Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(8): e0290155, 2023.
Article in English | MEDLINE | ID: mdl-37594969

ABSTRACT

Exosomes derived from endothelial cells and Schwann cells have been employed as novel treatments of neurological diseases, including peripheral neuropathy. Exosomal cargo plays a critical role in mediating recipient cell function. In this study, we thus performed a comprehensive proteomic analysis of exosomes derived from healthy mouse dermal microvascular endothelial cells (EC-Exo) and healthy mouse Schwann cells (SC-Exo). We detected 1,817and 1,579 proteins in EC-Exo and SC-Exo, respectively. Among them, 1506 proteins were present in both EC-Exo and SC-Exo, while 311 and 73 proteins were detected only in EC-Exo and SC-Exo, respectively. Bioinformatic analysis revealed that EC-Exo enriched proteins were involved in neurovascular function, while SC-Exo enriched proteins were related to lipid metabolism. Western blot analysis of 14 enriched proteins revealed that EC-Exo contained proteins involved in mediating endothelial function such as delta-like 4 (DLL4) and endothelial NOS (NOS3), whereas SC-Exo had proteins involved in mediating glial function such as apolipoprotein A-I (APOA1) and phospholipid transfer protein (PLTP). Collectively, the present study identifies differences in the cargo protein profiles of EC-Exo and SC-Exo, thus providing new molecular insights into their biological functions for the treatment of peripheral neuropathy.


Subject(s)
Endothelial Cells , Exosomes , Animals , Mice , Proteomics , Schwann Cells , Neuroglia
2.
J Extracell Vesicles ; 10(5): e12073, 2021 03.
Article in English | MEDLINE | ID: mdl-33728031

ABSTRACT

There are no effective treatments for chemotherapy induced peripheral neuropathy (CIPN). Small extracellular vesicles (sEVs) facilitate intercellular communication and mediate nerve function and tumour progression. We found that the treatment of mice bearing ovarian tumour with sEVs derived from cerebral endothelial cells (CEC-sEVs) in combination with a chemo-drug, oxaliplatin, robustly reduced oxaliplatin-induced CIPN by decreasing oxaliplatin-damaged myelination and nerve fibres of the sciatic nerve and significantly amplified chemotherapy of oxaliplatin by reducing tumour size. The combination therapy substantially increased a set of sEV cargo-enriched miRNAs, but significantly reduced oxaliplatin-increased proteins in the sciatic nerve and tumour tissues. Bioinformatics analysis revealed the altered miRNAs and proteins formed two distinct networks that regulate neuropathy and tumour growth, respectively. Intravenously administered CEC-sEVs were internalized by axons of the sciatic nerve and cancer cells. Reduction of CEC-sEV cargo miRNAs abolished the effects of CEC-sEVs on oxaliplatin-inhibited axonal growth and on amplification of the anti-cancer effect in ovarian cancer cells, suggesting that alterations in the networks of miRNAs and proteins in recipient cells contribute to the therapeutic effect of CEC-sEVs on CIPN. Together, the present study demonstrates that CEC-sEVs suppressed CIPN and enhanced chemotherapy of oxaliplatin in the mouse bearing ovarian tumour.


Subject(s)
Antineoplastic Agents/therapeutic use , Extracellular Vesicles/metabolism , Ovarian Neoplasms/drug therapy , Oxaliplatin/therapeutic use , Peripheral Nervous System Diseases/therapy , Animals , Antineoplastic Agents/adverse effects , Axons/drug effects , Cell Line, Tumor , Extracellular Vesicles/transplantation , Female , Humans , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Nerve Fibers/metabolism , Nerve Fibers, Myelinated/metabolism , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/chemically induced
3.
Diabetes ; 69(4): 749-759, 2020 04.
Article in English | MEDLINE | ID: mdl-31915154

ABSTRACT

Schwann cell-derived exosomes communicate with dorsal root ganglia (DRG) neurons. The current study investigated the therapeutic effect of exosomes derived from healthy Schwann cells (SC-Exos) on diabetic peripheral neuropathy (DPN). We found that intravenous administration of SC-Exos to type 2 diabetic db/db mice with peripheral neuropathy remarkably ameliorated DPN by improving sciatic nerve conduction velocity and increasing thermal and mechanical sensitivity. These functional improvements were associated with the augmentation of epidermal nerve fibers and remyelination of sciatic nerves. Quantitative RT-PCR and Western blot analysis of sciatic nerve tissues showed that SC-Exo treatment reversed diabetes-reduced mature form of miRNA (miR)-21, -27a, and -146a and diabetes-increased semaphorin 6A (SEMA6A); Ras homolog gene family, member A (RhoA); phosphatase and tensin homolog (PTEN); and nuclear factor-κB (NF-κB). In vitro data showed that SC-Exos promoted neurite outgrowth of diabetic DRG neurons and migration of Schwann cells challenged by high glucose. Collectively, these novel data provide evidence that SC-Exos have a therapeutic effect on DPN in mice and suggest that SC-Exo modulation of miRs contributes to this therapy.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/physiopathology , Exosomes/transplantation , Schwann Cells/cytology , Animals , Ganglia, Spinal/physiopathology , Male , Mice , Sciatic Nerve/physiopathology
4.
Diabetologia ; 63(2): 431-443, 2020 02.
Article in English | MEDLINE | ID: mdl-31740984

