Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
ACS Pharmacol Transl Sci ; 6(2): 306-319, 2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36798471

ABSTRACT

Charcot-Marie-Tooth X1 (CMTX1) disease is an inherited peripheral neuropathy that arises from loss-of-function mutations in the protein connexin 32 (Cx32). CMTX1 currently lacks a pharmacologic approach toward disease management, and we have previously shown that modulating the expression of molecular chaperones using novologue therapy may provide a viable disease-modifying approach to treat metabolic and demyelinating neuropathies. Cemdomespib is an orally bioavailable novologue that manifests neuroprotective activity by modulating the expression of heat shock protein 70 (Hsp70). We examined if 1 to 5 months of daily cemdomespib therapy may improve neuropathic symptoms in three mouse models of CMTX1 (Cx32 deficient (Cx32def), T55I-Cx32def, and R75W-Cx32 mice). Daily drug therapy significantly improved motor nerve conduction velocity (MNCV) and grip strength in all three models, but the compound muscle action potential was only improved in Cx32def mice. Drug efficacy required Hsp70 as improvements in MNCV, and the grip strength was abrogated in Cx32def × Hsp70 knockout mice. Five months of novologue therapy was associated with improved neuromuscular junction morphology, femoral motor nerve myelination, reduction in foamy macrophages, and a decrease in Schwann cell c-jun levels. To determine if c-jun may be downstream of Hsp70 and necessary for drug efficacy, c-jun expression was specifically deleted in Schwann cells of Cx32def mice. While the deletion of c-jun worsened the neuropathy, cemdomespib therapy remained effective in improving MNCV and grip strength. Our data show that cemdomespib therapy improves CMTX1-linked neuropathy in an Hsp70-dependent but a c-jun-independent manner and without regard to the nature of the underlying Cx32 mutation.

2.
Bioorg Med Chem ; 70: 116940, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35905686

ABSTRACT

KU-32 (2) and KU-596 (3), are first and second generation cytoprotective novologues that are derivatives of novobiocin (1), a heat shock protein 90 (Hsp90) C-terminal inhibitor. Although 2 and 3 improve mitochondrial bioenergetics and have demonstrated considerable cytoprotective activity, they contain a synthetically demanding noviose sugar. This issue was initially addressed by creating noviomimetics, such as KU-1202 (4), which replaced the noviose sugar with ether-linked cyclohexyl derivatives that retained some cytoprotective potential due to their ability to increase mitochondrial bioenergetics. Based on structure-activity relationship (SAR) studies of KU-1202 (4), the current study investigated 3'- and 4'-substituted cyclohexyl scaffolds as noviomimetics and determined their efficacy at increasing mitochondrial bioenergetic as a marker for cytoprotective potential.


Subject(s)
HSP90 Heat-Shock Proteins , Novobiocin , Mitochondria/metabolism , Novobiocin/pharmacology , Respiration , Sugars
3.
ACS Chem Neurosci ; 12(16): 3049-3059, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34340312

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a complication of diabetes whose pathophysiology is linked to altered mitochondrial bioenergetics (mtBE). KU-596 is a small molecule neurotherapeutic that reverses symptoms of DPN, improves sensory neuron mtBE, and decreases the pro-oxidant protein, thioredoxin-interacting protein (Txnip) in a heat shock protein 70 (Hsp70)-dependent manner. However, the mechanism by which KU-596 improves mtBE and the role of Txnip in drug efficacy remains unknown. Mitophagy is a quality-control mechanism that selectively targets damaged mitochondria for degradation. The goal of this study was to determine if KU-596 therapy improved DPN, mtBE, and mitophagy in an Hsp70- and Txnip-dependent manner. Mito-QC (MQC) mice express a mitochondrially targeted mCherry-GFP fusion protein that enables visualizing mitophagy. Diabetic MQC, MQC × Hsp70 knockout (KO), and MQC × Txnip KO mice developed sensory and nerve conduction dysfunctions consistent with the onset of DPN. KU-596 therapy improved these measures, and this was dependent on Hsp70 but not Txnip. In MQC mice, diabetes decreased mtBE and increased mitophagy and KU-596 treatment reversed these effects. In contrast, KU-596 was unable to improve mtBE and decrease mitophagy in MQC × Hsp70 and MQC × Txnip KO mice. These data suggest that Txnip is not necessary for the development of the sensory symptoms and mitochondrial dysfunction induced by diabetes. KU-596 therapy may improve mitochondrial tolerance to diabetic stress to decrease mitophagic clearance in an Hsp70- and Txnip-dependent manner.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Animals , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Energy Metabolism , HSP70 Heat-Shock Proteins/metabolism , Mice , Mitochondria/metabolism , Mitophagy , Sensory Receptor Cells/metabolism , Thioredoxins/metabolism
4.
Exp Neurol ; 313: 88-97, 2019 03.
Article in English | MEDLINE | ID: mdl-30557564

