Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 229(3): 876-887, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-37671668

ABSTRACT

Mycobacterium tuberculosis (Mtb)-specific γ9δ2 T cells secrete granzyme A (GzmA) protective against intracellular Mtb growth. However, GzmA-enzymatic activity is unnecessary for pathogen inhibition, and the mechanisms of GzmA-mediated protection remain unknown. We show that GzmA homodimerization is essential for opsonization of mycobacteria, altered uptake into human monocytes, and subsequent pathogen clearance within the phagolysosome. Although monomeric and homodimeric GzmA bind mycobacteria, only homodimers also bind cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4). Without access to surface-expressed CD14 and TLR4, GzmA fails to inhibit intracellular Mtb. Upregulation of Rab11FIP1 was associated with inhibitory activity. Furthermore, GzmA colocalized with and was regulated by protein disulfide isomerase AI (PDIA1), which cleaves GzmA homodimers into monomers and prevents Mtb inhibitory activity. These studies identify a previously unrecognized role for homodimeric GzmA structure in opsonization, phagocytosis, and elimination of Mtb in human monocytes, and they highlight PDIA1 as a potential host-directed therapy for prevention and treatment of tuberculosis, a major human disease.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Granzymes/metabolism , Monocytes/metabolism , Toll-Like Receptor 4/metabolism , Tuberculosis/microbiology
2.
Peptides ; 150: 170718, 2022 04.
Article in English | MEDLINE | ID: mdl-34954230

ABSTRACT

Connecting peptide, or C-peptide, is a part of the insulin prohormone and is essential for the proper folding and processing of the mature insulin peptide. C-peptide is released from the same beta cell secretory granules as insulin in equimolar amounts. However, due to their relative stabilities in plasma, the two peptides are detected in the circulation at ratios of approximately 4:1 to 6:1 (C-peptide to insulin), depending on metabolic state. C-peptide binds specifically to human cell membranes and induces intracellular signaling cascades, likely through an interaction with the G protein coupled receptor, GPR146. C-peptide has been shown to exert protective effects against the vascular, renal, and ocular complications of diabetes. The effects of C-peptide appear to be dependent upon the presence of insulin and the absolute, extracellular concentration of glucose. In this study, we employed HEK293 cells to further examine the interactive effects of C-peptide, insulin, and glucose on cell signaling. We observed that C-peptide's cellular effects are dampened significantly when cells are exposed to physiologically relevant concentrations of both insulin and C-peptide. Likewise, the actions of C-peptide on cFos and GPR146 mRNA expressions were affected by changes in extracellular glucose concentration. In particular, C-peptide induced significant elevations in cFos expression in the setting of high (25 mmol) extracellular glucose concentration. These data indicate that future experimentation on the actions of C-peptide should control for the presence or absence of insulin and the concentration of glucose. Furthermore, these findings should be considered prior to the development of C-peptide-based therapeutics for the treatment of diabetes-associated complications.


Subject(s)
Diabetes Complications , Insulin , Blood Glucose , C-Peptide , Glucose/pharmacology , HEK293 Cells , Humans , Insulin/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Front Immunol ; 12: 701227, 2021.
Article in English | MEDLINE | ID: mdl-34489949

ABSTRACT

Neutrophils are the most abundant white blood cells recruited to the sites of infection and inflammation. During neutrophil activation, myeloperoxidase (MPO) is released and converts hydrogen peroxide to hypochlorous acid (HOCl). HOCl reacts with plasmalogen phospholipids to liberate 2-chlorofatty aldehyde (2-ClFALD), which is metabolized to 2-chlorofatty acid (2-ClFA). 2-ClFA and 2-ClFALD are linked with inflammatory diseases and induce endothelial dysfunction, neutrophil extracellular trap formation (NETosis) and neutrophil chemotaxis. Here we examine the neutrophil-derived chlorolipid production in the presence of pathogenic E. coli strain CFT073 and non-pathogenic E. coli strain JM109. Neutrophils cocultured with CFT073 E. coli strain and JM109 E. coli strain resulted in 2-ClFALD production. 2-ClFA was elevated only in CFT073 coculture. NETosis is more prevalent in CFT073 cocultures with neutrophils compared to JM109 cocultures. 2-ClFA and 2-ClFALD were both shown to have significant bactericidal activity, which is more severe in JM109 E. coli. 2-ClFALD metabolic capacity was 1000-fold greater in neutrophils compared to either strain of E. coli. MPO inhibition reduced chlorolipid production as well as bacterial killing capacity. These findings indicate the chlorolipid profile is different in response to these two different strains of E. coli bacteria.