ABSTRACT

AIMS/HYPOTHESIS: Diabetic peripheral neuropathy (DPN) is one of the major complications of diabetes, which contributes greatly to morbidity and mortality. There is currently no effective treatment for this disease. Exosomes are cell-derived nanovesicles and play an important role in intercellular communications. The present study investigated whether mesenchymal stromal cell (MSC)-derived exosomes improve neurological outcomes of DPN. METHODS: Exosomes were isolated from the medium of cultured mouse MSCs by ultracentrifugation. Diabetic mice (BKS.Cg-m+/+Leprdb/J, db/db) at the age of 20 weeks were used as DPN models. Heterozygous mice (db/m) of the same age were used as the control. MSC-exosomes were administered weekly via the tail vein for 8 weeks. Neurological function was evaluated by testing motor and sensory nerve conduction velocities, and thermal and mechanical sensitivity. Morphometric analysis was performed by myelin sheath staining and immunohistochemistry. Macrophage markers and circulating cytokines were measured by western blot and ELISA. MicroRNA (miRNA) array and bioinformatics analyses were performed to examine the exosomal miRNA profile and miRNA putative target genes involved in DPN. RESULTS: Treatment of DPN with MSC-exosomes markedly decreased the threshold for thermal and mechanical stimuli and increased nerve conduction velocity in diabetic mice. Histopathological analysis showed that MSC-exosomes markedly augmented the density of FITC-dextran perfused blood vessels and increased the number of intraepidermal nerve fibres (IENFs), myelin thickness and axonal diameters of sciatic nerves. Western blot analysis revealed that MSC-exosome treatment decreased and increased M1 and M2 macrophage phenotype markers, respectively. Moreover, MSC-exosomes substantially suppressed proinflammatory cytokines. Bioinformatics analysis revealed that MSC-exosomes contained abundant miRNAs that target the Toll-like receptor (TLR)4/NF-κB signalling pathway. CONCLUSIONS/INTERPRETATION: MSC-derived exosomes alleviate neurovascular dysfunction and improve functional recovery in mice with DPN by suppression of proinflammatory genes.


Subject(s)
Diabetic Neuropathies/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental , Disease Models, Animal , Immunohistochemistry , Macrophages/cytology , Macrophages/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , MicroRNAs/metabolism , Sciatic Nerve/physiology , Vasa Nervorum/cytology , Vasa Nervorum/metabolism
5.
Brain Res ; 1707: 198-207, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30500399

ABSTRACT

Diabetes induces neurovascular dysfunction leading to peripheral neuropathy. MicroRNAs (miRNAs) affect many biological processes and the development of diabetic peripheral neuropathy. In the present study, we investigated whether thymosin-ß4 (Tß4) ameliorates diabetic peripheral neuropathy and whether miR-146a mediates the effect of Tß4 on improved neurovascular function. Male Type II diabetic BKS. Cg-m+/+Leprdb/J (db/db) mice at age 20 weeks were treated with Tß4 for 8 consecutive weeks, and db/db mice treated with saline were used as a control group. Compared to non-diabetic mice, diabetic mice exhibited substantially reduced miR-146a expression, and increased IL-1R-associated kinase-1 (IRAK1), tumor necrosis factor (TNFR)-associated factor 6 (TRAF6) levels and nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) activity in sciatic nerve tissues. Treatment of diabetic mice with Tß4 significantly elevated miR-146a levels and overcame the effect of diabetes on these proteins. Tß4 treatment substantially improved motor and sensory conduction velocity of the sciatic nerve, which was associated with improvements in sensory function. Tß4 treatment significantly increased intraepidermal nerve fiber density and augmented local blood flow and the density of fluorescein isothiocyanate (FITC)-dextran perfused vessels in the sciatic nerve tissue. In vitro, treatment of dorsal root ganglion (DRG) neurons and mouse dermal endothelial cells (MDEs) with Tß4 significantly increased axonal outgrowth and capillary-like tube formation, whereas blocking miR-146a attenuated Tß4-induced axonal outgrowth and capillary tube formation, respectively. Our data indicate that miR-146a may mediate Tß4-induced neurovascular remodeling in diabetic mice, by suppressing pro-inflammatory signals.