ABSTRACT

Neuronal mitochondrial dysfunction and oxidative stress are key pathophysiologic mechanisms of diabetic peripheral neuropathy (DPN). KU-596 is a small molecule modulator of heat shock protein 90 (Hsp90) that can reverse clinically relevant measures of DPN in diabetic animal models. Mechanistically, drug efficacy requires Hsp70 and correlates with improving mitochondrial maximal respiratory capacity (MRC) and decreasing oxidative stress in diabetic sensory neurons. The goal of this study was to determine if ex vivo treatment of diabetic neurons with KU-596 improves MRC by decreasing glucose-induced oxidative stress in an Hsp70-dependent manner. Sensory neurons were isolated from non-diabetic or diabetic mice wild type (WT) or Hsp70 knockout (Hsp70 KO) mice and treated with KU-596 in the presence of low or high glucose concentrations. In diabetic WT and Hsp70 KO neurons, hyperglycemia significantly increased superoxide levels, but KU-596 only decreased superoxide in WT neurons. Similarly, KU-596 significantly improved MRC in diabetic WT neurons maintained in high glucose but did not improve MRC in diabetic Hsp70 KO neurons under the same conditions. Since manganese superoxide dismutase (MnSOD) is the main mechanism to detoxify mitochondrial superoxide radicals, the cause and effect relationship between improved respiration and decreased oxidative stress was examined after knocking down MnSOD. Downregulating MnSOD in diabetic WT neurons increased hyperglycemia-induced superoxide levels, which was still significantly decreased by KU-596. However, KU-596 did not improve MRC following MnSOD knockdown. These data suggest that the ability of KU-596 to improve MRC is not necessarily dependent on decreasing mitochondrial superoxide in a MnSOD-dependent manner.


Subject(s)
Energy Metabolism/drug effects , Glycosides/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Phenethylamines/pharmacology , Sensory Receptor Cells/metabolism , Superoxide Dismutase/biosynthesis , Superoxides/metabolism , Animals , Diabetic Neuropathies/metabolism , Down-Regulation/drug effects , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hyperglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption/drug effects
5.
Bioorg Med Chem Lett ; 28(16): 2701-2704, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29759728

ABSTRACT

KU-596 is a second-generation C-terminal heat shock protein 90 KDa (Hsp90) modulator based on the natural product, novobiocin. KU-596 has been shown to induce Hsp70 levels and manifest neuroprotective activity through induction of the heat shock response. A ring-constrained analog of KU-596 was designed and synthesized to probe its binding orientation and ability to induce Hsp70 levels. Compound 2 was found to exhibit comparable or increased activity compared to KU-596, which is under clinical investigation for the treatment of neuropathy.


