Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-28775932

ABSTRACT

BACKGROUND: Mitochondrial dysfunction is purported as a contributory mechanism underlying diabetic neuropathy, but a defined role for damaged mitochondria in diabetic nerves remains unclear, particularly in standard diabetes models. Experiments here used a high-fat diet in attempt to exacerbate the severity of diabetes and expedite the time-course in which mitochondrial dysfunction may occur. We hypothesized a high-fat diet in addition to diabetes would increase stress on sensory neurons and worsen mitochondrial dysfunction. METHODS: Oxidative phosphorylation proteins and proteins associated with mitochondrial function were quantified in lumbar dorsal root ganglia. Comparisons were made between non-diabetic and streptozotocin-induced (STZ) C57Bl/6 mice fed a standard or high-fat diet for 8 weeks. RESULTS: Complex III subunit Core-2 and voltage dependent anion channel were increased (by 36% and 28% respectively, p<0.05) in diabetic mice compared to nondiabetic mice fed the standard diet. There were no differences among groups in UCP2, PGC-1α, PGC-1ß levels or Akt, mTor, or AMPK activation. These data suggest compensatory mitochondrial biogenesis occurs to offset potential mitochondrial dysfunction after 8 weeks of STZ-induced diabetes, but a high-fat diet does not alter these parameters. CONCLUSION: Our results indicate mitochondrial protein changes early in STZ-induced diabetes. Interestingly, a high-fat diet does not appear to affect mitochondrial proteins in either nondiabetic or STZ- diabetic mice.

2.
Neuroscience ; 285: 312-23, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25451280

ABSTRACT

Diabetic peripheral neuropathy is a common complication of diabetes mellitus, and a significant proportion of individuals suffer debilitating pain that significantly affects their quality of life. Unfortunately, symptomatic treatment options have limited efficacy, and often carry significant risk of systemic adverse effects. Activation of the adenosine A1 receptor (A1R) by the analgesic small molecule adenosine has been shown to have antinociceptive benefits in models of inflammatory and neuropathic pain. The current study used a mouse model of painful diabetic neuropathy to determine the effect of diabetes on endogenous adenosine production, and if central or peripheral delivery of adenosine receptor agonists could alleviate signs of mechanical allodynia in diabetic mice. Diabetes was induced using streptozocin in male A/J mice. Mechanical withdrawal thresholds were measured weekly to characterize neuropathy phenotype. Hydrolysis of AMP into adenosine by ectonucleotidases was determined in the dorsal root ganglia (DRG) and spinal cord at 8 weeks post-induction of diabetes. AMP, adenosine and the specific A1R agonist, N(6)-cyclopentyladenosine (CPA), were administered both centrally (intrathecal) and peripherally (intraplantar) to determine the effect of activation of adenosine receptors on mechanical allodynia in diabetic mice. Eight weeks post-induction, diabetic mice displayed significantly decreased hydrolysis of extracellular AMP in the DRG; at this same time, diabetic mice displayed significantly decreased mechanical withdrawal thresholds compared to nondiabetic controls. Central delivery AMP, adenosine and CPA significantly improved mechanical withdrawal thresholds in diabetic mice. Surprisingly, peripheral delivery of CPA also improved mechanical allodynia in diabetic mice. This study provides new evidence that diabetes significantly affects endogenous AMP hydrolysis, suggesting that altered adenosine production could contribute to the development of painful diabetic neuropathy. Moreover, central and peripheral activation of A1R significantly improved mechanical sensitivity, warranting further investigation into this important antinociceptive pathway as a novel therapeutic option for the treatment of painful diabetic neuropathy.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Analgesics, Non-Narcotic/administration & dosage , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/drug therapy , Hyperalgesia/drug therapy , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Foot , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Hindlimb , Hydrolysis/drug effects , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Injections, Spinal , Male , Mice , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Receptor, Adenosine A1/metabolism , Spinal Cord/drug effects , Spinal Cord/physiopathology , Touch
3.
Neuroscience ; 263: 216-30, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24462609

