 , Insulina/metabolismo , Resistencia a la Insulina , Intestinos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nogo/metabolismo , Proteínas Nogo/genética , Proglucagón/metabolismo , Proglucagón/genética , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 1/genética , Unión Proteica , Proteolisis
2.
Hippocampus ; 34(9): 491-502, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39091158

RESUMEN

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.


Asunto(s)
Región CA2 Hipocampal , Plasticidad Neuronal , Proteínas Nogo , Sinapsis , Animales , Proteínas Nogo/metabolismo , Masculino , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Región CA2 Hipocampal/fisiología , Región CA2 Hipocampal/metabolismo , Región CA2 Hipocampal/efectos de los fármacos , Ratas Sprague-Dawley , Ratas , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Corteza Entorrinal/fisiología , Corteza Entorrinal/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Vías Nerviosas/fisiología , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/genética , Proteínas del Tejido Nervioso , Receptores de Factores de Crecimiento
3.
Cell Rep ; 43(7): 114357, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38955182

RESUMEN

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.


Asunto(s)
Calcio , Retículo Endoplásmico , Neuronas , Proteínas Nogo , Retículo Endoplásmico/metabolismo , Proteínas Nogo/metabolismo , Humanos , Calcio/metabolismo , Neuronas/metabolismo , Neuritas/metabolismo , Transporte Biológico , Proyección Neuronal/efectos de los fármacos
4.
Neurosci Biobehav Rev ; 163: 105767, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38885889

RESUMEN

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.


Asunto(s)
Mitocondrias , Esclerosis Múltiple , Proteínas Nogo , Humanos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Nogo/metabolismo , Animales , Transducción de Señal/fisiología , Axones/metabolismo , Axones/patología
5.
Hum Cell ; 37(5): 1378-1393, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38858338

RESUMEN

We aim to investigate the effect of RVG-Lamp2b-modified exosomes (exos) loaded with neurotrophin-3 (NT-3) on facial nerve injury. Exos were collected from control cells (Ctrl Exo) or bone marrow mesenchymal stem cells co-transfected with RVG-Lamp2b and NT-3 plasmids (RVG-NT-3 Exo) by gradient centrifugation and identified by western blotting, transmission electron microscopy, and nanoparticle tracking analysis. Effect of RVG-NT-3 Exo on oxidative stress damage was determined by analysis of the morphology, viability, and ROS production of neurons. Effect of RVG-NT-3 Exo on facial nerve axotomy (FNA) was determined by detecting ROS production, neuroinflammatory reaction, microglia activation, facial motor neuron (FMN) death, and myelin sheath repair. Loading NT-3 and modifying with RVG-Lamp2b did not alter the properties of the exos. Moreover, RVG-NT-3 Exo could effectively target neurons to deliver NT-3. Treatment with RVG-NT-3 Exo lowered H2O2-induced oxidative stress damage in primary neurons and Nsc-34 cells. RVG-NT-3 Exo treatment significantly decreased ROS production, neuroinflammatory response, FMN death, and elevated microglia activation and myelin sheath repair in FNA rat models. Our findings suggested that RVG-NT-3 Exo-mediated delivery of NT-3 is effective for the treatment of facial nerve injury.


Asunto(s)
Exosomas , Traumatismos del Nervio Facial , Células Madre Mesenquimatosas , Neurotrofina 3 , Estrés Oxidativo , Exosomas/metabolismo , Exosomas/trasplante , Neurotrofina 3/administración & dosificación , Neurotrofina 3/metabolismo , Neurotrofina 3/genética , Traumatismos del Nervio Facial/terapia , Traumatismos del Nervio Facial/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Ratas , Células Cultivadas , Humanos , Modelos Animales de Enfermedad , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratas Sprague-Dawley , Sistemas de Liberación de Medicamentos/métodos
6.
Tissue Eng Regen Med ; 21(5): 777-789, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38630369

RESUMEN

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.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Autofagia , Cirrosis Hepática , Proteínas Nogo , Animales , Humanos , Masculino , Ratones , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Tetracloruro de Carbono , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , Proteínas Nogo/metabolismo
7.
Redox Biol ; 68: 102944, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37890359