Subject(s)
Glycosides/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams/pharmacology , Neuroprotective Agents/pharmacology , Phenanthridines/pharmacology , Animals , Binding Sites , Cell Line, Transformed , Glycosides/chemical synthesis , Glycosides/chemistry , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/chemistry , Hydrogen Bonding , Lactams/chemical synthesis , Lactams/chemistry , Mitochondria/metabolism , Molecular Docking Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Phenanthridines/chemical synthesis , Phenanthridines/chemistry , Phenethylamines/chemistry , Rats , Transcriptional Activation
6.
ACS Chem Neurosci ; 9(2): 381-390, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29120605

ABSTRACT

Increased expression of the c-jun transcription factor occurs in a variety of human neuropathies and is critical in promoting Schwann cell (SC) dedifferentiation and loss of the myelinated phenotype. Using cell culture models, we previously identified KU-32 as a novobiocin-based C-terminal heat shock protein 90 (Hsp90) inhibitor that decreased c-jun expression and the extent of demyelination. Additional chemical optimization has yielded KU-596 as a neuroprotective novologue whose mechanistic efficacy to improve a metabolic neuropathy requires the expression of Hsp70. The current study examined whether KU-596 therapy could decrease c-jun expression and improve motor function in an inducible transgenic model of a SC-specific demyelinating neuropathy (MPZ-Raf mice). Treating MPZ-Raf mice with tamoxifen activates the MAPK kinase pathway, increases c-jun expression and produces a profound demyelinating neuropathy characterized by a loss of motor function and paraparesis. KU-596 therapy did not interfere with MAPK activation but reduced c-jun expression, significantly improved motor performance, and ameliorated the extent of peripheral nerve demyelination in both prevention and intervention studies. Hsp70 was necessary for the drug's neuroprotective efficacy since MPZ-Raf × Hsp70 knockout mice did not respond to KU-596 therapy. Collectively, our data indicate that modulating Hsp70 may provide a novel therapeutic approach to attenuate SC c-jun expression and ameliorate the onset of certain demyelinating neuropathies in humans.


Subject(s)
Demyelinating Diseases/drug therapy , Glycosides/pharmacology , HSP70 Heat-Shock Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Peripheral Nervous System Diseases/drug therapy , Phenethylamines/pharmacology , Animals , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Female , Gene Expression/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Random Allocation , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Tamoxifen , raf Kinases/genetics , raf Kinases/metabolism
7.
Eur J Med Chem ; 143: 1428-1435, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29137866

ABSTRACT

Heat shock protein 90 (Hsp90) is a chaperone under investigation for the treatment of cancer and neurodegenerative diseases. Neuroprotective Hsp90 C-terminal inhibitors derived from novobiocin (novologues) include KU-32 and KU-596. These novologues modulate molecular chaperones and result in an induction of Heat Shock Protein 70 (Hsp70). "Noviomimetics" replace the synthetically complex noviose sugar with a simple cyclohexyl moiety to maintain biological efficacy as compared to novologues KU-596 and KU-32. In this study, we further explore the development of noviomimetics and evaluate their efficacy using a luciferase refolding assay, immunoblot analysis, a c-jun assay, and an assay measuring mitochondrial bioenergetics. These new noviomimetics were designed and synthesized and found to induce Hsp70 and improve biological activity. Noviomimetics 39e and 40a were found to induce Hsp70 and exhibit promising effects in cellular assays.


Subject(s)
Drug Discovery , HSP90 Heat-Shock Proteins/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Novobiocin/chemistry , Novobiocin/pharmacology , Cell Line , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Models, Molecular , Protein Conformation
8.
ACS Med Chem Lett ; 7(8): 813-8, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27563408

ABSTRACT

Novobiocin is a natural product that binds the Hsp90 C-terminus and manifests Hsp90 inhibitory activity. Structural investigations on novobiocin led to the development of both anti-cancer and neuroprotective agents. The varied pharmacological activity manifested by these novobiocin analogs prompted the investigation of structure-function studies to identify these contradictory effects, which revealed that modifications to the amide side chain produce either anti-cancer or neuroprotective activity. Compounds that exhibit neuroprotective activity contain a short alkyl or cycloalkyl amide side chain. In contrast, anti-cancer agents contain five or more carbons, disrupt interactions between Hsp90α and Aha1, and induce the degradation of Hsp90-dependent client proteins.

