Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Eur J Neurosci ; 59(10): 2628-2645, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38491850

RESUMEN

Over the past few decades, diabetes gradually has become one of the top non-communicable disorders, affecting 476.0 million in 2017 and is predicted to reach 570.9 million people in 2025. It is estimated that 70 to 100% of all diabetic patients will develop some if not all, diabetic complications over the course of the disease. Despite different symptoms, mechanisms underlying the development of diabetic complications are similar, likely stemming from deficits in both neuronal and vascular components supplying hyperglycaemia-susceptible tissues and organs. Diaph1, protein diaphanous homolog 1, although mainly known for its regulatory role in structural modification of actin and related cytoskeleton proteins, in recent years attracted research attention as a cytoplasmic partner of the receptor of advanced glycation end-products (RAGE) a signal transduction receptor, whose activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines in diabetes and its related complications. Both Diaph1 and RAGE are also a part of the RhoA signalling cascade, playing a significant role in the development of neurovascular disturbances underlying diabetes-related complications. In this review, based on the existing knowledge as well as compelling findings from our past and present studies, we address the role of Diaph1 signalling in metabolic stress and neurovascular degeneration in diabetic complications. In light of the most recent developments in biochemical, genomic and transcriptomic research, we describe current theories on the aetiology of diabetes complications, highlighting the function of the Diaph1 signalling system and its role in diabetes pathophysiology.


Asunto(s)
Forminas , Transducción de Señal , Humanos , Animales , Forminas/metabolismo , Transducción de Señal/fisiología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Complicaciones de la Diabetes/metabolismo , Neuropatías Diabéticas/metabolismo
2.
J Mol Med (Berl) ; 101(8): 1015-1028, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37462767

RESUMEN

Multiple molecular pathways including the receptor for advanced glycation end-products-diaphanous related formin 1 (RAGE-Diaph1) signaling are known to play a role in diabetic peripheral neuropathy (DPN). Evidence suggests that neuropathological alterations in type 1 diabetic spinal cord may occur at the same time as or following peripheral nerve abnormalities. We demonstrated that DPN was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. More than 500 differentially expressed genes (DEGs) belonging to multiple functional pathways were identified in diabetic spinal cord and of those the most enriched was RAGE-Diaph1 related PI3K-Akt pathway. Only seven of spinal cord DEGs overlapped with DEGs from type 1 diabetic sciatic nerve and only a single gene cathepsin E (CTSE) was common for both type 1 and type 2 diabetic mice. In silico analysis suggests that molecular changes in spinal cord may act synergistically with RAGE-Diaph1 signaling axis in the peripheral nerve. KEY MESSAGES: Molecular perturbations in spinal cord may be involved in the progression of diabetic peripheral neuropathy. Diabetic peripheral neuropathy was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. In silico analysis revealed that PI3K-Akt signaling axis related to RAGE-Diaph1 was the most enriched biological pathway in diabetic spinal cord. Cathepsin E may be the target molecular hub for intervention against diabetic peripheral neuropathy.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Neuropatías Diabéticas , Hiperglucemia , Animales , Ratones , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/complicaciones , Catepsina E , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Nervio Ciático/patología , Hiperglucemia/genética , Hiperglucemia/patología
3.
Eur J Neurosci ; 57(10): 1642-1656, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37070486

RESUMEN

This review focuses on receptor for advanced glycation endproducts/diaphonous related formin 1 (RAGE/Diaph1) interaction as a modulator of actin cytoskeleton dynamics in peripheral nervous system (PNS) in diabetes. Deciphering the complex molecular interactions between RAGE and Diaph1 is crucial in expanding our understanding of diabetic length dependent neuropathy (DLDN). DLDN is a common neurological disorder in patients with diabetes. It is well known that actin cytoskeletal homeostasis is disturbed during DLDN. Thus, we review the current status of knowledge about RAGE/Diaph1 impact on actin cytoskeletal malfunctions in PNS and DLDN progression in diabetes. We also survey studies about small molecules that may block RAGE/Diaph1 axis and thus inhibit the progression of DLDN. Finally, we explore examples of cytoskeletal long-non coding RNAs (lncRNAs) currently unrelated to DLDN, to discuss their potential role in this disease. Most recent studies indicated that lncRNAs have a great potential in many research areas, including RAGE/Diaph1 axis as well as DLDN. Altogether, this review is aimed at giving us an insight into the involvement of cytoskeletal lncRNAs in DLDN.