Subject(s)
Escherichia coli/immunology , Fatty Acids/metabolism , Neutrophil Activation/immunology , Neutrophils/immunology , Peroxidase/immunology , Cells, Cultured , Extracellular Traps/immunology , Humans , Neutrophils/enzymology
4.
Mo Med ; 118(4): 352-357, 2021.
Article in English | MEDLINE | ID: mdl-34373671

ABSTRACT

G protein-coupled receptors (GPCRs) transmit the signals of a variety of hormones and neurotransmitters and are targets of more than 30% of all FDA-approved drugs. We developed an approach for identifying the endogenous ligands for a family of orphan GPCRs that enables the development of novel therapeutics for the potential treatment of a wide variety of disorders including pain, diabetes, appetitive behaviors, infertility and obesity. With this approach, we have deorphanized five previously orphaned GPCRs.


Subject(s)
Obesity , Humans , Ligands
5.
Physiol Behav ; 235: 113380, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33705816

ABSTRACT

The existence of the peptide encoded by the cocaine- and amphetamine-regulated transcript (Cartpt) has been recognized since 1981, but it was not until 1995, that the gene encoding CART peptide (CART) was identified. With the availability of the predicted protein sequence of CART investigators were able to identify sites of peptide localization, which then led to numerous approaches attempting to clarify CART's multiple pharmacologic effects and even provide evidence of potential physiologic relevance. Although not without controversy, a picture emerged of the importance of CART in ingestive behaviors, reward behaviors and even pain sensation. Despite the wealth of data hinting at the significance of CART, in the absence of an identified receptor, the full potential for this peptide or its analogs to be developed into therapeutic agents remained unrealized. There was evidence favoring the action of CART via a G protein-coupled receptor (GPCR), but despite multiple attempts the identity of that receptor eluded investigators until recently. Now with the identification of the previously orphaned GPCR, GPR160, as a receptor for CART, focus on this pluripotent neuropeptide will in all likelihood experience a renaissance and the potential for the development of pharmcotherapies targeting GPR160 seems within reach.


Subject(s)
Cocaine , Neuropeptides , Feeding Behavior , Nerve Tissue Proteins/genetics , Reward
6.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R236-R249, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33206556

ABSTRACT

Recent work identified Gpr160 as a candidate receptor for cocaine- and amphetamine-regulated transcript peptide (CARTp) and described its role in pain modulation. The aims of the present study were to determine if Gpr160 is required for the CARTp's ability to reduce food intake and water intake and to initially identify the distribution of Gpr160-like immunoreactivity (Gpr160ir) in the rat brain. A passive immunoneutralization approach targeting Gpr160 was used to block the behavioral effects of a pharmacological dose of CARTp in the fourth cerebroventricle (4V) of rats and to determine the importance of endogenously produced CARTp in the control of ingestive behaviors. Passive immunoneutralization of Gpr160 in the 4V blocked the actions of CARTp to inhibit food intake and water intake. Blockade of Gpr160 in the 4V, independent of pharmacological CART treatment, caused an increase in both overnight food intake and water intake. The decrease in food intake, but not water intake, caused by central injection of CARTp was demonstrated to be interrupted by prior administration of a glucagon-like peptide 1 (GLP-1) receptor antagonist. Gpr160ir was observed in several, distinct sites throughout the rat brain, where CARTp staining has been described. Importantly, Gpr160ir was observed to be present in both neuronal and nonneuronal cell types. These data support the hypothesis that Gpr160 is required for the anorexigenic actions of central CARTp injection and extend these findings to water drinking. Gpr160ir was observed in both neuronal and nonneuronal cell types in regions known to be important in the multiple pharmacological effects of CARTp, identifying those areas as targets for future compromise of function studies.