9.
Curr Diab Rep ; 16(8): 71, 2016 08.
Article in English | MEDLINE | ID: mdl-27318486

ABSTRACT

The chaperome constitutes a broad family of molecular chaperones and co-chaperones that facilitate the folding, refolding, and degradation of the proteome. Heat shock protein 90 (Hsp90) promotes the folding of numerous oncoproteins to aid survival of malignant phenotypes, and small molecule inhibitors of the Hsp90 chaperone complex offer a viable approach to treat certain cancers. One therapeutic attribute of this approach is the selectivity of these molecules to target high affinity oncogenic Hsp90 complexes present in tumor cells, which are absent in nontransformed cells. This selectivity has given rise to the idea that disease may contribute to forming a stress chaperome that is functionally distinct in its ability to interact with small molecule Hsp90 modulators. Consistent with this premise, modulating Hsp90 improves clinically relevant endpoints of diabetic peripheral neuropathy but has little impact in nondiabetic nerve. The concept of targeting the "diabetic chaperome" to treat diabetes and its complications is discussed.


Subject(s)
Diabetic Neuropathies/therapy , Molecular Chaperones/metabolism , Molecular Targeted Therapy , Proteome/metabolism , Animals , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans
10.
J Biol Chem ; 291(16): 8309-23, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26872972

ABSTRACT

Heat shock protein 90 (HSP90) is a molecular chaperone that is up-regulated in cancer and is required for the folding of numerous signaling proteins. Consequently, HSP90 represents an ideal target for the development of new anti-cancer agents. The human HSP90 isoform, glucose-regulated protein 94 (GRP94), resides in the endoplasmic reticulum and regulates secretory pathways, integrins, and Toll-like receptors, which contribute to regulating immunity and metastasis. However, the cellular function of GRP94 remains underinvestigated. We report that GRP94 knockdown cells are defective in intracellular transport and, consequently, negatively impact the trafficking of F-actin toward the cellular cortex, integrin α2 and integrin αL toward the cell membrane and filopodia, and secretory vesicles containing the HSP90α-AHA1-survivin complex toward the leading edge. As a result, GRP94 knockdown cells form a multipolar spindle instead of bipolar morphology and consequently manifest a defect in cell migration and adhesion.


Subject(s)
Cell Movement , Cell Polarity , HSP90 Heat-Shock Proteins/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Cell Line, Tumor , HSP90 Heat-Shock Proteins/genetics , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Transport , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
11.
ACS Med Chem Lett ; 7(1): 67-71, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26819668

ABSTRACT

KU-32 and KU-596 are novobiocin-derived, C-terminal heat shock protein 90 (Hsp90) modulators that induce Hsp70 levels and manifest neuroprotective activity. However, the synthetically complex noviose sugar requires 10 steps to prepare, which makes translational development difficult. In this study, we developed a series of "noviomimetic" analogues of KU-596, which contain noviose surrogates that can be easily prepared, while maintaining the ability to induce Hsp70 levels. Both sugar and sugar analogues were designed, synthesized, and evaluated in a luciferase reporter assay, which identified compound 37, a benzyl containing noviomimetic, as the most potent inducer of Hsp70.

12.
Cell Stem Cell ; 18(2): 214-28, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26627594

ABSTRACT

The mammalian imprinted Dlk1-Gtl2 locus produces multiple non-coding RNAs (ncRNAs) from the maternally inherited allele, including the largest miRNA cluster in the mammalian genome. This locus has characterized functions in some types of stem cell, but its role in hematopoietic stem cells (HSCs) is unknown. Here, we show that the Dlk1-Gtl2 locus plays a critical role in preserving long-term repopulating HSCs (LT-HSCs). Through transcriptome profiling in 17 hematopoietic cell types, we found that ncRNAs expressed from the Dlk1-Gtl2 locus are predominantly enriched in fetal liver HSCs and the adult LT-HSC population and sustain long-term HSC functionality. Mechanistically, the miRNA mega-cluster within the Dlk1-Gtl2 locus suppresses the entire PI3K-mTOR pathway. This regulation in turn inhibits mitochondrial biogenesis and metabolic activity and protects LT-HSCs from excessive reactive oxygen species (ROS) production. Our data therefore show that the imprinted Dlk1-Gtl2 locus preserves LT-HSC function by restricting mitochondrial metabolism.