Asunto(s)
Hiperglucemia , ARN Largo no Codificante , Humanos , Transducción de Señal , Actinas , Receptor para Productos Finales de Glicación Avanzada , Citoesqueleto de Actina/metabolismo , Sistema Nervioso Periférico/metabolismo , Forminas/metabolismo
4.
Neuropathology ; 43(1): 84-94, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35915909

RESUMEN

Type 1 diabetes (T1D) may affect the peripheral nervous system and alter the expression of proteins contributing to inflammation and cellular cytoskeleton dysfunction, in most cases leading to the development of diabetic length-dependent neuropathy (DLDN). In the present study, we performed immunohistochemistry (IHC) to probe the expression of the receptor for advanced glycation end products (RAGE); its key ligands, high-mobility group box 1 (HMGB1), S100 calcium-binding protein B (S100B), and carboxymethyl-lysine (CML - advanced glycation end products (AGE)); and its cytoplasmic tail-binding partner, diaphanous related formin 1 (DIAPH1) and associated molecules, beta-actin (ACTB) and profilin 1 (PFN1) proteins in sciatic nerves harvested from seven-month old FVB/OVE26 mice with genetically-mediated T1D. We found that the amount of RAGE, HMGB1, and S100B proteins was elevated in diabetic vs the non-diabetic groups, while the amount of DIAPH1, ACTB, as well as PFN1 proteins did not differ between these groups. Moreover, our data revealed linear dependence between RAGE and HMGB1 proteins. Interaction criss-cross of selected sets of proteins in the sciatic nerve revealed that there were connected in a singular network. Our results indicate that T1D may alter expression patterns of RAGE axis proteins and thus contribute to DLDN.


Asunto(s)
Diabetes Mellitus Tipo 1 , Neuropatías Diabéticas , Proteína HMGB1 , Ratones , Animales , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteína HMGB1/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Nervio Ciático
5.
Life (Basel) ; 11(11)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34833143

RESUMEN

The objective of the study was to compare the effects of experimentally induced type 1 or type 2 diabetes (T1D or T2D) on the functional, structural and biochemical properties of mouse peripheral nerves. Eight-week-old C57BL/6 mice were randomly assigned into three groups, including the control (CTRL, chow-fed), STZ (streptozotocin (STZ)-injected), and HFD (high-fat diet (HFD)-fed) group. After 18-weeks of experimental treatment, HFD mice had higher body weights and elevated levels of plasma lipids, while STZ mice developed hyperglycemia. STZ-treated mice, after an extended period of untreated diabetes, developed motor and sensory nerve conduction-velocity deficits. Moreover, relative to control fibers, pre- and diabetic axons were lower in number and irregular in shape. Animals from both treatment groups manifested a pronounced overexpression of nNOS and a reduced expression of SOD1 proteins in the sciatic nerve, indicating oxidative-nitrosative stress and ineffective antioxidant protection in the peripheral nervous system of these mice. Collectively, STZ- and HFD-treated mice revealed similar characteristics of peripheral nerve damage, including a number of morphological and electrophysiological pathologies in the sciatic nerve. While hyperglycemia is a large component of diabetic neuropathy pathogenesis, the non-hyperglycemic effects of diabetes, including dyslipidemia, may also be of importance in the development of this condition.