Subject(s)
Appetite Depressants/pharmacology , Brain Stem/drug effects , Drinking Behavior/drug effects , Drinking/drug effects , Eating/drug effects , Feeding Behavior/drug effects , Nerve Tissue Proteins/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Brain Stem/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
7.
J Pharmacol Exp Ther ; 375(2): 367-375, 2020 11.
Article in English | MEDLINE | ID: mdl-32913007

ABSTRACT

Neuropathic pain is a debilitating public health concern for which novel non-narcotic therapeutic targets are desperately needed. Using unbiased transcriptomic screening of the dorsal horn spinal cord after nerve injury we have identified that Gpr183 (Epstein-Barr virus-induced gene 2) is upregulated after chronic constriction injury (CCI) in rats. GPR183 is a chemotactic receptor known for its role in the maturation of B cells, and the endogenous ligand is the oxysterol 7α,25-dihydroxycholesterol (7α,25-OHC). The role of GPR183 in the central nervous system is not well characterized, and its role in pain is unknown. The profile of commercially available probes for GPR183 limits their use as pharmacological tools to dissect the roles of this receptor in pathophysiological settings. Using in silico modeling, we have screened a library of 5 million compounds to identify several novel small-molecule antagonists of GPR183 with nanomolar potency. These compounds are able to antagonize 7α,25-OHC-induced calcium mobilization in vitro with IC50 values below 50 nM. In vivo intrathecal injections of these antagonists during peak pain after CCI surgery reversed allodynia in male and female mice. Acute intrathecal injection of the GPR183 ligand 7α,25-OHC in naïve mice induced dose-dependent allodynia. Importantly, this effect was blocked using our novel GPR183 antagonists, suggesting spinal GPR183 activation as pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify this receptor as a potential target for therapeutic intervention. SIGNIFICANCE STATEMENT: We have identified several novel GPR183 antagonists with nanomolar potency. Using these antagonists, we have demonstrated that GPR183 signaling in the spinal cord is pronociceptive. These studies are the first to reveal a role for GPR183 in neuropathic pain and identify it as a potential target for therapeutic intervention.


Subject(s)
Neuralgia/metabolism , Oxysterols/metabolism , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/metabolism , Animals , Female , HL-60 Cells , Humans , Male , Mice , Neuralgia/drug therapy , Neuralgia/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction , Spinal Cord/pathology
8.
J Clin Invest ; 130(5): 2587-2592, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31999650

ABSTRACT

Treating neuropathic pain is challenging and novel non-opioid-based medicines are needed. Using unbiased receptomics, transcriptomic analyses, immunofluorescence, and in situ hybridization, we found that the expression of the orphan GPCR Gpr160 and GPR160 increased in the rodent dorsal horn of the spinal cord following traumatic nerve injury. Genetic and immunopharmacological approaches demonstrated that GPR160 inhibition in the spinal cord prevented and reversed neuropathic pain in male and female rodents without altering normal pain response. GPR160 inhibition in the spinal cord attenuated sensory processing in the thalamus, a key relay in the sensory discriminative pathways of pain. We also identified cocaine- and amphetamine-regulated transcript peptide (CARTp) as a GPR160 ligand. Inhibiting endogenous CARTp signaling in spinal cord attenuated neuropathic pain, whereas exogenous intrathecal CARTp evoked painful hypersensitivity through GPR160-dependent ERK and cAMP response element-binding protein (CREB). Our findings de-orphanize GPR160, identify it as a determinant of neuropathic pain and potential therapeutic target, and provide insights into its signaling pathways. CARTp is involved in many diseases including depression and reward and addiction; de-orphanization of GPR160 is a major step forward understanding the role of CARTp signaling in health and disease.