Subject(s)
Diabetic Neuropathies/therapy , MicroRNAs/genetics , Thymosin/pharmacology , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Ganglia, Spinal/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Neuronal Outgrowth/drug effects , Sciatic Nerve/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/metabolism , Thymosin/metabolism
6.
Neurosci Res ; 147: 1-8, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30326249

ABSTRACT

Angiopoietin-1 (Ang1) and its receptor Tie2 regulate vascular function. Our previous study demonstrated that thymosin beta 4 (Tß4) ameliorates neurological function of diabetic peripheral neuropathy. Mechanisms underlying the therapeutic effect of Tß4 on diabetic peripheral neuropathy have not been fully investigated. The present in vivo study investigated whether the Ang1/Tie2 signaling pathway is involved in Tß4-improved neurovascular remodeling in diabetic peripheral neuropathy. Diabetic BKS. Cg-m+/+Leprdb/J (db/db) mice at age 20 weeks were treated with Tß4 and neutralizing antibody against mouse Tie2 for 4 consecutive weeks. Neurological functional and neurovascular remodeling were measured. Administration of the neutralizing antibody against Tie2 attenuated the therapeutic effect of Tß4 on improved diabetic peripheral neuropathy as measured by motor and sensory nerve conduction velocity and thermal hypoesthesia compared to diabetic db/db mice treated with Tß4 only. Histopathological analysis revealed that the neutralizing antibody against Tie2 abolished Tß4-increased microvascular density in sciatic nerve and intraepidermal nerve fiber density, which were associated with suppression of Tß4-upregulated occludin expression and Tß4-reduced protein levels of nuclear factor-κB (NF-κB) and vascular cell adhesion molecule-1 (VCAM1). Our data provide in vivo evidence that the Ang1/Tie2 pathway contributes to the therapeutic effect of Tß4 on diabetic peripheral neuropathy.


Subject(s)
Angiopoietin-1/metabolism , Diabetic Neuropathies/metabolism , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/metabolism , Thymosin/pharmacology , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/drug therapy , Mice , Mice, Transgenic , Nerve Fibers/drug effects , Nerve Fibers/pathology , Sciatic Nerve/blood supply , Sciatic Nerve/pathology , Signal Transduction/drug effects
7.
Front Neurosci ; 12: 795, 2018.
Article in English | MEDLINE | ID: mdl-30429771

ABSTRACT

Cognition impairment and peripheral neuropathy (DPN) are two major complications of diabetes. The aim of the present study is to investigate the effect of sex differences on cognition and DPN in diabetic mice. Male and female BKS.Cg-m+/+Leprdb/J (db/db) and db/m mice were used. At ages of 20 and 30 weeks, all animals were subjected to learning, memory and neurological function tests. Regional blood flow in footpad and sciatic nerves were measured using laser Doppler flowmetry. Our data showed that male db/db mice aged 20 weeks and 30 weeks spent significantly more time to locate the hidden platform in the correct quadrant and spent significantly less time exploring the cage with a new stranger mouse compared to aged-matched female db/db mice. Electrophysiological recordings showed that male db mice aged 30 weeks had significantly reduced motor and sensory nerve conduction velocity compared with females. Hot plate and tactile allodynia tests revealed that males exhibited significantly higher thermal and mechanical latency than females. Male db mice aged 30 weeks displayed significantly reduced blood perfusion in sciatic nerve and footpad tissues compared with females. In addition, compared with male and female non-diabetic db/m mice, db/db mice exhibited increased time spent on locating the hidden platform, decreased time spent on exploring the novel odor bead and an unfamiliar mouse, as well as showed significantly lower levels of blood flow, lower velocity of MCV and SCV, higher thermal and mechanical latencies. Blood glucose levels and body weight were not significantly different between male and female diabetic animals (age 30 weeks), but male db mice showed a higher serum total cholesterol content. Together, our data suggest that males develop a greater extent of diabetes-induced cognition deficits and peripheral neurovascular dysfunction than females.

8.
FASEB J ; : fj201800597R, 2018 Jun 22.
Article in English | MEDLINE | ID: mdl-29932869

ABSTRACT

Schwann cells actively interact with axons of dorsal root ganglia (DRG) neurons. Exosomes mediate intercellular communication by transferring their biomaterials, including microRNAs (miRs) into recipient cells. We hypothesized that exosomes derived from Schwann cells stimulated by high glucose (HG) exosomes accelerate development of diabetic peripheral neuropathy and that exosomal cargo miRs contribute to this process. We found that HG exosomes contained high levels of miR-28, -31a, and -130a compared to exosomes derived from non-HG-stimulated Schwann cells. In vitro, treatment of distal axons with HG exosomes resulted in reduction of axonal growth, which was associated with elevation of miR-28, -31a, and -130a and reduction of their target proteins of DNA methyltransferase-3α, NUMB (an endocytic adaptor protein), synaptosome associated protein 25, and growth-associated protein-43 in axons. In vivo, administration of HG exosomes to sciatic nerves of diabetic db/db mice at 7 wk of age promoted occurrence of peripheral neuropathy characterized by impairment of nerve conduction velocity and induction of mechanic and thermal hypoesthesia, which was associated with substantial decreases in intraepidermal nerve fibers. Our findings demonstrate a functional role of exosomes derived from HG-stimulated Schwann cells in mediating development of diabetic peripheral neuropathy.-Jia, L., Chopp, M., Wang, L., Lu, X., Szalad, A., Zhang, Z. G. Exosomes derived from high-glucose-stimulated Schwann cells promote development of diabetic peripheral neuropathy.