6.
Biochem Biophys Res Commun ; 559: 92-98, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33933994

RESUMEN

Synaptic transmission is mediated by neurotransmitters that are stored in synaptic vesicles (SV) and released at the synaptic active zone (AZ). While in recent years major progress has been made in unraveling the molecular machinery responsible for SV docking, fusion and exocytosis, the mechanisms governing AZ protein and SV trafficking through axons still remain unclear. Here, we performed stop-flow nerve ligation to examine axonal trafficking of endogenous AZ and SV proteins. Rat sciatic nerves were collected 1 h, 3 h and 8 h post ligation and processed for immunohistochemistry and electron microscopy. First, we followed the transport of an integral synaptic vesicle protein, SV2A and a SV-associated protein involved in SV trafficking, Rab3a, and observed that while SV2A accumulated on both sides of ligation, Rab3a was only noticeable in the proximal segment of the ligated nerve indicating that only SV trans-membrane protein SV2A displayed a bi-directional axonal transport. We then demonstrate that multiple AZ proteins accumulate rapidly on either side of the ligation with a timescale similar to that of SV2A. Overall, our data uncovers an unexpected robust bi-directional, coordinated -trafficking of SV and AZ proteins in peripheral nerves. This implies that pathological disruption of axonal trafficking will not only impair trafficking of newly synthesized proteins to the synapse but will also affect retrograde transport, leading to neuronal dysfunction and likely neurodegeneration.


Asunto(s)
Transporte Axonal , Proteínas del Tejido Nervioso/metabolismo , Nervios Periféricos/fisiología , Transporte de Proteínas , Vesículas Sinápticas/metabolismo , Animales , Masculino , Ratas Sprague-Dawley , Transmisión Sináptica
7.
Int J Mol Sci ; 20(11)2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31141951

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal progressive neurodegenerative disease characterized by a permanent degeneration of both upper and lower motor neurons. Many different genes and pathophysiological processes contribute to this disease, however its exact cause remains unclear. Therefore, it is necessary to understand this heterogeneity to find effective treatments. In this review, we focus on selected environmental and genetic risk factors predisposing to ALS and highlight emerging treatments in ALS therapy. Of numerous defective genes associated with ALS, we focus on four principal genes that have been identified as definite causes of ALS: the SOD1 gene, C9orf72, TDP-43, as well as the recently identified TBK1. We also provide up-to-date information on selected environmental factors that have historically been considered as key players in ALS development and pathogenesis. In parallel to our survey of known risk factors, we also discuss emerging ALS stem cell therapies and experimental medicines currently undergoing phase II and III clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Predisposición Genética a la Enfermedad , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Animales , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Terapia Genética , Humanos , Terapia Molecular Dirigida , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
8.
Neurochem Int ; 126: 154-164, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30902646

RESUMEN

The Receptor for Advanced Glycation End Products (RAGE) is expressed by multiple cell types in the brain and spinal cord that are linked to the pathogenesis of neurovascular and neurodegenerative disorders, including neurons, glia (microglia and astrocytes) and vascular cells (endothelial cells, smooth muscle cells and pericytes). Mounting structural and functional evidence implicates the interaction of the RAGE cytoplasmic domain with the formin, Diaphanous1 (DIAPH1), as the key cytoplasmic hub for RAGE ligand-mediated activation of cellular signaling. In aging and diabetes, the ligands of the receptor abound, both in the central nervous system (CNS) and in the periphery. Such accumulation of RAGE ligands triggers multiple downstream events, including upregulation of RAGE itself. Once set in motion, cell intrinsic and cell-cell communication mechanisms, at least in part via RAGE, trigger dysfunction in the CNS. A key outcome of endothelial dysfunction is reduction in cerebral blood flow and increased permeability of the blood brain barrier, conditions that facilitate entry of activated leukocytes into the CNS, thereby amplifying primary nodes of CNS cellular stress. This contribution details a review of the ligands of RAGE, the mechanisms and consequences of RAGE signal transduction, and cites multiple examples of published work in which RAGE contributes to the pathogenesis of neurovascular perturbation. Insights into potential therapeutic modalities targeting the RAGE signal transduction axis for disorders of CNS vascular dysfunction and neurodegeneration are also discussed.