Subject(s)
Genetic Loci , Hematopoietic Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Antigens, CD/metabolism , Calcium-Binding Proteins , Fetus/metabolism , Genomic Imprinting , HEK293 Cells , Humans , Liver/cytology , Liver/embryology , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Organelle Biogenesis , Reactive Oxygen Species/metabolism , Signal Transduction , Sirolimus/pharmacology
13.
ACS Chem Neurosci ; 6(9): 1637-48, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26161583

ABSTRACT

We have previously demonstrated that modulating molecular chaperones with KU-32, a novobiocin derivative, ameliorates physiologic and bioenergetic deficits of diabetic peripheral neuropathy (DPN). Replacing the coumarin core of KU-32 with a meta-fluorinated biphenyl ring system created KU-596, a novobiocin analogue (novologue) that showed neuroprotective activity in a cell-based assay. The current study sought to determine whether KU-596 offers similar therapeutic potential for treating DPN. Administration of 2-20 mg/kg of KU-596 improved diabetes induced hypoalgesia and sensory neuron bioenergetic deficits in a dose-dependent manner. However, the drug could not improve these neuropathic deficits in diabetic heat shock protein 70 knockout (Hsp70 KO) mice. To gain further insight into the mechanisms by which KU-596 improved DPN, we performed transcriptomic analysis of sensory neuron RNA obtained from diabetic wild-type and Hsp70 KO mice using RNA sequencing. Bioinformatic analysis of the differentially expressed genes indicated that diabetes strongly increased inflammatory pathways and that KU-596 therapy effectively reversed these increases independent of Hsp70. In contrast, the effects of KU-596 on decreasing the expression of genes regulating the production of reactive oxygen species were more Hsp70-dependent. These data indicate that modulation of molecular chaperones by novologue therapy offers an effective approach toward correcting nerve dysfunction in DPN but that normalization of inflammatory pathways alone by novologue therapy seems to be insufficient to reverse sensory deficits associated with insensate DPN.


Subject(s)
Diabetic Neuropathies/drug therapy , Glycosides/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Phenethylamines/pharmacology , Sensory Receptor Cells/drug effects , Transcriptome/drug effects , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/metabolism , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Lumbar Vertebrae , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pain/metabolism , Reactive Oxygen Species/metabolism , Sensory Receptor Cells/metabolism
14.
ACS Chem Biol ; 10(2): 577-90, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25402753

ABSTRACT

Human Hsp90 isoforms are molecular chaperones that are often up-regulated in malignances and represent a primary target for Hsp90 inhibitors undergoing clinical evaluation. Hsp90α is a stress-inducible isoform of Hsp90 that plays a significant role in apoptosis and metastasis. Though Hsp90α is secreted into the extracellular space under metastatic conditions, its role in cancer biology is poorly understood. We report that Hsp90α associates with the Aha1 co-chaperone and found this complex to localize in secretory vesicles and at the leading edge of migrating cells. Knockdown of Hsp90α resulted in a defect in cell migration. The functional role of Hsp90α/Aha1 was studied by treating the cells with various novobiocin-based Hsp90 C-terminal inhibitors. These inhibitors disrupted the Hsp90α/Aha1 complex, caused a cytoplasmic redistribution of Hsp90α and Aha1, and decreased cell migration. Structure-function studies determined that disruption of Hsp90α/Aha1 association and inhibition of cell migration correlated with the presence of a benzamide side chain, since an acetamide substituted analog was less effective. Our results show that disruption of Hsp90α/Aha1 interactions with novobiocin-based Hsp90 C-terminal inhibitors may limit the metastatic potential of tumors.