9.
Mol Neurobiol ; 55(12): 9089-9099, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29637443

ABSTRACT

Hyperglycemia impairs nerve fibers of dorsal root ganglia (DRG) neurons, leading to diabetic peripheral neuropathy (DPN). However, the molecular mechanisms underlying DPN are not fully understood. Using a mouse model of type II diabetes (db/db mouse), we found that microRNA-34a (miR-34a) was over-expressed in DRG, sciatic nerve, and foot pad tissues of db/db mice. In vitro, high glucose significantly upregulated miR-34a in postnatal and adult DRG neurons, which was associated with inhibition of axonal growth. Overexpression and attenuation of miR-34a in postnatal and adult DRG neurons suppressed and promoted, respectively, axonal growth. Bioinformatic analysis suggested that miR-34a putatively targets forkhead box protein P2 (FOXP2) and vesicle amine transport 1 (VAT1), which were decreased in diabetic tissues and in cultured DRG neurons under high glucose conditions. Dual-luciferase assay showed that miR-34a downregulated FOXP2 and VAT1 expression by targeting their 3' UTR. Gain-of- and loss-of-function analysis showed an inverse relation between augmentation of miR-34a and reduction of FOXP2 and VAT1 proteins in postnatal and adult DRG neurons. Knockdown of FOXP2 and VAT1 reduced axonal growth. Together, these findings suggest that miR-34a and its target genes of FOXP2 and VAT1 are involved in DRG neuron damage under hyperglycemia.


Subject(s)
Axons/metabolism , Forkhead Transcription Factors/metabolism , Ganglia, Spinal/metabolism , MicroRNAs/metabolism , Repressor Proteins/metabolism , Vesicular Transport Proteins/metabolism , 3' Untranslated Regions/genetics , Animals , Animals, Newborn , Base Sequence , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/metabolism , Hyperglycemia/pathology , Mice, Inbred C57BL , MicroRNAs/genetics
10.
Mol Neurobiol ; 55(1): 851-858, 2018 01.
Article in English | MEDLINE | ID: mdl-28070856

ABSTRACT

Diabetes initially induces distal axonal damage of peripheral nerves, but molecular mechanisms that mediate axonal injury are not fully understood. MircoRNAs (miRNAs) regulate axonal growth. We found that diabetic db/db mice exhibited substantial upregulation of miR-29c in dorsal root ganglia (DRG) neurons, sciatic nerve, and foot pad tissues. Bioinformatic analysis revealed PRKCI, a gene that encodes a member of the protein kinase C (PKC) iota, as a putative target for miR-29c. Western blot analysis showed that diabetic mice exhibited a considerable reduction of PRKCI protein levels in sciatic nerve tissues and DRG neurons. Using dual-luciferase assay, we found that co-transfection of a plasmid containing miR-29c binding site at 3' UTR of PRKCI gene and miR-29c mimics effectively reduced luminescence activity, which was abolished when miR-29c seed sequences at 3' UTR of PRKCI gene were mutated. In vitro, high glucose substantially upregulated and reduced miR-29c and PRKCI protein levels, respectively, in DRG neurons, which were associated with significant reduction of axonal growth. Knockdown of endogenous miR-29c in DRG neurons by siRNAs overcame reduced PRKCI protein and axonal growth under high glucose condition. Moreover, knockdown of PRKCI in DRG neurons by siRNAs under regular glucose condition considerably inhibited axonal growth. Together, these findings suggest that miR-29c is a negative regulator of axonal growth of DRG neurons by targeting PRKCI under hyperglycemia.


Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , Hyperglycemia/metabolism , Isoenzymes/metabolism , MicroRNAs/metabolism , Protein Kinase C/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/genetics , Mice , MicroRNAs/genetics , Rats, Wistar , Up-Regulation/genetics
11.
Diabetes ; 66(12): 3111-3121, 2017 12.
Article in English | MEDLINE | ID: mdl-28899883