ABSTRACT

Early life stress can permanently alter functioning of the hypothalamic-pituitary-adrenal (HPA) axis, which regulates the stress response and influences the perception of pain. Chronic pelvic pain patients commonly report having experienced childhood neglect or abuse, which increases the likelihood of presenting with comorbid chronic pain and/or mood disorders. Animal models of neonatal stress commonly display enhanced anxiety-like behaviors, colorectal hypersensitivity, and disruption of proper neuro-immune interactions in adulthood. Here, we tested the hypothesis that early life stress impacts vaginal sensitivity by exposing mice to neonatal maternal separation (NMS) for 3h/day during the first two (NMS14) or three (NMS21) postnatal weeks. As adults, female mice underwent vaginal balloon distension (VBD), which was also considered an acute stress. Before or after VBD, mice were assessed for anxiety-like behavior, hindpaw sensitivity, and changes in gene and protein expression related to HPA axis function and regulation. NMS21 mice displayed significantly increased vaginal sensitivity compared to naïve mice, as well as significantly reduced anxiety-like behavior at baseline, which was heightened following VBD. NMS21 mice exhibited significant thermal and mechanical hindpaw hypersensitivity at baseline and following VBD. NMS14 mice displayed no change in anxiety-like behavior and only exhibited significantly increased hindpaw mechanical and thermal sensitivity following VBD. Centrally, a significant decrease in negative regulation of the HPA axis was observed in the hypothalamus and hippocampus of NMS21 mice. Peripherally, NMS and VBD affected the expression of inflammatory mediators in the vagina and bladder. Corticotropin-releasing factor (CRF) receptor and transient receptor potential (TRP) channel protein expression was also significantly, and differentially, affected in vagina, bladder, and colon by both NMS and VBD. Together these data indicate that NMS affects both central and peripheral aspects of the HPA axis, which may drive changes in vaginal sensitivity and the development of comorbid chronic pain and mood disorders.


Subject(s)
Anxiety, Separation/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Psychological/physiopathology , Vagina/physiopathology , Animals , Animals, Newborn , Female , Gene Expression , Mice , Mice, Inbred C57BL
4.
Exp Neurol ; 234(1): 62-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22201551

ABSTRACT

Diabetic neuropathy is a common complication of diabetes mellitus with over half of all patients developing neuropathy symptoms due to sensory nerve damage. Diabetes-induced hyperglycemia leads to the accelerated production of advanced glycation end products (AGEs) that alter proteins, thereby leading to neuronal dysfunction. The glyoxalase enzyme system, specifically glyoxalase I (GLO1), is responsible for detoxifying precursors of AGEs, such as methylglyoxal and other reactive dicarbonyls. The purpose of our studies was to determine if expression differences of GLO1 may play a role in the development of diabetic sensory neuropathy. BALB/cJ mice naturally express low levels of GLO1, while BALB/cByJ express approximately 10-fold higher levels on a similar genetic background due to increased copy numbers of GLO1. Five weeks following STZ injection, diabetic BALB/cJ mice developed a 68% increase in mechanical thresholds, characteristic of insensate neuropathy or loss of mechanical sensitivity. This behavior change correlated with a 38% reduction in intraepidermal nerve fiber density (IENFD). Diabetic BALB/cJ mice also had reduced expression of mitochondrial oxidative phosphorylation proteins in Complexes I and V by 83% and 47%, respectively. Conversely, diabetic BALB/cByJ mice did not develop signs of neuropathy, changes in IENFD, or alterations in mitochondrial protein expression. Reduced expression of GLO1 paired with diabetes-induced hyperglycemia may lead to neuronal mitochondrial damage and symptoms of diabetic neuropathy. Therefore, AGEs, the glyoxalase system, and mitochondrial dysfunction may play a role in the development and modulation of diabetic peripheral neuropathy.