RESUMEN

AIMS: Endothelial dysfunction plays a pivotal role in atherosclerosis, but the detailed mechanism remains incomplete understood. Nogo-B is an endoplasmic reticulum (ER)-localized protein mediating ER-mitochondrial morphology. We previously showed endothelial Nogo-B as a key regulator of endothelial function in the setting of hypertension. Here, we aim to further assess the role of Nogo-B in coronary atherosclerosis in ApoE-/- mice with pressure overload. METHODS AND RESULTS: We generated double knockout (DKO) mouse models of systemically or endothelium-specifically excising Nogo-A/B gene on an ApoE-/- background. After 7 weeks of transverse aortic constriction (TAC) surgery, compared to ApoE-/- mice DKO mice were resistant to the development of coronary atherosclerotic lesions and plaque rapture. Sustained elevation of Nogo-B and adhesion molecules (VCAM-1/ICAM-1), early markers of atherosclerosis, was identified in heart tissues and endothelial cells (ECs) isolated from TAC ApoE-/- mice, changes that were significantly repressed by Nogo-B deficiency. In cultured human umbilical vein endothelial cells (HUVECs) exposure to inflammatory cytokines (TNF-α, IL-1ß), Nogo-B was upregulated and activated reactive oxide species (ROS)-p38-p65 signaling axis. Mitofusin 2 (Mfn2) is a key protein tethering ER to mitochondria in ECs, and we showed that Nogo-B expression positively correlated with Mfn2 protein level. And Nogo-B deletion in ECs or in ApoE-/- mice reduced Mfn2 protein content and increased ER-mitochondria distance, reduced ER-mitochondrial Ca2+ transport and mitochondrial ROS generation, and prevented VCAM-1/ICAM-1 upregulation and EC dysfunction, eventually restrained atherosclerotic lesions development. CONCLUSION: Our study revealed that Nogo-B is a critical modulator in promoting endothelial dysfunction and consequent pathogenesis of coronary atherosclerosis in pressure overloaded hearts of ApoE-/- mice. Nogo-B may hold the promise to be a common therapeutic target in the setting of hypertension.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Hipertensión , Placa Aterosclerótica , Humanos , Animales , Ratones , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Placa Aterosclerótica/metabolismo , Estrés Oxidativo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/metabolismo , Endotelio/metabolismo , Hipertensión/metabolismo , Apolipoproteínas E/genética , Ratones Noqueados , Ratones Endogámicos C57BL
8.
J Biol Chem ; 299(10): 105232, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37690690

RESUMEN

After adult mammalian central nervous system injury, axon regeneration is extremely limited or absent, resulting in persistent neurological deficits. Axon regeneration failure is due in part to the presence of inhibitory proteins, including NogoA (Rtn4A), from which two inhibitory domains have been defined. When these inhibitory domains are deleted, but an amino-terminal domain is still expressed in a gene trap line, mice show axon regeneration and enhanced recovery from injury. In contrast, when there is no amino-terminal Nogo-A fragment in the setting of inhibitory domain deletion, then axon regeneration and recovery are indistinguishable from WT. These data indicated that an amino-terminal Nogo-A fragment derived from the gene trap might promote axon regeneration, but this had not been tested directly and production of this fragment without gene targeting was unclear. Here, we describe posttranslation production of an amino-terminal fragment of Nogo-A from the intact gene product. This fragment is created by proteolysis near amino acid G214-N215 and levels are enhanced by axotomy. Furthermore, this fragment promotes axon regeneration in vitro and acts cell autonomously in neurons, in contrast to the inhibitory extracellular action of other Nogo-A domains.Proteins interacting with the amino-terminal Nogo-A fragment by immunoprecipitation include HSPA8 (HSC70, HSP7C). Suppression of HSPA8 expression by shRNA decreases axon regeneration from cerebral cortical neurons and overexpression increases axon regeneration. Moreover, the amino-terminal Nogo-A fragment increases HSPA8 chaperone activity. These data provide an explanation for varied results in different gene-targeted Nogo-A mice, as well as revealing an axon regeneration promoting domain of Nogo-A.