ABSTRACT

MicroRNA-146a (miR-146a) regulates multiple immune diseases. However, the role of miR-146a in diabetic peripheral neuropathy (DPN) has not been investigated. We found that mice (db/db) with type 2 diabetes exhibited substantial downregulation of miR-146a in sciatic nerve tissue. Systemic administration of miR-146a mimics to diabetic mice elevated miR-146a levels in plasma and sciatic nerve tissue and substantially increased motor and sensory nerve conduction velocities by 29 and 11%, respectively, and regional blood flow by 50% in sciatic nerve tissue. Treatment with miR-146a mimics also considerably decreased the response in db/db mice to thermal stimuli thresholds. Histopathological analysis showed that miR-146a mimics markedly augmented the density of fluorescein isothiocyanate-dextran-perfused blood vessels and increased the number of intraepidermal nerve fibers, myelin thickness, and axonal diameters of sciatic nerves. In addition, miR-146a treatment reduced and increased classically and alternatively activated macrophage phenotype markers, respectively. Analysis of miRNA target array revealed that miR-146a mimics greatly suppressed expression of many proinflammatory genes and downstream related cytokines. Collectively, our data indicate that treatment of diabetic mice with miR-146a mimics robustly reduces DPN and that suppression of hyperglycemia-induced proinflammatory genes by miR-146a mimics may underlie its therapeutic effect.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/prevention & control , MicroRNAs/physiology , Animals , Cytokines/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Macrophage Activation , Male , Mice , Myelin Sheath/physiology , NF-kappa B/physiology , Regional Blood Flow , Sciatic Nerve/blood supply , Sciatic Nerve/physiology , TNF Receptor-Associated Factor 6/genetics
12.
PLoS One ; 11(7): e0159665, 2016.
Article in English | MEDLINE | ID: mdl-27438594

ABSTRACT

We previously demonstrated that treatment of diabetic peripheral neuropathy with the short (4 hours) half-life phosphodiesterase 5 (PDE5) inhibitor, sildenafil, improved functional outcome in diabetic db/db mice. To further examine the effect of PDE5 inhibition on diabetic peripheral neuropathy, we investigated the effect of another potent PDE5 inhibitor, tadalafil, on diabetic peripheral neuropathy. Tadalafil is pharmacokinetically distinct from sildenafil and has a longer half-life (17+hours) than sildenafil. Diabetic mice (BKS.Cg-m+/+Leprdb/J, db/db) at age 20 weeks were treated with tadalafil every 48 hours for 8 consecutive weeks. Compared with diabetic mice treated with saline, tadalafil treatment significantly improved motor and sensory conduction velocities in the sciatic nerve and peripheral thermal sensitivity. Tadalafil treatment also markedly increased local blood flow and the density of FITC-dextran perfused vessels in the sciatic nerve concomitantly with increased intraepidermal nerve fiber density. Moreover, tadalafil reversed the diabetes-induced reductions of axon diameter and myelin thickness and reversed the diabetes-induced increased g-ratio in the sciatic nerve. Furthermore, tadalafil enhanced diabetes-reduced nerve growth factor (NGF) and platelet-derived growth factor-C (PDGF-C) protein levels in diabetic sciatic nerve tissue. The present study demonstrates that tadalafil increases regional blood flow in the sciatic nerve tissue, which may contribute to the improvement of peripheral nerve function and the amelioration of diabetic peripheral neuropathy.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Peripheral Nervous System Diseases/drug therapy , Sciatic Nerve/drug effects , Tadalafil/administration & dosage , Animals , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Lymphokines/biosynthesis , Lymphokines/genetics , Mice , Mice, Inbred NOD/genetics , Motor Activity/drug effects , Nerve Fibers/drug effects , Nerve Fibers/pathology , Nerve Growth Factor/biosynthesis , Nerve Growth Factor/genetics , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Sciatic Nerve/blood supply , Sciatic Nerve/physiopathology
13.
Neuroscience ; 329: 43-53, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27167084

ABSTRACT

Axonal loss contributes to induction of diabetic peripheral neuropathy. Sildenafil, a phosphodiesterase type 5 inhibitor, ameliorates neurological dysfunction in diabetic peripheral neuropathy. However, the direct effect of high glucose and sildenafil on axonal growth has not been extensively investigated. Using rat primary dorsal root ganglia (DRG) neurons cultured in a microfluidic chamber, we investigated the effect of axonal application of high glucose and sildenafil on distal axonal growth. We found that axonal, but not cell body, application of high glucose locally inhibited distal axonal growth. However, axonal application of sildenafil overcame high glucose-reduced axonal growth. Quantitative real-time RT-PCR (qRT-PCR) and Western blot analysis of distal axonal samples revealed that high glucose reduced axonal miR-146a levels and substantially increased miR-146a target genes, IRAK1 and TRAF6 in the axon. In contrast, sildenafil significantly reversed high glucose-reduced miR-146a levels and high glucose-increased IRAK1 and TRAF6. Gain- and loss-of function of miR-146a in DRG neurons revealed that miR-146a mediated the local effect of high glucose on the distal axonal growth. These in vitro data provide new insights into molecular mechanisms of diabetic peripheral neuropathy.