Subject(s)
Cell Movement/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Molecular Chaperones/metabolism , Novobiocin/pharmacology , Cell Line , Gene Expression Regulation , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Protein Binding , Protein Folding , Protein Isoforms
15.
J Pharmacol Exp Ther ; 348(2): 281-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263156

ABSTRACT

Impaired neuronal mitochondrial bioenergetics contributes to the pathophysiologic progression of diabetic peripheral neuropathy (DPN) and may be a focal point for disease management. We have demonstrated that modulating heat shock protein (Hsp) 90 and Hsp70 with the small-molecule drug KU-32 ameliorates psychosensory, electrophysiologic, morphologic, and bioenergetic deficits of DPN in animal models of type 1 diabetes. The current study used mouse models of type 1 and type 2 diabetes to determine the relationship of changes in sensory neuron mitochondrial bioenergetics to the onset of and recovery from DPN. The onset of DPN showed a tight temporal correlation with a decrease in mitochondrial bioenergetics in a genetic model of type 2 diabetes. In contrast, sensory hypoalgesia developed 10 weeks before the occurrence of significant declines in sensory neuron mitochondrial bioenergetics in the type 1 model. KU-32 therapy improved mitochondrial bioenergetics in both the type 1 and type 2 models, and this tightly correlated with a decrease in DPN. Mechanistically, improved mitochondrial function following KU-32 therapy required Hsp70, since the drug was ineffective in diabetic Hsp70 knockout mice. Our data indicate that changes in mitochondrial bioenergetics may rapidly contribute to nerve dysfunction in type 2 diabetes, but not type 1 diabetes, and that modulating Hsp70 offers an effective approach toward correcting sensory neuron bioenergetic deficits and DPN in both type 1 and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/prevention & control , HSP70 Heat-Shock Proteins/metabolism , Hypoglycemic Agents/therapeutic use , Mitochondria/drug effects , Novobiocin/analogs & derivatives , Oxidative Phosphorylation/drug effects , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HSP70 Heat-Shock Proteins/genetics , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Neuritis/prevention & control , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Novobiocin/administration & dosage , Novobiocin/blood , Novobiocin/pharmacokinetics , Novobiocin/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism
16.
Acta Neuropathol Commun ; 1: 39, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-24252174

ABSTRACT

BACKGROUND: Aberrant neuron/glia interactions can contribute to a variety of neurodegenerative diseases and we have previously demonstrated that enhanced activation of Erb B2, which is a member of the epidermal growth factor receptor (EGFR) family, can contribute to the development of diabetic peripheral neuropathy (DPN). In peripheral nerves, Erb B receptors are activated by various members of the neuregulin-1 (NRG1) family including NRG1 Type I, NRG1 Type II and NRG1 Type III to regulate Schwann cell (SC) growth, migration, differentiation and dedifferentiation. Alternatively, Erb B2 activity can be negatively regulated by association with the Erb B2-interacting protein, erbin. Since the effect of diabetes on the expression of NRG1 isoforms and erbin in peripheral nerve are unknown, the current study determined whether changes in NRG1 isoforms and erbin may be associated with altered Erb B2 signaling in DPN. RESULTS: Swiss Webster mice were rendered diabetic with streptozotocin (STZ) and after 12 weeks of diabetes, treated with erlotinib, an inhibitor of Erb B2 activation. Inhibition of Erb B2 signaling partially reversed several pathophysiologic aspects of DPN including a pronounced sensory hypoalgesia, nerve conduction velocity deficits and the decrease in epidermal nerve fiber innervation. We also observed a decrease of NRG1 Type III but an increase of NRG1 Type I level in diabetic sural nerves at early stage of diabetes. With disease progression, we detected reduced erbin expression and enhanced MAPK pathway activity in diabetic mice. Inhibition of Erb B2 receptor suppressed MAPK pathway activity in treated-diabetic sural nerves. CONCLUSIONS: These results support that hyperglycemia may impair NRG1/Erb B2 signaling by disrupting the balance between NRG1 isoforms, decreasing the expression of erbin and correspondingly activating the MAPK pathway. Together, imbalanced NRG1 isoforms and downregulated erbin may contribute to the dysregulation of Erb B2 signaling in the development of DPN.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/physiopathology , Neuregulin-1/metabolism , Receptor, ErbB-2/metabolism , Sural Nerve/physiopathology , Animals , Diabetic Neuropathies/drug therapy , Disease Progression , Down-Regulation , Erlotinib Hydrochloride , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Neural Conduction/drug effects , Neural Conduction/physiology , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Sural Nerve/drug effects
17.
ASN Neuro ; 4(7): e00102, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23240583