Asunto(s)
Axones , Proteínas de la Mielina , Animales , Ratones , Axones/metabolismo , Inhibidores de Crecimiento/metabolismo , Mamíferos/metabolismo , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Regeneración Nerviosa/fisiología , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Proteolisis , Femenino , Ratones Endogámicos C57BL
9.
Glia ; 71(10): 2473-2494, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37401784

RESUMEN

Nogo-A, B, and C are well described members of the reticulon family of proteins, most well known for their negative regulatory effects on central nervous system (CNS) neurite outgrowth and repair following injury. Recent research indicates a relationship between Nogo-proteins and inflammation. Microglia, the brain's immune cells and inflammation-competent compartment, express Nogo protein, although specific roles of the Nogo in these cells is understudied. To examine inflammation-related effects of Nogo, we generated a microglial-specific inducible Nogo KO (MinoKO) mouse and challenged the mouse with a controlled cortical impact (CCI) traumatic brain injury (TBI). Histological analysis shows no difference in brain lesion sizes between MinoKO-CCI and Control-CCI mice, although MinoKO-CCI mice do not exhibit the levels of ipsilateral lateral ventricle enlargement as injury matched controls. Microglial Nogo-KO results in decreased lateral ventricle enlargement, microglial and astrocyte immunoreactivity, and increased microglial morphological complexity compared to injury matched controls, suggesting decreased tissue inflammation. Behaviorally, healthy MinoKO mice do not differ from control mice, but automated tracking of movement around the home cage and stereotypic behavior, such as grooming and eating (termed cage "activation"), following CCI is significantly elevated. Asymmetrical motor function, a deficit typical of unilaterally brain lesioned rodents, was not detected in CCI injured MinoKO mice, while the phenomenon was present in CCI injured controls 1-week post-injury. Overall, our studies show microglial Nogo as a negative regulator of recovery following brain injury. To date, this is the first evaluation of the roles microglial specific Nogo in a rodent injury model.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Proteínas Nogo , Animales , Ratones , Lesiones Encefálicas/patología , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Proteínas Nogo/metabolismo
10.
Int J Mol Sci ; 24(5)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36901909

RESUMEN

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that causes progressive neurological disability in most patients due to neurodegeneration. Activated immune cells infiltrate the CNS, triggering an inflammatory cascade that leads to demyelination and axonal injury. Non-inflammatory mechanisms are also involved in axonal degeneration, although they are not fully elucidated yet. Current therapies focus on immunosuppression; however, no therapies to promote regeneration, myelin repair, or maintenance are currently available. Two different negative regulators of myelination have been proposed as promising targets to induce remyelination and regeneration, namely the Nogo-A and LINGO-1 proteins. Although Nogo-A was first discovered as a potent neurite outgrowth inhibitor in the CNS, it has emerged as a multifunctional protein. It is involved in numerous developmental processes and is necessary for shaping and later maintaining CNS structure and functionality. However, the growth-restricting properties of Nogo-A have negative effects on CNS injury or disease. LINGO-1 is also an inhibitor of neurite outgrowth, axonal regeneration, oligodendrocyte differentiation, and myelin production. Inhibiting the actions of Nogo-A or LINGO-1 promotes remyelination both in vitro and in vivo, while Nogo-A or LINGO-1 antagonists have been suggested as promising therapeutic approaches for demyelinating diseases. In this review, we focus on these two negative regulators of myelination while also providing an overview of the available data on the effects of Nogo-A and LINGO-1 inhibition on oligodendrocyte differentiation and remyelination.


Asunto(s)
Proteínas de la Membrana , Proteínas Nogo , Remielinización , Proteínas de la Membrana/metabolismo , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nogo/metabolismo , Oligodendroglía/metabolismo , Humanos
11.
Cardiovasc Toxicol ; 23(3-4): 147-160, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36964845

RESUMEN

Among the three isoforms encoded by neurite outgrowth inhibitor proteins has been intensely investigated as a central nervous system inhibitor. Although neurite outgrowth inhibitor protein-A (Nogo-A) expression is increased in plasma of patients who have experienced a coronary heart disease, its role in heart disease is not well elucidated. In this study, we discovered a significant increase in Nogo-A expression in diabetic myocardial ischemia reperfusion (MI/R) injury conditions. Accelerated Nogo-A and MI/R injury in diabetic rats was attenuated by tauroursodeoxycholic acid treatment and knockdown of Nogo-A per se is sufficient to decrease endoplasmic reticulum (ER) stress as well as prevents cardiomyocyte apoptosis. We hypothesized that decreased Nogo-A levels might reducing diabetic MI/R injury. Nogo-A interacted with C/EBP homologous protein, suggesting a role for Nogo-A in ER stress during diabetic MI/R. In conclusion, Nogo-A mediated ER stress plays a major role in diabetic MI/R injury, and pathologically altered Nogo-A expression mediates diabetic MI/R injury, suggesting Nogo-A as a novel target for the treatment of diabetic MI/R injury in clinical settings.