Subject(s)
Axons/metabolism , Diabetic Neuropathies/metabolism , Ganglia, Spinal/metabolism , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , Sildenafil Citrate/pharmacology , Animals , Argonaute Proteins/metabolism , Axons/drug effects , Axons/pathology , Cells, Cultured , DEAD-box RNA Helicases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/pathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Glucose , Interleukin-1 Receptor-Associated Kinases/metabolism , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/physiology , Phosphodiesterase 5 Inhibitors/pharmacology , Rats, Wistar , Ribonuclease III/metabolism , TNF Receptor-Associated Factor 6/metabolism
14.
J Diabetes Res ; 2015: 173656, 2015.
Article in English | MEDLINE | ID: mdl-25945352

ABSTRACT

Peripheral neuropathy is a chronic complication of diabetes mellitus. To investigated the efficacy and safety of the extended treatment of diabetic peripheral neuropathy with thymosin ß4 (Tß4), male diabetic mice (db/db) at the age of 24 weeks were treated with Tß4 or saline for 16 consecutive weeks. Treatment of diabetic mice with Tß4 significantly improved motor (MCV) and sensory (SCV) conduction velocity in the sciatic nerve and the thermal and mechanical latency. However, Tß4 treatment did not significantly alter blood glucose levels. Treatment with Tß4 significantly increased intraepidermal nerve fiber density. Furthermore, Tß4 counteracted the diabetes-induced axon diameter and myelin thickness reductions and the g-ratio increase in sciatic nerve. In vitro, compared with dorsal root ganglia (DRG) neurons derived from nondiabetic mice, DRG neurons derived from diabetic mice exhibited significantly decreased neurite outgrowth, whereas Tß4 promoted neurite growth in these diabetic DRG neurons. Blockage of the Ang1/Tie2 signaling pathway with a neutralized antibody against Tie2 abolished Tß4-increased neurite outgrowth. Our data demonstrate that extended Tß4 treatment ameliorates diabetic-induced axonal degeneration and demyelination, which likely contribute to therapeutic effect of Tß4 on diabetic neuropathy. The Ang1/Tie2 pathway may mediate Tß4-induced axonal remodeling.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/drug therapy , Thymosin/therapeutic use , Animals , Diabetes Mellitus, Type 2/blood , Diabetic Neuropathies/blood , Disease Models, Animal , Ganglia, Spinal/drug effects , Male , Mice , Nerve Fibers/drug effects , Sciatic Nerve/drug effects , Signal Transduction/drug effects , Thymosin/pharmacology , Treatment Outcome
15.
PLoS One ; 10(2): e0118134, 2015.
Article in English | MEDLINE | ID: mdl-25689401

ABSTRACT

Diabetic peripheral neuropathy is a common complication of long-standing diabetes mellitus. To mimic clinical trials in which patients with diabetes enrolled have advanced peripheral neuropathy, we investigated the effect of sildenafil, a specific inhibitor of phosphodiesterase type 5 enzyme, on long term peripheral neuropathy in middle aged male mice with type II diabetes. Treatment of diabetic mice (BKS.Cg-m+/+Leprdb/J, db/db) at age 36 weeks with sildenafil significantly increased functional blood vessels and regional blood flow in the sciatic nerve, concurrently with augmentation of intra-epidermal nerve fiber density in the skin and myelinated axons in the sciatic nerve. Functional analysis showed that the sildenafil treatment considerably improved motor and sensory conduction velocities in the sciatic nerve and peripheral thermal stimulus sensitivity compared with the saline treatment. In vitro studies showed that mouse dermal endothelial cells (MDE) cultured under high glucose levels exhibited significant down regulation of angiopoietin 1 (Ang1) expression and reduction of capillary-like tube formation, which were completely reversed by sildenafil. In addition, incubation of dorsal root ganglia (DRG) neurons with conditioned medium harvested from MDE under high glucose levels suppressed neurite outgrowth, where as conditional medium harvested from MDE treated with sildenafil under high glucose levels did not inhibit neurite outgrowth of DRG neurons. Moreover, blockage of the Ang1 receptor, Tie2, with a neutralized antibody against Tie2 abolished the beneficial effect of sildenafil on tube formation and neurite outgrowth. Collectively, our data indicate that sildenafil has a therapeutic effect on long term peripheral neuropathy of middle aged diabetic mice and that improvement of neurovascular dysfunction by sildenafil likely contributes to the amelioration of nerve function. The Ang1/Tie2 signaling pathway may play an important role in these restorative processes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy , Sildenafil Citrate/pharmacology , Angiopoietin-1/metabolism , Animals , Male , Mice , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Receptor, TIE-2/metabolism , Recovery of Function/drug effects , Signal Transduction/drug effects , Sildenafil Citrate/therapeutic use , Treatment Outcome
16.
Stroke ; 44(7): 1965-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696546

