Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Handb Clin Neurol ; 126: 429-42, 2014.
Article in English | MEDLINE | ID: mdl-25410237

ABSTRACT

Diabetic neuropathy is the most common complication of diabetes. It shows a progressive development with sensory loss, pain and autonomic dysfunction as common symptoms. Pathologically it is characterized by a series of interrelated metabolic abnormalities with insulin deficiency and hyperglycemia as the initiating culprits. The neuropathy accompanying type 2DM (insulin resistance) and type 1DM (insulin deficiency) appears to differ as to their structural changes; the former showing a milder axonal involvement and segmental myelin breakdown, whereas the latter shows a more severe axonal atrophy and axonal loss. Based mainly on animal data we will describe the sequential neuropathologic changes and differences in the two types of diabetes. These differences are related to differences in a myriad of underlying sequential metabolic abnormalities, which will be dealt with in detail. How metabolic defects affect nerve function will be elaborated upon. The disorder does not only involve somatic peripheral nerves but also autonomic and central nerve tracts. Today no successful therapy exists for diabetic neuropathy. During the last 30 years several experimental drugs targeting the polyol-pathway and oxidative stress have been tested, but with limited or no success. Instead therapies targeting the initiating and overriding pathogenetic abnormalities, such as insulin-deficiency and hyperglycemia need to be employed. One such agent is the insulinomimetic C-peptide which has demonstrated significant therapeutic and preventive effects in type 1 diabetic patients. Not surprisingly this has been particularly successful following early intervention. However diabetic neuropathy still remains a major medical problem affecting millions of patients.


Subject(s)
Axons/pathology , Diabetes Mellitus/diagnosis , Diabetic Neuropathies/diagnosis , Animals , Axons/metabolism , Diabetes Mellitus/epidemiology , Diabetes Mellitus/metabolism , Diabetic Neuropathies/epidemiology , Diabetic Neuropathies/metabolism , Humans , Insulin/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress/physiology
3.
Brain ; 134(Pt 11): 3222-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21926103

ABSTRACT

Diabetic neuropathy is a common complication of diabetes. While multiple pathways are implicated in the pathophysiology of diabetic neuropathy, there are no specific treatments and no means to predict diabetic neuropathy onset or progression. Here, we identify gene expression signatures related to diabetic neuropathy and develop computational classification models of diabetic neuropathy progression. Microarray experiments were performed on 50 samples of human sural nerves collected during a 52-week clinical trial. A series of bioinformatics analyses identified differentially expressed genes and their networks and biological pathways potentially responsible for the progression of diabetic neuropathy. We identified 532 differentially expressed genes between patient samples with progressing or non-progressing diabetic neuropathy, and found these were functionally enriched in pathways involving inflammatory responses and lipid metabolism. A literature-derived co-citation network of the differentially expressed genes revealed gene subnetworks centred on apolipoprotein E, jun, leptin, serpin peptidase inhibitor E type 1 and peroxisome proliferator-activated receptor gamma. The differentially expressed genes were used to classify a test set of patients with regard to diabetic neuropathy progression. Ridge regression models containing 14 differentially expressed genes correctly classified the progression status of 92% of patients (P < 0.001). To our knowledge, this is the first study to identify transcriptional changes associated with diabetic neuropathy progression in human sural nerve biopsies and describe their potential utility in classifying diabetic neuropathy. Our results identifying the unique gene signature of patients with progressive diabetic neuropathy will facilitate the development of new mechanism-based diagnostics and therapies.


Subject(s)
Diabetic Neuropathies/genetics , Disease Progression , Sural Nerve/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Down-Regulation , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Sural Nerve/physiopathology , Up-Regulation
4.
Acta Diabetol ; 47(4): 279-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20798963