Asunto(s)
Enfermedades del Sistema Nervioso Central/metabolismo , Forminas/metabolismo , Mediadores de Inflamación/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Enfermedades Vasculares/metabolismo , Animales , Enfermedades del Sistema Nervioso Central/patología , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Vasculares/patología
9.
Brain Behav ; 7(1): e00558, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28127506

RESUMEN

INTRODUCTION: Chemotherapy-induced neuropathies are one of the most common side effects of cancer treatment, surpassing bone marrow suppression and kidney dysfunction. Chemotherapy effects on the nervous system vary between different classes of drugs and depend on specific chemical and physical properties of the drug used. The three most neurotoxic classes of anti-cancer drugs are: platinum-based drugs, taxanes, and thalidomide and its analogs; other, less neurotoxic but also commonly used drugs are: bortezomib, ixabepilone, and vinca alkaloids. METHODS: Here, in this paper, based on our experience and current knowledge, we provide a short review of the most common, neuropathy-inducing anti-cancer drugs, describe the most prevalent neuropathy symptoms produced by each of them, and outline preventive measures and treatment guidelines for cancer patients suffering from neuropathy and for their health care providers. RESULTS: Patients should be encouraged to report any signs of neuropathic pain, alteration in sensory perception, tingling, numbness, burning, increased hot/cold sensitivity and motor dysfunctions as early as possible. If known neurotoxic chemotherapeutics are used, a neurological examination with electrophysiological evaluation should be implemented early in the course of treatment so, both patients and physicians would be better prepared to cope with possible neurotoxic effects. CONCLUSIONS: The use of neurotoxic chemotherapeutics should be closely monitored and if clinically permitted, that is, if a patient shows signs of cancer regression, drug doses should be reduced or combined with other less neurotoxic anti-cancer medication. If not counteractive, the use of over the counter antineuropathic supplements such as calcium or magnesium might be encouraged. If physically possible, patients should also be encouraged to exercise regularly and avoid factors that might increase nerve damage such as excessive drinking, smoking, or sitting in a cramped position.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Humanos
10.
Front Cell Neurosci ; 10: 117, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27242430

RESUMEN

The etiology of amyotrophic lateral sclerosis (ALS), a fatal motor neuron disorder characterized by progressive muscle weakness and spasticity, remains largely unknown. Approximately 5-10% of cases are familial, and of those, 15-20% are associated with mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1). Mutations of the SOD1 gene interrupt cellular homeostasis and contribute to cellular toxicity evoked by the presence of altered SOD1, along with other toxic species, such as advanced glycation end products (AGEs). AGEs trigger activation of their chief cell surface receptor, RAGE (receptor for advanced glycation end products), and induce RAGE-dependent cellular stress and inflammation in neurons, thereby affecting their function and leading to apoptosis. Here, we show for the first time that the expression of RAGE is higher in the SOD1 transgenic mouse model of ALS vs. wild-type mouse spinal cord. We tested whether pharmacological blockade of RAGE may delay the onset and progression of disease in this mouse model. Our findings reveal that treatment of SOD1 transgenic mice with soluble RAGE (sRAGE), a natural competitor of RAGE that sequesters RAGE ligands and blocks their interaction with cell surface RAGE, significantly delays the progression of ALS and prolongs life span compared to vehicle treatment. We demonstrate that in sRAGE-treated SOD1 transgenic animals at the final stage of the disease, a significantly higher number of neurons and lower number of astrocytes is detectable in the spinal cord. We conclude that RAGE antagonism may provide a novel therapeutic strategy for ALS intervention.