Subject(s)
Diabetic Neuropathies/enzymology , Gene Expression Regulation, Enzymologic/physiology , Lactoylglutathione Lyase/metabolism , Analysis of Variance , Animals , Blood Glucose , Body Weight , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/complications , Diabetic Neuropathies/genetics , Disease Models, Animal , Gene Expression Regulation, Enzymologic/genetics , Lactoylglutathione Lyase/genetics , Male , Mice , Mice, Inbred BALB C , Mitochondrial Diseases/etiology , Multienzyme Complexes/metabolism , Nerve Fibers/pathology , Neural Conduction/physiology , Pain Measurement , Psychomotor Performance , Streptozocin/adverse effects
5.
Exp Diabetes Res ; 2011: 848307, 2011.
Article in English | MEDLINE | ID: mdl-22144990

ABSTRACT

Emerging evidence suggests that dyslipidemia is an independent risk factor for diabetic neuropathy (DN) (reviewed by Vincent et al. 2009). To experimentally determine how dyslipidemia alters DN, we quantified neuropathic symptoms in diabetic mice fed a high-fat diet. Streptozotocin-induced diabetic C57BL/6 mice fed a high-fat diet developed dyslipidemia and a painful neuropathy (mechanical allodynia) instead of the insensate neuropathy (mechanical insensitivity) that normally develops in this strain. Nondiabetic mice fed a high-fat diet also developed dyslipidemia and mechanical allodynia. Thermal sensitivity was significantly reduced in diabetic compared to nondiabetic mice, but was not worsened by the high-fat diet. Moreover, diabetic mice fed a high-fat diet had significantly slower sensory and motor nerve conduction velocities compared to nondiabetic mice. Overall, dyslipidemia resulting from a high-fat diet may modify DN phenotypes and/or increase risk for developing DN. These results provide new insight as to how dyslipidemia may alter the development and phenotype of diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/etiology , Diet, High-Fat/adverse effects , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/physiopathology , Dyslipidemias/complications , Dyslipidemias/physiopathology , Male , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Neural Conduction , Oxidative Stress , Phenotype , Risk Factors , Sensory Receptor Cells/physiology , Skin/innervation
6.
Exp Diabetes Res ; 2011: 212571, 2011.
Article in English | MEDLINE | ID: mdl-21754917

ABSTRACT

Insulin signaling depends on tyrosine phosphorylation of insulin receptor substrates (IRSs) to mediate downstream effects; however, elevated serine phosphorylation of IRS impairs insulin signaling. Here, we investigated IRS protein expression patterns in dorsal root ganglia (DRG) of mice and whether their signaling was affected by diabetes. Both IRS1 and IRS2 are expressed in DRG; however, IRS2 appears to be the prevalent isoform and is expressed by many DRG neuronal subtypes. Phosphorylation of Ser(731)IRS2 was significantly elevated in DRG neurons from type 1 and type 2 diabetic mice. Additionally, Akt activation and neurite outgrowth in response to insulin were significantly decreased in DRG cultures from diabetic ob/ob mice. These results suggest that DRG neurons express IRS proteins that are altered by diabetes similar to other peripheral tissues, and insulin signaling downstream of the insulin receptor may be impaired in sensory neurons and contribute to the pathogenesis of diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/physiopathology , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Neurons/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Ganglia, Spinal/metabolism , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/genetics , Leptin/genetics , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/drug effects , Obesity/genetics , Obesity/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Streptozocin/adverse effects
7.
Diabetologia ; 54(8): 2174-82, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21633909