ABSTRACT

Diabetic encephalopathies are now accepted complications of diabetes. They appear to differ in type 1 and type 2 diabetes as to underlying mechanisms and the nature of resulting cognitive deficits. The increased incidence of Alzheimer's disease in type 2 diabetes is associated with insulin resistance, hyperinsulinemia and hyperglycemia, and commonly accompanying attributes such as hypercholesterolemia, hypertension and obesity. The relevance of these disorders as to the emergence of dementia and Alzheimer's disease is discussed based on epidemiological studies. The pathobiology of accumulation of ß-amyloid and tau the hallmarks of Alzheimer's disease are discussed based on experimental data. Type 1 diabetic encephalopathy is likely to increase as a result of the global increase in the incidence of type 1 diabetes and its occurrence in increasingly younger patients. Alzheimer-like changes and dementia are not prominently increased in type 1 diabetes. Instead, the type 1 diabetic encephalopathy involves learning abilities, intelligence development and memory retrieval resulting in impaired school and professional performances. The major underlying component here appears to be insulin deficiency with downstream effects on the expression of neurotrophic factors, neurotransmitters, oxidative and apoptotic stressors resulting in defects in neuronal integrity, connectivity and loss commonly occurring in the still developing brain. Recent experimental data emphasize the role of impaired central insulin action and provide information as to potential therapies. Therefore, the underlying mechanisms resulting in diabetic encephalopathies are complex and appear to differ between the two types of diabetes. Major headway has been made in our understanding of their pathobiology; however, many questions remain to be clarified. In view of the increasing incidence of both type 1 and type 2 diabetes, intensified investigations are called for to expand our understanding of these complications and to find therapeutic means by which these disastrous consequences can be prevented and modified.


Subject(s)
Brain Diseases, Metabolic/etiology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Amyloid/metabolism , Animals , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/epidemiology , Diabetes Complications/diagnosis , Diabetes Complications/epidemiology , Diabetes Complications/psychology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/psychology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/psychology , Disease Models, Animal , Humans , Models, Biological , tau Proteins/metabolism
5.
Brain ; 133(Pt 5): 1391-402, 2010 May.
Article in English | MEDLINE | ID: mdl-20395263

ABSTRACT

We have studied a family with severe mental retardation characterized by the virtual absence of speech, autism spectrum disorder, epilepsy, late-onset ataxia, weakness and dystonia. Post-mortem examination of two males revealed widespread neuronal loss, with the most striking finding being neuronal and glial tau deposition in a pattern reminiscent of corticobasal degeneration. Electron microscopic examination of isolated tau filaments demonstrated paired helical filaments and ribbon-like structures. Biochemical studies of tau demonstrated a preponderance of 4R tau isoforms. The phenotype was linked to Xq26.3, and further analysis identified an in-frame 9 base pair deletion in the solute carrier family 9, isoform A6 (SLC9A6 gene), which encodes sodium/hydrogen exchanger-6 localized to endosomal vesicles. Sodium/hydrogen exchanger-6 is thought to participate in the targeting of intracellular vesicles and may be involved in recycling synaptic vesicles. The striking tau deposition in our subjects reveals a probable interaction between sodium/proton exchangers and cytoskeletal elements involved in vesicular transport, and raises the possibility that abnormalities of vesicular targeting may play an important role in more common disorders such as Alzheimer's disease and autism spectrum disorders.


Subject(s)
Intellectual Disability/genetics , Intellectual Disability/metabolism , Mutation , Sodium-Hydrogen Exchangers/genetics , tau Proteins/metabolism , Brain/metabolism , Brain/pathology , Child, Preschool , Gene Deletion , Genetic Linkage , Humans , Intellectual Disability/diagnosis , Intellectual Disability/psychology , Male , Microscopy, Electron , Molecular Biology/methods , Neuroglia/metabolism , Neurons/metabolism , Pedigree , Phenotype , Protein Isoforms/metabolism , Severity of Illness Index , tau Proteins/ultrastructure
6.
Brain Res ; 1343: 168-77, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20420811

ABSTRACT

Gray and white matter structural deficits may accompany type 1 diabetes. Earlier experimental studies have demonstrated neuronal deficits associated with impaired neurotrophic support, inflammation and oxidative stress. In this study we demonstrate in two patients with histories of poorly controlled type 1 diabetes and fatal brain edema of ketoacidosis neuronal deficits associated with a decreased presence of insulin and IGF-1 receptors and accumulation of nitrotyrosin in neurons of affected areas and the choroid plexus. The findings add support to the suggested genesis of T1DM encephalopathy due to compromised neurotrophic protection, oxidative stress, inflammation and neuronal deficits, as demonstrated in T1DM encephalopathy in the BB/Wor-rat.