ABSTRACT

Modulating molecular chaperones is emerging as an attractive approach to treat neurodegenerative diseases associated with protein aggregation, DPN (diabetic peripheral neuropathy) and possibly, demyelinating neuropathies. KU-32 [N-(7-((2R,3R,4S,5R)-3,4-dihydroxy-5-methoxy-6,6-dimethyl-tetrahydro-2H-pyran-2-yloxy)-8-methyl-2-oxo-2H-chromen-3-yl)acetamide] is a small molecule inhibitor of Hsp90 (heat shock protein 90) and reverses sensory deficits associated with myelinated fibre dysfunction in DPN. Additionally, KU-32 prevented the loss of myelinated internodes induced by treating myelinated SC (Schwann cell)-DRG (dorsal root ganglia) sensory neuron co-cultures with NRG1 (neuregulin-1 Type 1). Since KU-32 decreased NRG1-induced demyelination in an Hsp70-dependent manner, the goal of the current study was to clarify how Hsp70 may be mechanistically linked to preventing demyelination. The activation of p42/p44 MAPK (mitogen-activated protein kinase) and induction of the transcription factor c-Jun serve as negative regulators of myelination. NRG1 activated MAPK, induced c-Jun expression and promoted a loss of myelin segments in DRG explants isolated from both WT (wild-type) and Hsp70 KO (knockout) mice. Although KU-32 did not block the activation of MAPK, it blocked c-Jun induction and protected against a loss of myelinated segments in WT mice. In contrast, KU-32 did not prevent the NRG1-dependent induction of c-Jun and loss of myelin segments in explants from Hsp70 KO mice. Overexpression of Hsp70 in myelinated DRG explants prepared from WT or Hsp70 KO mice was sufficient to block the induction of c-Jun and the loss of myelin segments induced by NRG1. Lastly, inhibiting the proteasome prevented KU-32 from decreasing c-Jun levels. Collectively, these data support that Hsp70 induction is sufficient to prevent NRG1-induced demyelination by enhancing the proteasomal degradation of c-Jun.


Subject(s)
Demyelinating Diseases/metabolism , HSP70 Heat-Shock Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neuregulin-1/metabolism , Signal Transduction/physiology , Animals , Coculture Techniques , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
18.
Pharmacol Rev ; 64(4): 880-900, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22885705

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a common complication of diabetes that is associated with axonal atrophy, demyelination, blunted regenerative potential, and loss of peripheral nerve fibers. The development and progression of DPN is due in large part to hyperglycemia but is also affected by insulin deficiency and dyslipidemia. Although numerous biochemical mechanisms contribute to DPN, increased oxidative/nitrosative stress and mitochondrial dysfunction seem intimately associated with nerve dysfunction and diminished regenerative capacity. Despite advances in understanding the etiology of DPN, few approved therapies exist for the pharmacological management of painful or insensate DPN. Therefore, identifying novel therapeutic strategies remains paramount. Because DPN does not develop with either temporal or biochemical uniformity, its therapeutic management may benefit from a multifaceted approach that inhibits pathogenic mechanisms, manages inflammation, and increases cytoprotective responses. Finally, exercise has long been recognized as a part of the therapeutic management of diabetes, and exercise can delay and/or prevent the development of painful DPN. This review presents an overview of existing therapies that target both causal and symptomatic features of DPN and discusses the role of up-regulating cytoprotective pathways via modulating molecular chaperones. Overall, it may be unrealistic to expect that a single pharmacologic entity will suffice to ameliorate the multiple symptoms of human DPN. Thus, combinatorial therapies that target causal mechanisms and enhance endogenous reparative capacity may enhance nerve function and improve regeneration in DPN if they converge to decrease oxidative stress, improve mitochondrial bioenergetics, and increase response to trophic factors.