ABSTRACT

BACKGROUND AND PURPOSE: Cerebrolysin, a mixture of neurotrophic peptides, enhances neurogenesis and improves neurological outcome in experimental neurodegenerative diseases and stroke. The Sonic hedgehog (Shh) signaling pathway stimulates neurogenesis after stroke. The present study tests whether the Shh pathway mediates cerebrolysin-induced neurogenesis and improves neurological outcome after stroke. METHODS: Rats subjected to embolic stroke were treated with cerebrolysin with or without cyclopamine. RESULTS: Using neural progenitor cells derived from the subventricular zone of the lateral ventricle of adult rats, we found that cerebrolysin significantly increased neural progenitor cells proliferation and their differentiation into neurons and myelinating oligodendrocytes, which were associated with upregulation of Shh and its receptors patched and smoothened. Blockage of the Shh signaling pathway with a pharmacological smoothened inhibitor, cyclopamine, abolished cerebrolysin-induced in vitro neurogenesis and oligodendrogenesis. In the ischemic rats, treatment with cerebrolysin starting 24 hours after stroke significantly increased neural progenitor cell proliferation in the subventricular zone and enhanced neurogenesis, oligodendrogenesis, and axonal remodeling in the peri-infarct area. Moreover, profound neurological function improvements were observed in rats treated with cerebrolysin from week 3 to week 5 after stroke onset compared with vehicle-treated rats. However, in vivo inhibition of the Shh pathway with cyclopamine completely reversed the effects of cerebrolysin on neurorestoration and functional recovery. CONCLUSIONS: These results demonstrate that the Shh pathway mediates cerebrolysin-enhanced neurogenesis and white matter remodeling and improves functional recovery in rats after stroke.


Subject(s)
Amino Acids/pharmacology , Antihypertensive Agents/pharmacology , Cell Proliferation/drug effects , Hedgehog Proteins/physiology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Amino Acids/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Cell Differentiation/drug effects , Disease Models, Animal , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurogenesis/drug effects , Neurogenesis/physiology , Neuroprotective Agents/administration & dosage , Patched Receptors , Random Allocation , Rats , Rats, Wistar , Recovery of Function/physiology , Signal Transduction/drug effects
17.
Neurobiol Dis ; 48(3): 546-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22922221

ABSTRACT

Peripheral neuropathy is one of the most common complications of diabetes mellitus. Using a mouse model of diabetic peripheral neuropathy, we tested the hypothesis that thymosin ß4 (Tß4) ameliorates diabetes-induced neurovascular dysfunction in the sciatic nerve and promotes recovery of neurological function from diabetic peripheral neuropathy. Tß4 treatment of diabetic mice increased functional vascular density and regional blood flow in the sciatic nerve, and improved nerve function. Tß4 upregulated angiopoietin-1 (Ang1) expression, but suppressed Ang2 expression in endothelial and Schwann cells in the diabetic sciatic nerve. In vitro, incubation of Human Umbilical Vein Endothelial Cells (HUVECs) with Tß4 under high glucose condition completely abolished high glucose-downregulated Ang1 expression and high glucose-reduced capillary-like tube formation. Moreover, incubation of HUVECs under high glucose with conditioned medium collected from Human Schwann Cells (HSCs) treated with Tß4 significantly reversed high glucose-decreased capillary-like tube formation. PI3K/Akt signaling pathway is involved in Tß4-regulated Ang1 expression on endothelial and Schwann cells. These data indicate that Tß4 likely acts on endothelial cells and Schwann cells to preserve and/or restore vascular function in the sciatic nerve which facilitates improvement of peripheral nerve function under diabetic neuropathy. Thus, Tß4 has potential for the treatment of diabetic peripheral neuropathy.


Subject(s)
Diabetic Neuropathies/metabolism , Neuroprotective Agents/pharmacology , Sciatic Nerve/drug effects , Thymosin/pharmacology , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Electrophysiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Immunohistochemistry , Laser-Doppler Flowmetry , Mice , Real-Time Polymerase Chain Reaction , Regional Blood Flow , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/blood supply , Sciatic Nerve/metabolism , Signal Transduction/drug effects
18.
Phys Rev E Stat Nonlin Soft Matter Phys ; 83(3 Pt 1): 031920, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21517536

ABSTRACT

We consider emergent collective behavior of a multicellular biological system. Specifically, we investigate the role of hypoxia (lack of oxygen) in migration of brain tumor cells. We performed two series of cell migration experiments. In the first set of experiments, cell migration away from a tumor spheroid was investigated. The second set of experiments was performed in a typical wound-healing geometry: Cells were placed on a substrate, a scratch was made, and cell migration into the gap was investigated. Experiments show a surprising result: Cells under normal and hypoxic conditions have migrated the same distance in the "spheroid" experiment, while in the "scratch" experiment cells under normal conditions migrated much faster than under hypoxic conditions. To explain this paradox, we formulate a discrete stochastic model for cell dynamics. The theoretical model explains our experimental observations and suggests that hypoxia decreases both the motility of cells and the strength of cell-cell adhesion. The theoretical predictions were further verified in independent experiments.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Hypoxia , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Diffusion , Glioma/metabolism , Humans , Models, Biological , Mutation , Oxygen/metabolism , Stochastic Processes , Time Factors
19.
J Cereb Blood Flow Metab ; 31(2): 640-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20700128