11.
Neurosci Biobehav Rev ; 62: 48-55, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26724598

RESUMEN

RAGE, the receptor of advanced glycation end-products, is thought to be one of the potential contributors to the neurodegeneration. It has been shown that RAGE activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines. RAGE involvement has been documented in the pathogenesis of a number of neurodegenerative diseases such amyotrophic lateral sclerosis (ALS), Alzheimer's, Parkinson's, Huntington's, Creutzfeld-Jakob' diseases and various neurodegenerative conditions such as diabetic neuropathy, familial amyloid polyneuropathy, Charcot neuroarthropathy and vasculitic neuropathy. Although the detailed mechanisms of RAGE contribution to the neurodegeneration remains unclear, studies indicate that RAGE detrimental actions are exerted via its binding to the pro-inflammatory ligands such as advanced glycation end-products, S100/calgranulin and amphoterin and subsequent activation of downstream regulatory pathways such as NF-κB, STAT and JKN pathways. Here, in this review we attempt to shed light onto molecular events and pathological pathways involved in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis (ALS)--a progressive and fatal neurodegenerative disorder, summarizing current knowledge and the prospect of RAGE in the pathogenesis of this disastrous disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Inflamación/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Transducción de Señal/fisiología , Animales , Proteínas Portadoras/metabolismo , Humanos , Enfermedades Neurodegenerativas/inmunología , Estrés Oxidativo/inmunología , Estrés Oxidativo/fisiología
12.
Front Cell Neurosci ; 9: 485, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26733811

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder of largely unknown pathogenesis. Recent studies suggest that enhanced oxidative stress and neuroinflammation contribute to the progression of the disease. Mounting evidence implicates the receptor for advanced glycation end-products (RAGE) as a significant contributor to the pathogenesis of certain neurodegenerative diseases and chronic conditions. It is hypothesized that detrimental actions of RAGE are triggered upon binding to its ligands, such as AGEs (advanced glycation end products), S100/calgranulin family members, and High Mobility Group Box-1 (HMGB1) proteins. Here, we examined the expression of RAGE and its ligands in human ALS spinal cord. Tissue samples from age-matched human control and ALS spinal cords were tested for the expression of RAGE, carboxymethyllysine (CML) AGE, S100B, and HMGB1, and intensity of the immunofluorescent and immunoblotting signals was assessed. We found that the expression of both RAGE and its ligands was significantly increased in the spinal cords of ALS patients versus age-matched control subjects. Our study is the first report describing co-expression of both RAGE and its ligands in human ALS spinal cords. These findings suggest that further probing of RAGE as a mechanism of neurodegeneration in human ALS is rational.

13.
J Mol Neurosci ; 55(2): 297-304, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24854048

RESUMEN

The superior cervical ganglion (SCG) is a center of sympathetic innervation of all head and neck organs. SCG sympathetic preganglionic neurons (SPN) were found in the nucleus intermediolateralis pars principalis (IMLpp), the nucleus intermediolateralis pars funicularis (IMLpf), the nucleus intercalatus spinalis (IC), and the nucleus intercalatus spinalis pars paraependymalis (ICpe). Despite its importance, little is known of SCG innervation and chemical coding in the laboratory pig, a model that is physiologically and anatomically representative of humans. Here in our study, we established the distribution and chemical coding of Fast Blue (FB) retrogradely labelled SPN innervating porcine SCG. After unilateral injection of FB retrograde tracer into the left SCG, labeled neurons were found solely on the ipsilateral side with approximately 98% located in Th1-Th3 segments and predominantly distributed in the IMLpp and IMLpf. Neurochemical analysis revealed that approximately 80% of SPN were positive both to choline acetyltransferase (ChAT) and nitric oxide synthase (NOS) and were surrounded by a plethora of opioidergic and peptiergic nerve terminals. The results of our study provide a detailed description of the porcine preganglionic neuroarchitecture of neurons controlling the SCG, setting the stage for further studies concerning SPN plasticity under experimental/pathological conditions.