Subject(s)
Brain Edema/complications , Brain Edema/pathology , Diabetes Mellitus, Type 1/pathology , Diabetic Ketoacidosis/complications , Diabetic Ketoacidosis/pathology , Insulin/deficiency , Receptor, IGF Type 1/deficiency , Tyrosine/analogs & derivatives , Adolescent , Biomarkers/metabolism , Brain Edema/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetic Ketoacidosis/metabolism , Disease Progression , Fatal Outcome , Female , Humans , Insulin/biosynthesis , Receptor, IGF Type 1/biosynthesis , Tyrosine/biosynthesis , Tyrosine/deficiency
8.
Rev Diabet Stud ; 6(3): 187-202, 2009.
Article in English | MEDLINE | ID: mdl-20039008

ABSTRACT

Diabetic polyneuropathy (DPN) is a common complication in diabetes. At present, there is no adequate treatment, and DPN is often debilitating for patients. It is a heterogeneous disorder and differs in type 1 and type 2 diabetes. An important underlying factor in type 1 DPN is insulin deficiency. Proinsulin C-peptide is a critical element in the cascade of events. In this review, we describe the physiological role of C-peptide and how it provides an insulin-like signaling function. Such effects translate into beneficial outcomes in early metabolic perturbations of neural Na+/K+-ATPase and nitric oxide (NO) with subsequent preventive effects on early nerve dysfunction. Further corrective consequences resulting from this signaling cascade have beneficial effects on gene regulation of early gene responses, neurotrophic factors, their receptors, and the insulin receptor itself. This may lead to preventive and corrective results to nerve fiber degeneration and loss, as well as, promotion of nerve fiber regeneration with respect to sensory somatic fibers and small nociceptive nerve fibers. A characteristic abnormality of type 1 DPN is nodal and paranodal degeneration with severe consequences for myelinated fiber function. This review deals in detail with the underlying insulin-deficiency-related molecular changes and their correction by C-peptide. Based on these observations, it is evident that continuous maintenance of insulin-like actions by C-peptide is needed in peripheral nerve to minimize the sequences of metabolic and molecular abnormalities, thereby ameliorating neuropathic complications. There is now ample evidence demonstrating that C-peptide replacement in type 1 diabetes promotes insulin action and signaling activities in a more enhanced, prolonged, and continuous fashion than does insulin alone. It is therefore necessary to replace C-peptide to physiological levels in diabetic patients. This will have substantial beneficial effects on type 1 DPN.

10.
Rev Diabet Stud ; 6(3): 211-22, 2009.
Article in English | MEDLINE | ID: mdl-20039010

ABSTRACT

Diabetic encephalopathy is a recently recognized complication in type 1 diabetes. In this review, we summarize a series of experimental results obtained longitudinally in the spontaneously type 1 diabetic BB/Wor-rat, and bringing out the beneficial effects of C-peptide replacement. It is increasingly clear that lack of insulin and C-peptide, and perturbations of their signaling cascades in type 1 diabetes are detrimental to the regulation of neurotrophic factors and their receptors. Other consequences of such deficits and perturbations are innate inflammatory responses with effects on synaptogenesis, neurite degeneration, and early behavioral abnormalities. Replacement of C-peptide, which does not effect hyperglycemia, has beneficial effects on a variety of pro-apoptotic stressors, oxidative stressors, and finally on apoptosis. Eventually, this cascade of events leads to neuronal loss and decreased densities of white matter myelinating cells, with more profound deficits in behavioral and cognitive function. Such changes are likely to underlie gray and white matter atrophy in type 1 diabetes, and are significantly prevented by full C-peptide replacement. Present data demonstrate that C-peptide replacement has beneficial effects on numerous sequential and partly interrelated pathogenetic mechanisms, resulting in prevention of neuronal and oligodendroglial cell loss, with significant prevention of neurobehavioral and cognitive functions.