Subject(s)
Diabetic Neuropathies/therapy , Analgesics/therapeutic use , Animals , Diabetic Neuropathies/metabolism , Exercise Therapy , Humans , Hypoglycemic Agents/therapeutic use , Molecular Chaperones
19.
J Med Chem ; 55(12): 5797-812, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22702513

ABSTRACT

Compound 2 (KU-32) is a first-generation novologue (a novobiocin-based, C-terminal, heat shock protein 90 (Hsp90) inhibitor) that decreases glucose-induced death of primary sensory neurons and reverses numerous clinical indices of diabetic peripheral neuropathy in mice. The current study sought to exploit the C-terminal binding site of Hsp90 to determine whether the optimization of hydrogen bonding and hydrophobic interactions of second-generation novologues could enhance neuroprotective activity. Using a series of substituted phenylboronic acids to replace the coumarin lactone of 2, we identified that electronegative atoms placed at the meta-position of the B-ring exhibit improved cytoprotective activity, which is believed to result from favorable interactions with Lys539 in the Hsp90 C-terminal binding pocket. Consistent with these results, a meta-3-fluorophenyl substituted novologue (13b) exhibited a 14-fold lower ED(50) for protection against glucose-induced toxicity of primary sensory neurons compared to 2.


Subject(s)
Cytoprotection/drug effects , Glucose/adverse effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Sensory Receptor Cells/drug effects , Chemistry Techniques, Synthetic , Humans , Models, Molecular , Neuroprotective Agents/chemistry , Protein Conformation , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
20.
Exp Neurol ; 235(1): 388-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22465570

ABSTRACT

Quantification of intra-epidermal nerve fibers (iENFs) is an important approach to stage diabetic peripheral neuropathy (DPN) and is a promising clinical endpoint for identifying beneficial therapeutics. Mechanistically, diabetes decreases neuronal mitochondrial function and enhancing mitochondrial respiratory capacity may aid neuronal recovery from glucotoxic insults. We have proposed that modulating the activity and expression of heat shock proteins (Hsp) may be of benefit in treating DPN. KU-32 is a C-terminal Hsp90 inhibitor that improved thermal hypoalgesia in diabetic C57Bl/6 mice but it was not determined if this was associated with an increase in iENF density and mitochondrial function. After 16 weeks of diabetes, Swiss Webster mice showed decreased electrophysiological and psychosensory responses and a >30% loss of iENFs. Treatment of the mice with ten weekly doses of 20mg/kg KU-32 significantly reversed pre-existing deficits in nerve conduction velocity and responses to mechanical and thermal stimuli. KU-32 therapy significantly reversed the pre-existing loss of iENFs despite the identification of a sub-group of drug-treated diabetic mice that showed improved thermal sensitivity but no increase in iENF density. To determine if the improved clinical indices correlated with enhanced mitochondrial activity, sensory neurons were isolated and mitochondrial bioenergetics assessed ex vivo using extracellular flux technology. Diabetes decreased maximal respiratory capacity in sensory neurons and this deficit was improved following KU-32 treatment. In conclusion, KU-32 improved physiological and morphologic markers of degenerative neuropathy and drug efficacy may be related to enhanced mitochondrial bioenergetics in sensory neurons.


Subject(s)
Diabetic Neuropathies/physiopathology , Epidermis/innervation , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Mitochondria/physiology , Molecular Chaperones/metabolism , Nerve Fibers/physiology , Sensory Receptor Cells/physiology , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mice , Neural Conduction/physiology , Pain Measurement
SELECTION OF CITATIONS
SEARCH DETAIL
...