Asunto(s)
Neuronas/metabolismo , Ganglio Cervical Superior/citología , Amidinas/farmacocinética , Animales , Colina O-Acetiltransferasa/metabolismo , Óxido Nítrico Sintasa/metabolismo , Ganglio Estrellado/citología , Porcinos
14.
Acta Histochem ; 116(1): 106-11, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23830992

RESUMEN

Peripheral neuropathy (PN) involves widespread peripheral nerve disorders affecting a large human population worldwide. In Europe and the United States, the first single most prominent cause of peripheral neuropathy is diabetes, affecting 60-70% patients with long-term diabetes followed by idiopathic neuropathy, peripheral nerve damage of unknown etiology, diagnosed in 10-40% of all patients admitted to hospitals with symptoms of peripheral nerve damage. The molecular mechanisms underlying the pathogenesis of this disorder are not yet fully understood, however a few potential molecular contributors, such as Munc13-1, have been recently suggested. Munc13-1 is a diacylglycerol (DAG) receptor and a multifunction active zone protein essential for synaptic vesicle priming and crucial for insulin release from pancreatic beta cells. Here, for the first time, we focused on the comparative expression of Munc13-1 in human and porcine peripheral nerves. Our results revealed significantly reduced number of Munc13-1 in human (64.26% ± 6.68%) and porcine (84.09% ± 2.21%) diabetic nerve fibers and lower number of the double stained, neuronal marker, Neurofilament (NF) and Munc13-1 positive, human (56.83% ± 3.77%) and porcine (65.87% ± 4.86%) nerve fibers. Optical density quantification of Western blots showed similar results. Our study indicates that Munc13-1, on account of its role in both insulin and neurotransmitter exocytosis and through its binding properties, may be an important factor contributing to the development or progression of diabetic neuropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Neuropatías Diabéticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Humanos , Filamentos Intermedios/metabolismo , Masculino , Persona de Mediana Edad , Nervios Periféricos/metabolismo , Sus scrofa
15.
Brain Behav ; 3(6): 701-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24363972

RESUMEN

BACKGROUND: Diabetic neuropathy and idiopathic neuropathy are among the most prevalent neuropathies in human patients. The molecular mechanism underlying pathological changes observed in the affected nerve remains unclear but one candidate molecule, the receptor for advanced glycation end-products (RAGE), has recently gained attention as a potential contributor to neuropathy. Our previous studies revealed that RAGE expression is higher in porcine and murine diabetic nerve, contributing to the inflammatory mechanisms leading to diabetic neuropathy. Here, for the first time, we focused on the expression of RAGE in human peripheral nerve. METHODS: Our study utilized de-identified human sural nerve surplus obtained from 5 non-neuropathic patients (control group), 6 patients with long-term mild-to-moderate diabetic neuropathy (diabetic group) and 5 patients with mild-to-moderate peripheral neuropathy of unknown etiology (idiopathic group). By using immunofluorescent staining and protein immunoblotting we studied the expression and colocalization patterns of RAGE and its ligands: carboxymethyllysine (CML), high mobility group box 1 (HMBG1) and mammalian Diaphanous 1 (mDia1) in control and neuropathic nerves. RESULTS: We found that in a normal, healthy human nerve, RAGE is expressed in almost 30% of all nerve fibers and that number is higher in pathological states such as peripheral neuropathy. We established that the levels of RAGE and its pro-inflammatory ligands, CML and HMBG1, are higher in both idiopathic and diabetic nerve, while the expression of the RAGE cytoplasmic domain-binding partner, mDia1 is similar among control, diabetic, and idiopathic nerve. The highest number of double stained nerve fibers was noted for RAGE and CML: ∼76% (control), ∼91% (idiopathic) and ∼82% (diabetic) respectively. CONCLUSIONS: Our data suggest roles for RAGE and its inflammatory ligands in human peripheral neuropathies and lay the foundation for further, more detailed and clinically oriented investigation involving these proteins and their roles in disorders of the human peripheral nerve.