11.
Exp Diabetes Res ; 2009: 793281, 2009.
Article in English | MEDLINE | ID: mdl-19834568

ABSTRACT

We investigated mechanisms underlying progressive axonal dysfunction and structural deficits in type 1 BB/Wor-rats from 1 week to 10 month diabetes duration. Motor and sensory conduction velocities were decreased after 4 and 6 weeks of diabetes and declined further over the remaining 9 months. Myelinated sural nerve fibers showed progressive deficits in fiber numbers and sizes. Structural deficits in unmyelinated axonal size were evident at 2 month and deficits in number were present at 4 mo. These changes were preceded by decreased availability of insulin, C-peptide and IGF-1 and decreased expression of neurofilaments and beta-III-tubulin. Upregulation of phosphorylating stress kinases like Cdk5, p-GSK-3beta, and p42/44 resulted in increased phosphorylation of neurofilaments. Increasing activity of p-GSK-3beta correlated with increasing phosphorylation of NFH, whereas decreasing Cdk5 correlated with diminishing phosphorylation of NFM. The data suggest that impaired neurotrophic support results in sequentially impaired synthesis and postranslational modifications of neuroskeletal proteins, resulting in progressive deficits in axonal function, maturation and size.


Subject(s)
Axons/physiology , Diabetes Mellitus, Type 1/complications , Ganglia, Spinal/metabolism , Motor Neurons/metabolism , Nerve Degeneration/pathology , Animals , Axons/metabolism , Axons/pathology , Cyclin-Dependent Kinase 5/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Ganglia, Spinal/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Motor Neurons/pathology , Nerve Degeneration/complications , Nerve Degeneration/physiopathology , Nerve Fibers, Myelinated/metabolism , Neural Conduction/physiology , Physical Stimulation , Prediabetic State/metabolism , Prediabetic State/pathology , Rats , Temperature
12.
Mol Biosyst ; 5(10): 1157-62, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19756305

ABSTRACT

Insulin resistance can broadly be defined as the diminished ability of cells to respond to the action of insulin in transporting glucose from the bloodstream into cells and tissues. Here, we report that erythrocytes (ERYs) obtained from type 2 diabetic rats display an apparent resistance to Zn(2+)-activated C-peptide. Thus, the aims of this study were to demonstrate that Zn(2+)-activated C-peptide exerts potentially beneficial effects on healthy ERYs and that these same effects on type 2 diabetic ERYs are enhanced in the presence of metformin. Incubation of ERYs (obtained from type 2 diabetic BBZDR/Wor-rats) with Zn(2+)-activated C-peptide followed by chemiluminescence measurements of ATP resulted in a 31.2 +/- 4.0% increase in ATP release from these ERYs compared to a 78.4 +/- 4.9% increase from control ERYs. Glucose accumulation in diabetic ERYs, measured by scintillation counting of (14)C-labeled glucose, increased by 35.8 +/- 1.3% in the presence of the Zn(2+)-activated C-peptide, a value significantly lower than results obtained from control ERYs (64.3 +/- 5.1%). When Zn(2+)-activated C-peptide was exogenously added to diabetic ERYs, immunoassays revealed a 32.5 +/- 8.2% increase in C-peptide absorbance compared to a 64.4 +/- 10.3% increase in control ERYs. Phosphatidylserine (PS) externalization and metformin sensitization of Zn(2+)-activated C-peptide were examined spectrofluorometrically by measuring the binding of FITC-labeled annexin to PS. The incubation of diabetic ERYs with metformin prior to the addition of Zn(2+)-activated C-peptide resulted in values that were statistically equivalent to those of controls. Summarily, data obtained here demonstrate an apparent resistance to Zn(2+)-activated C-peptide by the ERY that is corrected by metformin.


Subject(s)
C-Peptide/metabolism , Diabetes Mellitus, Type 2/pathology , Erythrocytes/metabolism , Hyperglycemia/complications , Metformin/pharmacology , Phosphatidylserines/metabolism , Zinc/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antibodies , Diabetes Mellitus, Type 2/complications , Erythrocytes/drug effects , Exocytosis/drug effects , Glucose/metabolism , Humans , Hyperglycemia/pathology , Rats , Spectrometry, Mass, Electrospray Ionization
13.
Rev Diabet Stud ; 6(1): 37-42, 2009.
Article in English | MEDLINE | ID: mdl-19557294