ABSTRACT

Erythropoietin (EPO) enhances angiogenesis in the ischemic brain. Stroke induces secretion of tumor necrosis factor α (TNF-α). We investigated the effect of TNF-α on EPO-induced in vitro angiogenesis in cerebral endothelial cells. Using a capillary-like tubular formation assay, we found that transient incubation of primary rat cerebral microvascular endothelial cells (RECs) with TNF-α substantially upregulated EPO receptor (EPOR) expression and addition of EPO into TNF-α-treated RECs significantly augmented the capillary-like tube formation. Blockage of TNF receptor 1 (TNFR1) suppressed TNF-α-upregulated EPOR expression and abolished EPO-induced tube formation. Attenuation of endogenous EPOR with small interfering RNA (siRNA) also inhibited EPO-enhanced tube formation. Treatment of RECs with EPO activated nuclear factor-kappa B (NF-κB) and Akt. Incubation of the TNF-α-treated endothelial cells with EPO activated vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), angiopoietin 1 (Ang1), and Tie2. Blockage of VEGFR2 and Tie2 resulted in reduction of EPO-augmented tube formation. These data indicate that interaction of TNF-α with TNFR1 sensitizes cerebral endothelial cells for EPO-induced angiogenesis by upregulation of EPOR, which amplifies the effect of EPO on activation of the VEGF/VEGFR2 and Ang1/Tie2 pathways. Our results provide the evidence for crosslink between TNF and EPOR to coordinate the onset of angiogenesis in cerebral endothelial cells.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Endothelial Cells/drug effects , Erythropoietin/pharmacology , Neovascularization, Physiologic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Angiogenesis Inducing Agents/metabolism , Angiopoietin-1/biosynthesis , Animals , Blotting, Western , Capillaries/growth & development , Cell Proliferation/drug effects , Drug Synergism , Immunohistochemistry , Male , NF-kappa B/biosynthesis , Oncogene Protein v-akt/biosynthesis , Rats , Rats, Wistar , Receptors, Erythropoietin/drug effects , Receptors, Erythropoietin/genetics , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vascular Endothelial Growth Factor A/biosynthesis
20.
J Neurosci Res ; 88(15): 3275-81, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20857512

ABSTRACT

Cerebrolysin is a peptide preparation mimicking the action of neurotrophic factors and has beneficial effects on neurodegenerative diseases and stroke. The present study investigated the effect of Cerebrolysin on neurogenesis in a rat model of embolic middle cerebral artery occlusion (MCAo). Treatment with Cerebrolysin at doses of 2.5 and 5 ml/kg significantly increased the number of bromodeoxyuridine-positive (BrdU(+)) subventricular zone (SVZ) neural progenitor cells and doublecortin (DCX) immunoreactivity (migrating neuroblasts) in the ipsilateral SVZ and striatal ischemic boundary 28 days after stroke when the treatment was initiated 24 hr after stroke. The treatment also reduced TUNEL(+) cells by ∼50% in the ischemic boundary. However, treatment with Cerebrolysin at a dose of 2.5 ml/kg initiated at 24 and 48 hr did not significantly reduce infarct volume but substantially improved neurological outcomes measured by an array of behavioral tests 21 and 28 days after stroke. Incubation of SVZ neural progenitor cells from ischemic rats with Cerebrolysin dose dependently augmented BrdU(+) cells and increased the number of Tuj1(+) cells (a marker of immature neurons). Blockage of the PI3K/Akt pathway abolished Cerebrolysin-increased BrdU(+) cells. Moreover, Cerebrolysin treatment promoted neural progenitor cell migration. Collectively, these data indicate that Cerebrolysin treatment when initiated 24 and 48 hr after stroke enhances neurogenesis in the ischemic brain and improves functional outcome and that Cerebrolysin-augmented proliferation, differentiation, and migration of adult SVZ neural progenitor cells contribute to Cerebrolysin-induced neurogenesis, which may be related to improvement of neurological outcome. The PI3K/Akt pathway mediates Cerebrolysin-induced progenitor cell proliferation.


Subject(s)
Amino Acids/pharmacology , Brain Ischemia/drug therapy , Neurogenesis/drug effects , Recovery of Function/drug effects , Stroke/drug therapy , Animals , Blotting, Western , Brain Ischemia/pathology , Cell Differentiation/drug effects , Doublecortin Protein , Immunohistochemistry , Male , Neurogenesis/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Stroke/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...