16.
Eur J Neurosci ; 38(8): 3159-68, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23941591

RESUMEN

Diabetic peripheral nerve dysfunction is a common complication occurring in 30-50% of long-term diabetic patients. The pathogenesis of this dysfunction remains unclear but growing evidence suggests that it might be attributed, in part, to alteration in axonal transport. Our previous studies demonstrated that RAGE (Receptor for Advanced Glycation Endproducts) contributes to the pathogenesis of diabetic peripheral neuropathy and impairs nerve regeneration consequent to sciatic nerve crush, particularly in diabetes. We hypothesize that RAGE plays a role in axonal transport impairment via the interaction of its cytoplasmic domain with mammalian Diaphanous 1 (mDia1) - actin interacting molecule. Studies showed that mDia1-RAGE interaction is necessary for RAGE-ligand-dependent cellular migration, AKT phosphorylation, macrophage inflammatory response and smooth muscle migration. Here, we studied RAGE, mDia1 and markers of axonal transport rates in the peripheral nerves of wild-type C57BL/6 and RAGE null control and streptozotocin-injected diabetic mice at 1, 3 and 6 h after sciatic nerve crush. The results show that in both control and diabetic nerves, the amount of RAGE accumulated at the proximal and distal side of the crush area is similar, indicating that the recycling rate for RAGE is very high and that it is evenly transported from and towards the neuronal cell body. Furthermore, we show that slow axonal transport of proteins such as Neurofilament is affected by diabetes in a RAGE-independent manner. Finally, our study demonstrates that mDia1 axonal transport is impaired in diabetes, suggesting that diabetes-related changes affecting actin binding proteins occur early in the course of the disease.


Asunto(s)
Transporte Axonal , Diabetes Mellitus Experimental/metabolismo , Receptores Inmunológicos/metabolismo , Nervio Ciático/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/metabolismo , Diabetes Mellitus Experimental/patología , Forminas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Compresión Nerviosa , Proteínas de Neurofilamentos/metabolismo , Unión Proteica , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Nervio Ciático/patología
17.
Diabetes ; 62(3): 931-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23172920

RESUMEN

Peripheral neuropathy and insensate limbs and digits cause significant morbidity in diabetic individuals. Previous studies showed that deletion of the receptor for advanced end-glycation products (RAGE) in mice was protective in long-term diabetic neuropathy. Here, we tested the hypothesis that RAGE suppresses effective axonal regeneration in superimposed acute peripheral nerve injury attributable to tissue-damaging inflammatory responses. We report that deletion of RAGE, particularly in diabetic mice, resulted in significantly higher myelinated fiber densities and conduction velocities consequent to acute sciatic nerve crush compared with wild-type control animals. Consistent with key roles for RAGE-dependent inflammation, reconstitution of diabetic wild-type mice with RAGE-null versus wild-type bone marrow resulted in significantly improved axonal regeneration and restoration of function. Diabetic RAGE-null mice displayed higher numbers of invading macrophages in the nerve segments postcrush compared with wild-type animals, and these macrophages in diabetic RAGE-null mice displayed greater M2 polarization. In vitro, treatment of wild-type bone marrow-derived macrophages with advanced glycation end products (AGEs), which accumulate in diabetic nerve tissue, increased M1 and decreased M2 gene expression in a RAGE-dependent manner. Blockade of RAGE may be beneficial in the acute complications of diabetic neuropathy, at least in part, via upregulation of regeneration signals.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Neuropatías Diabéticas/metabolismo , Regeneración Nerviosa , Receptores Inmunológicos/metabolismo , Nervio Ciático/fisiopatología , Neuropatía Ciática/metabolismo , Animales , Trasplante de Médula Ósea , Células Cultivadas , Neuropatías Diabéticas/inmunología , Neuropatías Diabéticas/patología , Neuropatías Diabéticas/prevención & control , Productos Finales de Glicación Avanzada/metabolismo , Inmunohistoquímica , Ligandos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/trasplante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Compresión Nerviosa/efectos adversos , Conducción Nerviosa , Especificidad de Órganos , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Nervio Ciático/inmunología , Nervio Ciático/lesiones , Nervio Ciático/patología , Neuropatía Ciática/inmunología , Neuropatía Ciática/patología , Neuropatía Ciática/prevención & control
18.
Neurosci Lett ; 500(1): 57-62, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21683765