ABSTRACT

Encephalopathy is an increasingly recognized complication of type 1 diabetes. The underlying mechanisms are not well understood, although insulin deficiency has been implicated. The spontaneously diabetic BB/Wor-rat develops neuro-behavioral deficits and neuronal cell death in hippocampus and frontal cortex, which can be prevented by insulinomimetic C-peptide. Here we examined whether contributing factors such as activation of innate immune mediators are responsive to C-peptide replacement. Seven-month diabetic BB/Wor-rats and those treated with full C-peptide replacement were compared to age-matched control rats. Hippocampi of diabetic rats showed upregulation of RAGE and NF-kappaB, the former being localized to proliferating astrocytes. These changes were associated with increased expression of TNF-alpha, IL-1beta, IL-2 and IL-6 in hippocampi of diabetic rats. Full C-peptide replacement, which did not induce hyperglycemia, resulted in significant prevention of upregulation of RAGE expression, activation of NF-kappaB and activation of pro-inflammatory factors. In conclusion, impaired insulin activity is associated with upregulation of RAGE and pro-inflammatory factors, and these are likely to contribute to previously described oxidative and apoptotic neuronal cell death. Replacement of insulinomimetic C-peptide significantly prevents this cascade of events.

14.
Acta Neuropathol ; 118(4): 531-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19562355

ABSTRACT

Mutations affecting proteolipid protein 1 (PLP1), the major protein in central nervous system myelin, cause the X-linked leukodystrophy Pelizaeus-Merzbacher disease (PMD). We describe the neuropathologic findings in a series of eight male PMD subjects with confirmed PLP1 mutations, including duplications, complete gene deletion, missense and exon-skipping. While PLP1 mutations have effects on oligodendrocytes that result in mutation-specific degrees of dysmyelination, our findings indicate that there are also unexpected effects in the central nervous system resulting in neuronal loss. Although length-dependent axonal degeneration has been described in PLP1 null mutations, there have been no reports on neuronal degeneration in PMD patients. We now demonstrate widespread neuronal loss in PMD. The patterns of neuronal loss appear to be dependent on the mutation type, suggesting selective vulnerability of neuronal populations that depends on the nature of the PLP1 disturbance. Nigral neurons, which were not affected in patients with either null or severe misfolding mutations, and thalamic neurons appear particularly vulnerable in PLP1 duplication and deletion patients, while hippocampal neuronal loss was prominent in a patient with complete PLP1 gene deletion. All subjects showed cerebellar neuronal loss. The patterns of neuronal involvement may explain some clinical findings, such as ataxia, being more prominent in PMD than in other leukodystrophies. While the precise pathogenetic mechanisms are not known, these observations suggest that defective glial functions contribute to neuronal pathology.


Subject(s)
Brain/pathology , Cell Death/genetics , Myelin Proteolipid Protein/genetics , Neurons/pathology , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/pathology , Adult , Age Factors , Chromosomes, Human, X , Genetic Markers , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Myelin Sheath/genetics , Myelin Sheath/pathology , Neuroglia/pathology
15.
Diabetes ; 58(7): 1634-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19411614

ABSTRACT

OBJECTIVE: To evaluate mechanisms underlying diabetic neuropathy progression using indexes of sural nerve morphometry obtained from two identical randomized, placebo-controlled clinical trials. RESEARCH DESIGN AND METHODS: Sural nerve myelinated fiber density (MFD), nerve conduction velocities (NCVs), vibration perception thresholds, clinical symptom scores, and a visual analog scale for pain were analyzed in participants with diabetic neuropathy. A loss of > or =500 fibers/mm(2) in sural nerve MFD over 52 weeks was defined as progressing diabetic neuropathy, and a MFD loss of < or =100 fibers/mm(2) during the same time interval as nonprogressing diabetic neuropathy. The progressing and nonprogressing cohorts were matched for baseline characteristics using an O'Brien rank-sum and baseline MFD. RESULTS: At 52 weeks, the progressing cohort demonstrated a 25% decrease (P < 0.0001) from baseline in MFD, while the nonprogressing cohort remained unchanged. MFD was not affected by active drug treatment (P = 0.87), diabetes duration (P = 0.48), age (P = 0.11), or BMI (P = 0.30). Among all variables tested, elevated triglycerides and decreased peroneal motor NCV at baseline significantly correlated with loss of MFD at 52 weeks (P = 0.04). CONCLUSIONS: In this cohort of participants with mild to moderate diabetic neuropathy, elevated triglycerides correlated with MFD loss independent of disease duration, age, diabetes control, or other variables. These data support the evolving concept that hyperlipidemia is instrumental in the progression of diabetic neuropathy.