Asunto(s)
Diabetes Mellitus Experimental , Daño por Reperfusión Miocárdica , Ratas , Animales , Daño por Reperfusión Miocárdica/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Proteínas Nogo/metabolismo , Estrés del Retículo Endoplásmico , Apoptosis , Miocitos Cardíacos/metabolismo
12.
Cardiovasc Res ; 119(2): 506-519, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35815623

RESUMEN

AIMS: Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS: We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION: Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.


Asunto(s)
Insuficiencia Cardíaca , Esfingolípidos , Humanos , Ratones , Animales , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Esfingolípidos/metabolismo , Ceramidas/metabolismo , Insuficiencia Cardíaca/genética , Miocitos Cardíacos/metabolismo
13.
Biol Trace Elem Res ; 201(7): 3381-3386, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36057764

RESUMEN

Metabolic dysfunction is a critical step in the etiopathogenesis of Alzheimer's disease. In this progressive neurological disorder, impaired zinc homeostasis has a key role that needs to be clarified. The aim of this study was to investigate the effect of zinc deficiency and administration on hippocampal Nogo-A receptor and osteocalcin gene expression in rats injected with intracerebroventricular streptozotocin (icv-STZ). Forty male Wistar rats were divided into 5 groups in equal numbers: Sham 1 group received icv artificial cerebrospinal fluid (aCSF); Sham 2 group received icv a CSF and i.p. saline; STZ group received 3 mg/kg icv STZ; STZ-Zn-deficient group received 3 mg/kg icv STZ and fed a zinc-deprived diet; STZ-Zn-supplemented group received 3 mg/kg icv STZ and i.p. zinc sulfate (5 mg/kg/day). Hippocampus tissue samples were taken following the cervical dislocation of the animals under general anesthesia. Nogo-A receptor and osteocalcin gene expression levels were determined by real-time-PCR method. Zinc supplementation attenuated the increase in hippocampal Nogo-A receptor gene expression, which was significantly increased in zinc deficiency. Again, zinc supplementation upregulated the intrinsic protective mechanisms of the brain by activating osteocalcin-expressing cells in the brain. The results of the study show that zinc has critical effects on Nogo-A receptor gene expression and hippocampal osteocalcin gene expression levels in the memory-sensitive rat hippocampus that is impaired by icv-STZ injection. These results are the first to examine the effect of zinc deficiency and supplementation on hippocampal Nogo-A receptor and osteocalcin gene expression in icv-STZ injection in rats.


Asunto(s)
Enfermedad de Alzheimer , Zinc , Ratas , Masculino , Animales , Estreptozocina/farmacología , Ratas Wistar , Proteínas Nogo/metabolismo , Proteínas Nogo/farmacología , Osteocalcina/genética , Osteocalcina/metabolismo , Zinc/farmacología , Zinc/metabolismo , Enfermedad de Alzheimer/patología , Hipocampo/metabolismo , Modelos Animales de Enfermedad , Aprendizaje por Laberinto
14.
Semin Cell Dev Biol ; 139: 111-120, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35431138

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive decline. Synaptic impairment is one of the first events to occur in the progression of this disease. Synaptic plasticity and cellular association of various plastic events have been shown to be affected in AD models. Nogo-A, a well-known axonal growth inhibitor with a recently discovered role as a plasticity suppressor, and its main receptor Nogo-66 receptor 1 (NGR1) have been found to be overexpressed in the hippocampus of Alzheimer's patients. However, the role of Nogo-A and its receptor in the pathology of AD is still widely unknown. In this work we set out to investigate whether Nogo-A is working as a plasticity suppressor in AD. Our results show that inhibition of the Nogo-A pathway via the Nogo-R antibody in an Alzheimer's mouse model, APP/PS1, leads to the restoration of both synaptic plasticity and associativity in a protein synthesis and NMDR-dependent manner. We also show that inhibition of the p75NTR pathway, which is strongly associated with NGR1, restores synaptic plasticity as well. Mechanistically, we propose that the restoration of synaptic plasticity in APP/PS1 via inhibition of the Nogo-A pathway is due to the modulation of the RhoA-ROCK2 pathway and increase in plasticity related proteins. Our study identifies Nogo-A as a plasticity suppressor in AD models hence targeting Nogo-A could be a promising strategy to understanding AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Proteínas Nogo/metabolismo , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Modelos Animales de Enfermedad , Precursor de Proteína beta-Amiloide/genética
15.
Cells ; 11(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36497029