RESUMEN

The main goal of our study was to investigate the chemical coding of the superior cervical ganglion (SCG) sympathetic neurons supplying the porcine parotid gland. Additionally, the chemical nature of the vicinal nerve fibers surrounding the parotid SCG perikarya was investigated. Fast blue (FB) retrograde tracing of the parotid gland and immunofluorescent labelling of SCG neurons were studied in juvenile female pigs. Microscopic analysis revealed that only ipsilateral SCG neurons were retrogradely labelled. The labelled neurons formed a discrete cluster in the middle and caudal region of the ganglion. Immunofluorescent labelling revealed that virtually all of the FB-positive parotid gland neurons were immunoreactive to tyrosine hydroxylase (TH), confirming their sympathetic nature. In addition to TH, the majority of the FB-positive neurons were found to be immunoreactive to calbindin (CB) and to a lesser extent for neuropeptide Y (NPY), leu-enkephalin (LENK) and galanin (GAL). In the close proximity of the FB-traced perikarya, a large number of immunoreactive (IR) vasoactive intestinal peptide (VIP-IR), pituitary adenylate cyclase-activating polypeptide (PACAP-IR), nitric oxide synthase (NOS-IR) processes were identified. Moreover, calcitonin gene related peptide-immunoreactive (CGRP-IR), substance P-immunoreactive (SP-IR), vesicular acetylcholine transporter (VAChT-IR), calretinin (CRT-IR), GAL-IR, LENK-IR and CB-IR protrusions were observed. The results of the present study provide a detailed characteristic of the location and neurochemical coding of sympathetic SCG neurons innervating the parotid salivary gland of the pig and lay ground for more advanced, clinical studies on salivary gland innervations.


Asunto(s)
Neuronas/metabolismo , Glándula Parótida/inervación , Ganglio Cervical Superior/metabolismo , Animales , Femenino , Inmunohistoquímica , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , Ganglio Cervical Superior/citología , Porcinos
19.
Biochem Insights ; 2010(3): 47-59, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21852897

RESUMEN

The aim of our project was to study the effect of streptozotocin (STZ)-induced hyperglycemia on sciatic nerve morphology, blood plasma markers and immunohistochemical expression of RAGE (the Receptor for Advanced Glycation End-products), and its ligands-S100B and Carboxymethyl Lysine (CML)-advanced glycation endproduct (AGE) in the laboratory pig. Six months after STZ-injections, blood plasma measurements, morphometric analysis of sciatic nerve fiber density, immunofluorescent distribution of potential molecular neuropathy contributors, ELISA measurement of plasma AGE level and HPLC analysis of sciatic nerve levels of one of the pre-AGE and the glycolysis intermediate products-methyl-glyoxal (MG) were performed. The results of our study revealed that STZ-injected animals displayed elevated levels of plasma glucose, gamma glutamyl transferase (GGT) and triglycerides. The sciatic nerve of STZ-injected pigs revealed significantly lower numbers of small-diameter myelinated fibers, higher immunoreactivity for RAGE and S100B and increased levels of MG as compared to control animals. Our results correspond to clinical findings in human patients with hyperglycemia/diabetes-evoked peripheral neuropathy and suggest that the domestic pig may be a suitable large animal model for the study of mechanisms underlying hyperglycemia-induced neurological complications in the peripheral nerve and may serve as a relevant model for the pre-clinical assessment of candidate drugs in neuropathy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...