ABSTRACT

AIMS/HYPOTHESIS: Diabetic peripheral neuropathy (DN) is a common complication of diabetes; however, the mechanisms producing positive or negative symptoms are not well understood. The enzyme glyoxalase I (GLO1) detoxifies reactive dicarbonyls that form AGEs and may affect the way sensory neurons respond to heightened AGE levels in DN. We hypothesised that differential GLO1 levels in sensory neurons may lead to differences in AGE formation and modulate the phenotype of DN. METHODS: Inbred strains of mice were used to assess the variability of Glo1 expression by quantitative RT-PCR. Non-diabetic C57BL/6 mice were used to characterise the distribution of GLO1 in neural tissues by immunofluorescence. Behavioural assessments were conducted in diabetic A/J and C57BL/6 mice to determine mechanical sensitivity, and GLO1 abundance was determined by western blot. RESULTS: GLO1 immunoreactivity was found throughout the nervous system, but selectively in small, unmyelinated peptidergic dorsal root ganglia (DRG) neurons that are involved in pain transmission. GLO1 protein was present at various levels in DRG from different inbred mice strains. Diabetic A/J and C57BL/6 mice, two mouse strains with different levels of GLO1, displayed dramatically different behavioural responses to mechanical stimuli. Diabetic C57BL/6 mice also had a reduced abundance of GLO1 following diabetes induction. CONCLUSIONS/INTERPRETATION: These findings reveal that the abundance of GLO1 varies between different murine strains and within different sensory neuron populations. These differences could lead to different responses of sensory neurons to the toxic effects of hyperglycaemia and reactive dicarbonyls associated with diabetes.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetic Neuropathies/enzymology , Lactoylglutathione Lyase/metabolism , Animals , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Immunohistochemistry , Lactoylglutathione Lyase/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peripheral Nervous System/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Neuroscience ; 145(1): 303-13, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17223273

ABSTRACT

Human diabetic patients often lose touch and vibratory sensations, but to date, most studies on diabetes-induced sensory nerve degeneration have focused on epidermal C-fibers. Here, we explored the effects of diabetes on cutaneous myelinated fibers in relation to the behavioral responses to tactile stimuli from diabetic mice. Weekly behavioral testing began prior to streptozotocin (STZ) administration and continued until 8 weeks, at which time myelinated fiber innervation was examined in the footpad by immunohistochemistry using antiserum to neurofilament heavy chain (NF-H) and myelin basic protein (MBP). Diabetic mice developed reduced behavioral responses to non-noxious (monofilaments) and noxious (pinprick) stimuli. In addition, diabetic mice displayed a 50% reduction in NF-H-positive myelinated innervation of the dermal footpad compared with non-diabetic mice. To test whether two neurotrophins nerve growth factor (NGF) and/or neurotrophin-3 (NT-3) known to support myelinated cutaneous fibers could influence myelinated innervation, diabetic mice were treated intrathecally for 2 weeks with NGF, NT-3, NGF and NT-3. Neurotrophin-treated mice were then compared with diabetic mice treated with insulin for 2 weeks. NGF and insulin treatment both increased paw withdrawal to mechanical stimulation in diabetic mice, whereas NT-3 or a combination of NGF and NT-3 failed to alter paw withdrawal responses. Surprisingly, all treatments significantly increased myelinated innervation compared with control-treated diabetic mice, demonstrating that myelinated cutaneous fibers damaged by hyperglycemia respond to intrathecal administration of neurotrophins. Moreover, NT-3 treatment increased epidermal Merkel cell numbers associated with nerve fibers, consistent with increased numbers of NT-3-responsive slowly adapting A-fibers. These studies suggest that myelinated fiber loss may contribute as significantly as unmyelinated epidermal loss in diabetic neuropathy, and the contradiction between neurotrophin-induced increases in dermal innervation and behavior emphasizes the need for multiple approaches to accurately assess sensory improvements in diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Nerve Fibers, Myelinated/drug effects , Nerve Growth Factors/administration & dosage , Sensation Disorders/drug therapy , Sensation Disorders/etiology , Skin/innervation , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Experimental/pathology , Dose-Response Relationship, Drug , Drug Interactions , Immunohistochemistry/methods , Insulin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Neurofilament Proteins/metabolism , Pain Measurement/methods , Physical Stimulation/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...