RESUMEN

Current therapeutics targeting chronic phases of multiple sclerosis (MS) are considerably limited in reversing the neural damage resulting from repeated inflammation and demyelination insults in the multi-focal lesions. This inflammation is propagated by the activation of microglia, the endogenous immune cell aiding in the central nervous system homeostasis. Activated microglia may transition into polarized phenotypes; namely, the classically activated proinflammatory phenotype (previously categorized as M1) and the alternatively activated anti-inflammatory phenotype (previously, M2). These transitional microglial phenotypes are dynamic states, existing as a continuum. Shifting microglial polarization to an anti-inflammatory status may be a potential therapeutic strategy that can be harnessed to limit neuroinflammation and further neurodegeneration in MS. Our research has observed that the obstruction of signaling by inhibitory myelin proteins such as myelin-associated inhibitory factor, Nogo-A, with its receptor (NgR), can regulate microglial cell function and activity in pre-clinical animal studies. Our review explores the microglial role and polarization in MS pathology. Additionally, the potential therapeutics of targeting Nogo-A/NgR cellular mechanisms on microglia migration, polarization and phagocytosis for neurorepair in MS and other demyelination diseases will be discussed.


Asunto(s)
Microglía , Esclerosis Múltiple , Animales , Microglía/metabolismo , Proteínas Nogo/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Transducción de Señal , Proteínas de la Mielina/metabolismo , Inflamación/patología
16.
Cell Rep ; 41(4): 111505, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36288715

RESUMEN

Gene-based therapeutic strategies to lower ataxin-2 levels are emerging for the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia type 2 (SCA2). Additional strategies to lower levels of ataxin-2 could be beneficial. Here, we perform a genome-wide arrayed small interfering RNA (siRNA) screen in human cells and identify RTN4R, the gene encoding the RTN4/NoGo-Receptor, as a potent modifier of ataxin-2 levels. RTN4R knockdown, or treatment with a peptide inhibitor, is sufficient to lower ataxin-2 protein levels in mouse and human neurons in vitro, and Rtn4r knockout mice have reduced ataxin-2 levels in vivo. We provide evidence that ataxin-2 shares a role with the RTN4/NoGo-Receptor in limiting axonal regeneration. Reduction of either protein increases axonal regrowth following axotomy. These data define the RTN4/NoGo-Receptor as a novel therapeutic target for ALS and SCA2 and implicate the targeting of ataxin-2 as a potential treatment following nerve injury.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ataxias Espinocerebelosas , Animales , Ratones , Humanos , Ataxina-2/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , ARN Interferente Pequeño , Receptores Nogo/metabolismo , Ataxias Espinocerebelosas/genética , Ratones Noqueados , Péptidos/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo
17.
Cells ; 11(19)2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36231046

RESUMEN

Nogo-B has been reported to play a critical role in angiogenesis and the repair of damaged blood vessels; however, its role in the tumor microenvironment remains unclear. Here, we observed the differential expression of Nogo-B in endothelial cells from hepatocellular carcinoma (HCC) and glioma samples. Downregulation of Nogo-B expression correlated with the malignant phenotype of cancer and a poor prognosis for patients. In subsequent studies, endothelial Nogo-B inhibition robustly promoted the growth of HCC or glioma xenografts in nude mice. Intriguingly, endothelial Nogo-B silencing dramatically suppressed endothelial cell expansion and tumor angiogenesis, but potently enhanced the proliferation of neighboring HCC and glioma cells. Based on the results of the ELISA assay, Nogo-B silencing reduced TGF-ß production in endothelial cells, which attenuated the phosphorylation and nuclear translocation of Smad in neighboring cancer cells. The endothelial Nogo-B silencing-mediated increase in cancer cell proliferation was abolished by either a TGF-ß neutralizing antibody or TGF-ß receptor inhibitor, indicating the essential role for TGF-ß in endothelial Nogo-B-mediated suppression of cancer growth. These findings not only broaden our understanding of the crosstalk between cancer cells and endothelial cells but also provide a novel prognostic biomarker and a therapeutic target for cancer treatments.