Subject(s)
Diabetic Neuropathies/physiopathology , Disease Progression , Triglycerides/blood , Adult , Aged , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/blood , Diabetic Neuropathies/pathology , Electrophysiology , Female , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Multicenter Studies as Topic , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/physiology , Sural Nerve/pathology , Sural Nerve/physiopathology , Vibration
16.
Brain Res ; 1238: 154-62, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18775683

ABSTRACT

Radiologic and neuropsychologic studies suggest that diabetes mellitus causes structural changes in the brain and adversely effects cognitive development. Experimental animal models of type 1 diabetes mellitus (T1DM) have advanced these findings by demonstrating duration-related neuronal and cognitive deficits in T1DM BB/Wor rats. We studied the expression of receptor for advanced glycation end products (RAGE) and neuronal densities in the brains of two patients who died as the result of clinical brain edema(BE)that developed during the treatment of severe diabetic ketoacidosis (DKA). RAGE was markedly and diffusely expressed in blood vessels, neurons, and the choroid plexus and co-localized with glial fibrillary acidic protein (GFAP) in astrocytes. Significant neuronal loss was seen in the hippocampus and frontal cortex. Astrocytosis was present and white matter was atrophied in both cases when compared to age-matched controls. Our data supports that a neuroinflammatory response occurs in the BE associated with DKA, and that even after a relatively short duration of poorly controlled T1DM, the pathogenesis of primary diabetic encephalopathy can be initiated.


Subject(s)
Brain Edema/metabolism , Brain Edema/pathology , Diabetic Ketoacidosis/metabolism , Neurons/pathology , Receptors, Immunologic/biosynthesis , Adolescent , Brain Edema/etiology , Diabetes Mellitus, Type 1 , Diabetic Ketoacidosis/complications , Diabetic Ketoacidosis/pathology , Female , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein , Gliosis/etiology , Gliosis/pathology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Receptor for Advanced Glycation End Products
17.
Front Biosci ; 13: 4809-16, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508546

ABSTRACT

Diabetic neuropathy and its underlying pathogenesis are reviewed. It has been documented for some time that diabetic neuropathy differs in both human and experimental type 1 versus type 2 diabetes. Such differences are accounted for by impaired insulin action and signal transduction in type 1 diabetes, whereas hyperglycemia per se contributes equally to neuropathy in the two types of diabetes. Such differences in basic initiating factors and pathogenesis translate into differences in the functional and structural expressions of neuropathy in type 1 and type 2 diabetes. Type 1 neuropathy shows a more rapid progression with more severe functional and structural changes. Several experimental mono-therapies have been tested over the last decades which unfortunately have not been efficacious. Therefore discrepancies in underlying pathogenetic mechanisms in the two types of diabetic neuropathy will have to be taken into account in the design of future therapies, which should target several key pathogenetic mechanisms. Therapies that meet these criteria include replacement of acetyl-L-carnitine and replenishment of C-peptide in type 1 diabetic neuropathy.


Subject(s)
Diabetic Neuropathies/physiopathology , Diabetic Neuropathies/therapy , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/metabolism , Disease Models, Animal , Humans , Rats
18.
Exp Diabetes Res ; 2008: 230458, 2008.
Article in English | MEDLINE | ID: mdl-18437223

ABSTRACT

Diabetic polyneuropathy (DPN) occurs more frequently in type 1 diabetes resulting in a more severe DPN. The differences in DPN between the two types of diabetes are due to differences in the availability of insulin and C-peptide. Insulin and C-peptide provide gene regulatory effects on neurotrophic factors with effects on axonal cytoskeletal proteins and nerve fiber integrity. A significant abnormality in type 1 DPN is nodal degeneration. In the type 1 BB/Wor-rat, C-peptide replacement corrects metabolic abnormalities ameliorating the acute nerve conduction defect. It corrects abnormalities of neurotrophic factors and the expression of neuroskeletal proteins with improvements of axonal size and function. C-peptide corrects the expression of nodal adhesive molecules with prevention and repair of the functionally significant nodal degeneration. Cognitive dysfunction is a recognized complication of type 1 diabetes, and is associated with impaired neurotrophic support and apoptotic neuronal loss. C-peptide prevents hippocampal apoptosis and cognitive deficits. It is therefore clear that substitution of C-peptide in type 1 diabetes has a multitude of effects on DPN and cognitive dysfunction. Here the effects of C-peptide replenishment will be extensively described as they pertain to DPN and diabetic encephalopathy, underpinning its beneficial effects on neurological complications in type 1 diabetes.


Subject(s)
Brain Diseases/drug therapy , C-Peptide/therapeutic use , Diabetes Mellitus, Type 1/physiopathology , Diabetic Neuropathies/drug therapy , Animals , Brain Diseases/etiology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Hyperalgesia/drug therapy , Rats , Rats, Inbred BB
19.
Curr Drug Targets ; 9(1): 37-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18220711

ABSTRACT

In this review we will describe the interaction between insulin and C-peptide which enhances and attenuates insulin-signaling functions. We will describe how replenishment of C-peptide prevents and reverses the early metabolic abnormalities in type 1 diabetic polyneuropathy, such as Na(+)/K(+)-ATPase activity and endoneurial vascular NO release, resulting in prevention and reversal of early nerve dysfunction. The effects on expression of neurotrophic factors and their receptors, mediated by corrections of early gene responses and transcription factors, have downstream beneficial effects on cytoskeletal protein mRNAs and protein expression. Similar effects probably underlie corrections of cell adhesive molecules. The end-effects are prevention and reversal of myelinated and unmyelinated axonal degeneration, atrophy, and loss. Similarly, progressive degeneration of the node and paranode is prevented and repaired by C-peptide replacement with normalization of the molecular constituents of these functionally important structures. Cognitive dysfunction is now recognized as a complication of type 1 diabetes. Experimentally it is linked to impaired synaptic plasticity and eventually apoptotic neuronal loss caused by impaired insulin action and neurotrophic support. C-peptide replacement partially prevents hippocampal neuronal apoptosis and cognitive deficits. It is therefore becoming increasingly clear that C-peptide has major functions in supporting insulin action with a multitude of beneficial effects on diabetic polyneuropathy and primary diabetic encephalopathy in type 1 diabetes.


Subject(s)
C-Peptide/metabolism , Diabetes Mellitus, Type 1/complications , Diabetic Neuropathies/physiopathology , Insulin/metabolism , Animals , Brain Diseases, Metabolic/physiopathology , Cognition Disorders/etiology , Diabetes Complications/physiopathology , Gene Expression Regulation , Humans , Signal Transduction
20.
CNS Drugs ; 21 Suppl 1: 13-23; discussion 45-6, 2007.
Article in English | MEDLINE | ID: mdl-17696589

ABSTRACT

Diabetic polyneuropathy (DPN) is the most common late complication of diabetes mellitus. The underlying pathogenesis is multifaceted, with partly interrelated mechanisms that display a dynamic course. The mechanisms underlying DPN in type 1 and type 2 diabetes mellitus show overlaps or may differ. The differences are mainly due to insulin deficiency in type 1 diabetes which exacerbates the abnormalities caused by hyperglycaemia. Experimental DPN in rat models have identified early metabolic abnormalities with consequences for nerve conduction velocities and endoneurial blood flow. When corrected, the early functional deficits are usually normalised. On the other hand, if not corrected, they lead to abnormalities in lipid peroxidation and expression of neurotrophic factors which in turn result in axonal, nodal and paranodal degenerative changes with worsening of nerve function. As the structural changes progress, they become increasingly less amendable to metabolic interventions. In the past several years, experimental drugs--such as aldose reductase inhibitors, antioxidants and protein kinase C inhibitors--have undergone clinical trials, with disappointing outcomes. These drugs, targeting a single underlying pathogenetic factor, have in most cases been initiated at the advanced stage of DPN. In contrast, substitution of acetyl-L-carnitine (ALC) or C-peptide in type 1 DPN target a multitude of underlying mechanisms and are therefore more likely to be effective on a broader spectrum of the underlying pathogenesis. Clinical trials utilising ALC have shown beneficial effects on nerve conduction slowing, neuropathic pain, axonal degenerative changes and nerve fibre regeneration, despite relatively late initiation in the natural history of DPN. Owing to the good safety profile of ALC, early initiation of ALC therapy would be justified, with potentially greater benefits.


Subject(s)
Acetylcarnitine/therapeutic use , Diabetic Neuropathies/drug therapy , Nootropic Agents/therapeutic use , Animals , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...