Subject(s)
Neuralgia/etiology , Neuralgia/physiopathology , Receptors, G-Protein-Coupled/physiology , Animals , Cell Line , Female , Humans , Ligands , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/genetics , PC12 Cells , RNA, Small Interfering/genetics , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Signal Transduction , Spinal Cord/metabolism , Up-Regulation
9.
Brain Pathol ; 28(6): 806-821, 2018 11.
Article in English | MEDLINE | ID: mdl-30062819

ABSTRACT

BACKGROUND: Mutations in the three-prime repair exonuclease 1 (TREX1) gene have been associated with neurological diseases, including Retinal Vasculopathy with Cerebral Leukoencephalopathy (RVCL). However, the endogenous expression of TREX1 in human brain has not been studied. METHODS: We produced a rabbit polyclonal antibody (pAb) to TREX1 to characterize TREX1 by Western blotting (WB) of cell lysates from normal controls and subjects carrying an RVCL frame-shift mutation. Dual staining was performed to determine cell types expressing TREX1 in human brain tissue. TREX1 distribution in human brain was further evaluated by immunohistochemical analyses of formalin-fixed, paraffin-embedded samples from normal controls and patients with RVCL and ischemic stroke. RESULTS: After validating the specificity of our anti-TREX1 rabbit pAb, WB analysis was utilized to detect the endogenous wild-type and frame-shift mutant of TREX1 in cell lysates. Dual staining in human brain tissues from patients with RVCL and normal controls localized TREX1 to a subset of microglia and macrophages. Quantification of immunohistochemical staining of the cerebral cortex revealed that TREX1+ microglia were primarily in the gray matter of normal controls (22.7 ± 5.1% and 5.5 ± 1.9% of Iba1+ microglia in gray and white matter, respectively) and commonly in association with the microvasculature. In contrast, in subjects with RVCL, the TREX1+ microglia were predominantly located in the white matter of normal appearing cerebral cortex (11.8 ± 3.1% and 38.9 ± 5.8% of Iba1+ microglia in gray and white matter, respectively). The number of TREX1+ microglia was increased in ischemic brain lesions in central nervous system of RVCL and stroke patients. CONCLUSIONS: TREX1 is expressed by a subset of microglia in normal human brain, often in close proximity to the microvasculature, and increases in the setting of ischemic lesions. These findings suggest a role for TREX1+ microglia in vessel homeostasis and response to ischemic injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/physiology , Microglia/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Aged , Animals , Antibodies/metabolism , Brain/pathology , Exodeoxyribonucleases/genetics , Female , Frameshift Mutation , HEK293 Cells , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Homeostasis/physiology , Humans , Macrophages/metabolism , Male , Middle Aged , Phosphoproteins/genetics , Rabbits/immunology , Retinal Diseases/genetics , Retinal Diseases/pathology , Vascular Diseases/genetics , Vascular Diseases/pathology
10.
Peptides ; 106: 45-48, 2018 08.
Article in English | MEDLINE | ID: mdl-29933026

ABSTRACT

Using a series of classical protein purification techniques, coupled with more modern molecular approaches, a family of neuropeptides, the Phoenixins, was identified to be produced in brain and heart, and to bind selectively in pituitary gland, ovary and brain. These same binding sites were revealed, using a novel receptor identification strategy, to express the orphan G protein-coupled receptor, GPR173, the expression of which was required for the actions of phoenixin both in vivo and in vitro. In fact, studies using small interfering RNA molecules to compromise GPR173 expression revealed the physiologic relevance of the initially reported pharmacologic actions of the peptides. Those include not only the reproductive actions of the peptides in brain and pituitary gland, but also a CNS site of action in the maintenance of fluid and electrolyte homeostasis. Additional pharmacologic actions of the phoenixins have been described and the race is on to establish the physiologic relevance of those actions as well as the therapeutic potential of phoenixin analogs.


Subject(s)
Hypothalamic Hormones/physiology , Neuropeptides/physiology , Receptors, Peptide/metabolism , Animals , Brain/physiology , Disease Models, Animal , Female , Heart/physiology , Humans , Ovary/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics
11.
J Lipid Res ; 59(8): 1424-1432, 2018 08.
Article in English | MEDLINE | ID: mdl-29739865

ABSTRACT

Neutrophils form neutrophil extracellular traps (NETs), which have been implicated in microcirculatory plugging. NET formation (NETosis) involves the fusion of granule and nuclear contents, which are then released in the extracellular space. Myeloperoxidase (MPO) plays a major role in NETosis leading to the dissociation of DNA from histones. During neutrophil activation, MPO is released and activated to convert hydrogen peroxide and chloride to hypochlorous acid (HOCl). HOCl targets plasmalogens leading to the production of the chlorinated lipids, 2-chlorofatty aldehyde and 2-chlorofatty acid (2-ClFA). Here, we tested the hypothesis that 2-ClFAs are important lipid mediators of NETosis. Human neutrophils treated with physiological levels of 2-ClFAs formed NETs, characterized by MPO association with DNA and neutrophil elastase (NE) redistribution to the perinuclear area. 2-ClFA-induced NETs reduced Escerichia coli colony forming units. 2-ClFA-induced NETosis is calcium- and protein arginine deiminase 4-dependent. Interestingly, unlike PMA, 2-ClFA initiates the NETosis process without neutrophil activation and degranulation. Furthermore, 2-ClFA elicits NETosis in bone-marrow derived neutrophils from MPO-deficient mice. Taken together, these findings suggest 2-ClFA as an MPO product that triggers the NETosis pathway following neutrophil activation.


Subject(s)
Extracellular Traps/drug effects , Extracellular Traps/metabolism , Fatty Acids/pharmacology , Lipid Metabolism/drug effects , Neutrophils/cytology , Calcium/metabolism , DNA/metabolism , Fatty Acids/chemistry , Healthy Volunteers , Humans , Neutrophils/drug effects , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/metabolism
12.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R623-R628, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29364701

ABSTRACT

The newly described hypothalamic peptide, phoenixin, is produced in the hypothalamus and adenohypophysis, where it acts to control reproductive hormone secretion. Both phoenixin and its receptor GPR173 are expressed in the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei, suggesting additional, nonreproductive effects of the peptide to control vasopressin (AVP) or oxytocin (OT) secretion. Hypothalamo-neurohypophysial explants released AVP but not OT in response to phoenixin. Intracerebroventricular administration of phoenixin into conscious, unrestrained male and female rats significantly increased circulating AVP, but not OT, levels in plasma, and it increased immediate early gene expression in the supraoptic nuclei of male rats. Bath application of phoenixin in hypothalamic slice preparations resulted in depolarization of PVN neurons, indicating a direct, neural action of phoenixin in the hypothalamus. Our results suggest that the newly described, hypothalamic peptide phoenixin, in addition to its effects on hypothalamic and pituitary mechanisms controlling reproduction, may contribute to the physiological mechanisms regulating fluid and electrolyte homeostasis.


Subject(s)
Arginine Vasopressin/metabolism , Hypothalamic Hormones/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Peptide Hormones/physiology , Animals , Arginine Vasopressin/blood , Female , Gene Expression Regulation , Genes, fos , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/physiology , Hypothalamo-Hypophyseal System/metabolism , In Vitro Techniques , Injections, Intraventricular , Male , Membrane Potentials , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Sprague-Dawley , Secretory Pathway/drug effects , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism
13.
Development ; 144(17): 3080-3094, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760814

ABSTRACT

The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.


Subject(s)
Loop of Henle/embryology , Loop of Henle/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Multipotent Stem Cells/cytology , Organogenesis , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Gene Ontology , Homozygote , Kidney Tubules/metabolism , Loop of Henle/abnormalities , Mice , Multipotent Stem Cells/metabolism , Mutation/genetics , Organogenesis/genetics , Protein Binding/genetics , Transcription Factors/chemistry , Ureter/embryology , Ureter/metabolism
15.
Am J Physiol Regul Integr Comp Physiol ; 310(2): R143-55, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26561648

ABSTRACT

Neuronostatin (NST) is a recently described peptide that is produced from the somatostatin preprohormone in pancreatic δ-cells. NST has been shown to increase glucagon secretion from primary rat pancreatic islets in low-glucose conditions. Here, we demonstrate that NST increases proglucagon message in α-cells and identify a potential mechanism for NST's cellular activities, including the phosphorylation of PKA following activation of the G protein-coupled receptor, GPR107. GPR107 is abundantly expressed in the pancreas, particularly, in rodent and human α-cells. Compromise of GPR107 in pancreatic α-cells results in failure of NST to increase PKA phosphorylation and proglucagon mRNA levels. We also demonstrate colocalization of GPR107 and NST on both mouse and human pancreatic α-cells. Taken together with our group's observation that NST infusion in conscious rats impairs glucose clearance in response to a glucose challenge and that plasma levels of the peptide are elevated in the fasted compared with the fed or fasted-refed state, these studies support the hypothesis that endogenous NST regulates islet cell function by interacting with GPR107 and initiating signaling in glucagon-producing α-cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Glucagon-Secreting Cells/drug effects , Peptide Hormones/pharmacology , Proglucagon/genetics , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line , Glucagon-Secreting Cells/enzymology , Humans , Male , Mice , Peptide Fragments/metabolism , Peptide Hormones/metabolism , Phosphorylation , RNA Interference , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Somatostatin/metabolism , Transfection , Up-Regulation
16.
J Neurosurg ; 124(6): 1752-65, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26587655

ABSTRACT

Nonhemorrhagic neurological deficits are underrecognized symptoms of intracranial dural arteriovenous fistulas (dAVFs) having cortical venous drainage. These symptoms are the consequence of cortical venous hypertension and portend a clinical course with increased risk of neurological morbidity and mortality. One rarely documented and easily misinterpreted type of nonhemorrhagic neurological deficit is progressive dementia, which can result from venous hypertension in the cortex or in bilateral thalami. The latter, which is due to dAVF drainage into the deep venous system, is the less common of these 2 dementia syndromes. Herein, the authors report 4 cases of dAVF with venous drainage into the vein of Galen causing bithalamic edema and rapidly progressive dementia. Two patients were treated successfully with endovascular embolization, and the other 2 patients were treated successfully with endovascular embolization followed by surgery. The radiographic abnormalities and presenting symptoms rapidly resolved after dAVF obliteration in all 4 cases. Detailed descriptions of these 4 cases are presented along with a critical review of 15 previously reported cases. In our analysis of these 19 published cases, the following were emphasized: 1) the clinical and radiographic differences between dAVF-induced thalamic versus cortical dementia syndromes; 2) the differential diagnosis and necessary radiographic workup for patients presenting with a rapidly progressive thalamic dementia syndrome; 3) the frequency at which delays in diagnosis occurred and potentially dangerous and avoidable diagnostic procedures were used; and 4) the rapidity and completeness of symptom resolution following dAVF treatment.


Subject(s)
Central Nervous System Vascular Malformations/complications , Central Nervous System Vascular Malformations/diagnostic imaging , Dementia/diagnostic imaging , Dementia/etiology , Thalamus/diagnostic imaging , Aged , Central Nervous System Vascular Malformations/pathology , Central Nervous System Vascular Malformations/therapy , Cerebral Angiography , Delayed Diagnosis , Dementia/pathology , Dementia/therapy , Diagnosis, Differential , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Thalamus/pathology , Thalamus/surgery , Tomography, X-Ray Computed
17.
PLoS One ; 10(9): e0138958, 2015.
Article in English | MEDLINE | ID: mdl-26402857

ABSTRACT

Dry eye is a common disorder caused by inadequate hydration of the ocular surface that results in disruption of barrier function. The homeostatic protein clusterin (CLU) is prominent at fluid-tissue interfaces throughout the body. CLU levels are reduced at the ocular surface in human inflammatory disorders that manifest as severe dry eye, as well as in a preclinical mouse model for desiccating stress that mimics dry eye. Using this mouse model, we show here that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to the galectin LGALS3, a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. These findings define a fundamentally new mechanism for ocular surface protection and suggest CLU as a biotherapeutic for dry eye.


Subject(s)
Clusterin/therapeutic use , Dry Eye Syndromes/drug therapy , Eye/pathology , Administration, Topical , Animals , Clusterin/pharmacology , Cytoprotection/drug effects , Desiccation , Dry Eye Syndromes/pathology , Eye/drug effects , Female , Galectin 3/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/drug effects , Stress, Physiological/drug effects , Tears/metabolism
18.
Physiology (Bethesda) ; 30(4): 327-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26136546

ABSTRACT

C-peptide is produced, processed, and secreted with insulin, and appears to exert separate but intimately related effects. In this review, we address the existence of the C-peptide receptor, the interaction between C-peptide and insulin, and the potential physiological significance of proinsulin C-peptide.


Subject(s)
C-Peptide/metabolism , Endothelium, Vascular/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Models, Biological , Animals , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Endothelium, Vascular/physiopathology , Humans , Insulin Secretion , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
19.
Peptides ; 72: 192-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26051024

ABSTRACT

The Human Genome Project provided the opportunity to use bioinformatic approaches to discover novel, endogenous hormones. Using this approach we have identified two novel peptide hormones and review here our strategy for the identification and characterization of the hormone, neuronostatin. We describe in this mini-review our strategy for determining neuronostatin's actions in brain, heart and pancreas. More importantly, we detail our deductive reasoning strategy for the identification of a neuronostatin receptor and our progress in establishing the physiological relevance of the peptide.


Subject(s)
Brain/metabolism , Genome, Human , Myocardium/metabolism , Pancreas/metabolism , Peptide Hormones/metabolism , Humans , Peptide Hormones/genetics
20.
Mol Metab ; 4(4): 353-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25830098

ABSTRACT

OBJECTIVE: Non-alcoholic steatohepatitis (NASH) is characterized by hepatic steatosis, inflammation and fibrosis. There are currently no targeted therapies for NASH. We developed a liver-specific LXR inverse agonist, SR9238, which effectively reduces hepatic lipogenesis in models of obesity and hepatic steatosis. We hypothesized that suppression of lipogenesis, which is pathologically elevated in NASH may suppress progression of hepatic steatosis to NASH. METHODS: NASH was induced in B6 V-lep (ob)/J (ob/ob) mice using a custom complete rodent diet (HTF) containing high amounts of trans-fat, fructose, and cholesterol. Once NASH was induced, mice were treated with SR9238 for one month by i.p. injection. Plasma lipid levels and liver health were analyzed by clinical chemistry. QPCR, western blot, and immunohistochemistry were used to assess disease severity. RESULTS: Ob/ob mice are obese and diabetic thus they are commonly used as models for the study of metabolic diseases. These mice quickly developed the NASH phenotype when provided the HTF diet. The mice develop hepatic steatosis, severe hepatic inflammation and fibrosis on the HTF diet. Treatment with SR9238 significantly reduced the severity of hepatic steatosis and most importantly reduced hepatic inflammation and ameliorated hepatic fibrosis. CONCLUSIONS: Here, we demonstrate that an LXR inverse agonist, SR9238, is effective in reduction of hepatic steatosis, inflammation and fibrosis in an animal model of NASH. These results have important implications for the development of therapeutics for treatment NASH in humans.

SELECTION OF CITATIONS
SEARCH DETAIL
...