Asunto(s)
Carcinoma Hepatocelular , Glioma , Neoplasias Hepáticas , Proteínas Nogo/metabolismo , Animales , Anticuerpos Neutralizantes , Biomarcadores , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Neovascularización Patológica , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
18.
Zhen Ci Yan Jiu ; 47(9): 843-6, 2022 Sep 25.
Artículo en Chino | MEDLINE | ID: mdl-36153461

RESUMEN

The corticospinal tract (CST), descending from the frontoparietal cortex and traveling down to terminate at the anterior horn of the spinal cord to mediate voluntary movements, is frequently injured from the infarcted or hemorrhagic cerebrovascular insults due to stroke. Under the circumstances, motor dysfunction seriously affects the patient's quality of life. Acupuncture therapy has a sequelae, especially in improving motor deficits. In the present paper, we reviewed the current development of researches on acupuncture treatment of poststroke motor dysfunction and its biological mechanisms from 1) delaying patients' development of neuronal degeneration and white matter fibrosis (Wallerian degeneration), 2) improving patients' upper limb motor function and daily life ability by promoting the repair of white matter tracts and CST on the affected side, 3) promoting the compensation of CST on the healthy side, 4) reconstructing the motor conduction pathway to strengthen the bilateral brain connection in ex-perimental animals, and 5) strengthening the sprouting of the contralateral CST to dominate the affected side again across the midline. In addition, acupuncture stimulation induced improvement of axonal rewiring for corticospinal innervation is also possibly related to its functions in accelerating the synthesis and release of neurotrophic factors, down-regulating Nogo-A/RhoA signaling and activating vascular epithelial growth factor/Dll4/Notch signaling pathways.


Asunto(s)
Terapia por Acupuntura , Corteza Motora , Accidente Cerebrovascular , Animales , Corteza Motora/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Proteínas Nogo/metabolismo , Tractos Piramidales/metabolismo , Calidad de Vida , Recuperación de la Función/fisiología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/terapia
19.
Nat Commun ; 13(1): 5294, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36075894

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) is a restriction factor that limits viral pathogenesis and exerts poorly understood immunoregulatory functions. Here, using human and mouse models, we demonstrate that IFITM3 promotes MyD88-dependent, TLR-mediated IL-6 production following exposure to cytomegalovirus (CMV). IFITM3 also restricts IL-6 production in response to influenza and SARS-CoV-2. In dendritic cells, IFITM3 binds to the reticulon 4 isoform Nogo-B and promotes its proteasomal degradation. We reveal that Nogo-B mediates TLR-dependent pro-inflammatory cytokine production and promotes viral pathogenesis in vivo, and in the case of TLR2 responses, this process involves alteration of TLR2 cellular localization. Nogo-B deletion abrogates inflammatory cytokine responses and associated disease in virus-infected IFITM3-deficient mice. Thus, we uncover Nogo-B as a driver of viral pathogenesis and highlight an immunoregulatory pathway in which IFITM3 fine-tunes the responsiveness of myeloid cells to viral stimulation.


Asunto(s)
COVID-19 , Interleucina-6 , Proteínas Nogo/metabolismo , Animales , Citocinas/metabolismo , Humanos , Interleucina-6/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2 , Receptor Toll-Like 2/metabolismo
20.
Int J Mol Sci ; 23(15)2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35955811

RESUMEN

Thoracic spinal cord injury (SCI) results in urinary dysfunction, which majorly affects the quality of life of SCI patients. Abnormal sprouting of lumbosacral bladder afferents plays a crucial role in this condition. Underlying mechanisms may include changes in expression of regulators of axonal growth, including chondroitin sulphate proteoglycans (CSPGs), myelin-associated inhibitors (MAIs) and repulsive guidance molecules, known to be upregulated at the injury site post SCI. Here, we confirmed lumbosacral upregulation of the growth-associated protein GAP43 in SCI animals with bladder dysfunction, indicating the occurrence of axonal sprouting. Neurocan and Phosphacan (CSPGs), as well as Nogo-A (MAI), at the same spinal segments were upregulated 7 days post injury (dpi) but returned to baseline values 28 dpi. In turn, qPCR analysis of the mRNA levels for receptors of those repulsive molecules in dorsal root ganglia (DRG) neurons showed a time-dependent decrease in receptor expression. In vitro assays with DRG neurons from SCI rats demonstrated that exposure to high levels of NGF downregulated the expression of some, but not all, receptors for those regulators of axonal growth. The present results, therefore, show significant molecular changes at the lumbosacral cord and DRGs after thoracic lesion, likely critically involved in neuroplastic events leading to urinary impairment.


Asunto(s)
Traumatismos de la Médula Espinal , Vejiga Urinaria Hiperactiva , Animales , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Ganglios Espinales/metabolismo , Proteínas Nogo/metabolismo , Calidad de Vida , Ratas , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Vejiga Urinaria Hiperactiva/etiología , Vejiga Urinaria Hiperactiva/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA