Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Kidney Dis ; 74(1): 132-137, 2019 07.
Article in English | MEDLINE | ID: mdl-30876746

ABSTRACT

In older adults, the most common kidney biopsy diagnoses include pauci-immune crescentic glomerulonephritis, membranous nephropathy, and focal segmental glomerulosclerosis. Recently, investigators described a small series of older patients (aged 66-80 years) with acute kidney injury and a kidney biopsy demonstrating tubular basement membrane (TBM) immune deposits of polytypic immunoglobulin G (IgG) and C3, acute tubular injury, and tubulointerstitial inflammation. They identified a circulating antibody against kidney tubular low-density lipoprotein (LDL) receptor-related protein 2 (LRP2; also known as megalin) in patients' sera and colocalization of LRP2 with IgG in TBM deposits. We present a rare case of anti-LRP2 nephropathy/anti-brush border antibody disease and describe the novel feature of abundant IgG4-positive interstitial plasma cells. Along with the combination of TBM deposits, tubulointerstitial nephritis (TIN), and segmental glomerular subepithelial immune deposits seen in both entities, this newly described feature adds to the morphologic overlap with IgG4-related TIN. Identification of large TBM deposits using light microscopy and IgG staining of apical aspects of proximal tubules using immunofluorescence microscopy can point to the correct diagnosis of anti-LRP2 nephropathy and prompt confirmatory studies. Particularly in older patients with immune complex-mediated TIN who lack clinical, laboratory, radiographic, and/or characteristic histologic features of IgG4-TIN or other autoimmune, infectious, or drug-related injury, a diagnosis of anti-LRP2 nephropathy should be considered.


Subject(s)
Acute Kidney Injury , Glomerulonephritis, Membranous/diagnosis , Kidney Glomerulus , Kidney Tubules , Low Density Lipoprotein Receptor-Related Protein-2/immunology , Methylprednisolone/administration & dosage , Nephritis, Interstitial/diagnosis , Renal Dialysis/methods , Rituximab/administration & dosage , Acute Kidney Injury/diagnosis , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Acute Kidney Injury/therapy , Aged, 80 and over , Antibodies/blood , Biopsy/methods , Diagnosis, Differential , Female , Humans , Immunoglobulin G/immunology , Immunosuppressive Agents/administration & dosage , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Tubules/immunology , Kidney Tubules/pathology , Plasma Cells/immunology , Treatment Outcome
2.
Am J Physiol Cell Physiol ; 316(2): C264-C273, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30462538

ABSTRACT

Cancer-associated thrombosis is a common first presenting sign of malignancy and is currently the second leading cause of death in cancer patients after their malignancy. However, the molecular mechanisms underlying cancer-associated thrombosis remain undefined. In this study, we aimed to develop a better understanding of how cancer cells affect the coagulation cascade and platelet activation to induce a prothrombotic phenotype. Our results show that colon cancer cells trigger platelet activation in a manner dependent on cancer cell tissue factor (TF) expression, thrombin generation, activation of the protease-activated receptor 4 (PAR4) on platelets and consequent release of ADP and thromboxane A2. Platelet-colon cancer cell interactions potentiated the release of platelet-derived extracellular vesicles (EVs) rather than cancer cell-derived EVs. Our data show that single colon cancer cells were capable of recruiting and activating platelets and generating fibrin in plasma under shear flow. Finally, in a retrospective analysis of colon cancer patients, we found that the number of venous thromboembolism events was 4.5 times higher in colon cancer patients than in a control population. In conclusion, our data suggest that platelet-cancer cell interactions and perhaps platelet procoagulant EVs may contribute to the prothrombotic phenotype of colon cancer patients. Our work may provide rationale for targeting platelet-cancer cell interactions with PAR4 antagonists together with aspirin and/or ADP receptor antagonists as a potential intervention to limit cancer-associated thrombosis, balancing safety with efficacy.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/physiology , Colonic Neoplasms/blood , Thrombosis/blood , Blood Platelets/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Cross-Sectional Studies , Humans , Retrospective Studies , Thrombosis/pathology
3.
Neurobiol Dis ; 118: 22-39, 2018 10.
Article in English | MEDLINE | ID: mdl-29940337

ABSTRACT

Intraventricular hemorrhage (IVH) in preterm infants results in reduced proliferation and maturation of oligodendrocyte progenitor cells (OPCs), and survivors exhibit reduced myelination and neurological deficits. Wnt signaling regulates OPC maturation and myelination in a context dependent manner. Herein, we hypothesized that the occurrence of IVH would downregulate Wnt signaling, and that activating Wnt signaling by GSK-3ß inhibition or Wnt3A recombinant human protein (rh-Wnt3A) treatment might promote maturation of OPCs, myelination of the white matter, and neurological recovery in premature rabbits with IVH. These hypotheses were tested in autopsy samples from preterm infants and in a rabbit model of IVH. Induction of IVH reduced expressions of activated ß-catenin, TCF-4, and Axin2 transcription factors in preterm newborns. Both AR-A014418 (ARA) and Wnt-3A treatment activated Wnt signaling. GSK-3ß inhibition by intramuscular ARA treatment accelerated maturation of OPCs, myelination, and neurological recovery in preterm rabbits with IVH compared to vehicle controls. In contrast, intracerebroventricular rh-Wnt3A treatment failed to enhance myelination and neurological function in rabbits with IVH. ARA treatment reduced microglia infiltration and IL1ß expression in rabbits with IVH relative to controls, whereas Wnt3A treatment elevated TNFα, IL1ß, and IL6 expression without affecting microglia density. GSK-3ß inhibition downregulated, while rh-Wnt3A treatment upregulated Notch signaling; and none of the two treatments affected the Sonic-Hedgehog pathway. The administration of ARA or rh-Wnt3A did not affect gliosis. The data suggest that GSK-3ß inhibition promoted myelination by suppressing inflammation and Notch signaling; and Wnt3A treatment failed to enhance myelination because of its pro-inflammatory activity and synergy with Notch signaling. GSK-3ß inhibitors might improve the neurological outcome of preterm infants with IVH.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/biosynthesis , Infant, Premature/metabolism , Nerve Fibers, Myelinated/metabolism , Wnt3A Protein/biosynthesis , Animals , Brain/drug effects , Female , Humans , Infant, Newborn , Male , Nerve Fibers, Myelinated/drug effects , Rabbits , Recombinant Proteins/biosynthesis , Thiazoles/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
4.
JAMA Ophthalmol ; 135(7): 749-760, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28542676

ABSTRACT

Importance: Mutations in genes traditionally associated with syndromic retinal disease are increasingly found to cause nonsyndromic inherited retinal degenerations. Mutations in CLN3 are classically associated with juvenile neuronal ceroid lipofuscinosis, a rare neurodegenerative disease with early retinal degeneration and progressive neurologic deterioration, but have recently also been identified in patients with nonsyndromic inherited retinal degenerations. To our knowledge, detailed clinical characterization of such cases has yet to be reported. Objective: To provide detailed clinical, electrophysiologic, structural, and molecular genetic findings in nonsyndromic inherited retinal degenerations associated with CLN3 mutations. Design, Setting, and Participants: A multi-institutional case series of 10 patients who presented with isolated nonsyndromic retinal disease and mutations in CLN3. Patient ages ranged from 16 to 70 years; duration of follow-up ranged from 3 to 29 years. Main Outcomes and Measures: Longitudinal clinical evaluation, including full ophthalmic examination, multimodal retinal imaging, perimetry, and electrophysiology. Molecular analyses were performed using whole-genome sequencing or whole-exome sequencing. Electron microscopy studies of peripheral lymphocytes and CLN3 transcript analysis with polymerase chain reaction amplification were performed in a subset of patients. Results: There were 7 females and 3 males in this case series, with a mean (range) age at last review of 37.1 (16-70) years. Of the 10 patients, 4 had a progressive late-onset rod-cone dystrophy, with a mean (range) age at onset of 29.7 (20-40) years, and 6 had an earlier onset rod-cone dystrophy, with a mean (range) age at onset of 12.1 (7-17) years. Ophthalmoscopic examination features included macular edema, mild intraretinal pigment migration, and widespread atrophy in advanced disease. Optical coherence tomography imaging demonstrated significant photoreceptor loss except in patients with late-onset disease who had a focal preservation of the ellipsoid zone and outer nuclear layer in the fovea. Electroretinography revealed a rod-cone pattern of dysfunction in 6 patients and were completely undetectable in 2 patients. Six novel CLN3 variants were identified in molecular analyses. Conclusions and Relevance: This report describes detailed clinical, imaging, and genetic features of CLN3-associated nonsyndromic retinal degeneration. The age at onset and natural progression of retinal disease differs greatly between syndromic and nonsyndromic CLN3 disease, which may be associated with genotypic differences.


Subject(s)
DNA/genetics , Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Mutation , Retinal Degeneration/genetics , Visual Acuity , Adolescent , Adult , Aged , DNA Mutational Analysis , Electroretinography , Female , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Molecular Chaperones/metabolism , Ophthalmoscopy , Pedigree , Phenotype , Retinal Degeneration/diagnosis , Retinal Degeneration/metabolism , Tomography, Optical Coherence , Young Adult
5.
Glia ; 64(11): 1987-2004, 2016 11.
Article in English | MEDLINE | ID: mdl-27472419

ABSTRACT

Intraventricular hemorrhage (IVH) leads to reduced myelination and astrogliosis of the white matter in premature infants. No therapeutic strategy exists to minimize white matter injury in survivors with IVH. Epidermal growth factor (EGF) enhances myelination, astrogliosis, and neurologic recovery in animal models of white matter injury. Here, we hypothesized that recombinant human (rh) EGF treatment would enhance oligodendrocyte precursor cell (OPC) maturation, myelination, and neurological recovery in preterm rabbits with IVH. In addition, rhEGF would promote astrogliosis by inducing astroglial progenitor proliferation and GFAP transcription. We tested these hypotheses in a preterm rabbit model of IVH and evaluated autopsy samples from human preterm infants. We found that EGF and EGFR expression were more abundant in the ganglionic eminence relative to the cortical plate and white matter of human infants and that the development of IVH reduced EGF levels, but not EGFR expression. Accordingly, rhEGF treatment promoted proliferation and maturation of OPCs, preserved myelin in the white matter, and enhanced neurological recovery in rabbits with IVH. rhEGF treatment inhibited Notch signaling, which conceivably contributed to OPC maturation. rhEGF treatment contributed to astrogliosis by increasing astroglial proliferation and upregulating GFAP as well as Sox9 expression. Hence, IVH results in a decline in EGF expression; and rhEGF treatment preserves myelin, restores neurological recovery, and exacerbates astrogliosis by inducing proliferation of astrocytes and enhancing transcription of GFAP and Sox9 in pups with IVH. rhEGF treatment might improve the neurological outcome of premature infants with IVH. GLIA 2016;64:1987-2004.


Subject(s)
Astrocytes/drug effects , Cerebral Intraventricular Hemorrhage/complications , Cerebral Intraventricular Hemorrhage/pathology , Epidermal Growth Factor/pharmacology , Gliosis/etiology , Myelin Sheath/metabolism , Age Factors , Animals , Animals, Newborn , Astrocytes/ultrastructure , Brain/embryology , Brain/growth & development , Brain/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cerebral Intraventricular Hemorrhage/chemically induced , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant, Newborn , Infant, Premature , Ki-67 Antigen/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Rabbits , Signal Transduction/physiology
6.
J Neurosci ; 36(11): 3363-77, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985043

ABSTRACT

Intraventricular hemorrhage (IVH) in preterm infants leads to cerebral inflammation, reduced myelination of the white matter, and neurological deficits. No therapeutic strategy exists against the IVH-induced white matter injury. AMPA-kainate receptor induced excitotoxicity contributes to oligodendrocyte precursor cell (OPC) damage and hypomyelination in both neonatal and adult models of brain injury. Here, we hypothesized that IVH damages white matter via AMPA receptor activation, and that AMPA-kainate receptor inhibition suppresses inflammation and restores OPC maturation, myelination, and neurologic recovery in preterm newborns with IVH. We tested these hypotheses in a rabbit model of glycerol-induced IVH and evaluated the expression of AMPA receptors in autopsy samples from human preterm infants. GluR1-GluR4 expressions were comparable between preterm humans and rabbits with and without IVH. However, GluR1 and GluR2 levels were significantly lower in the embryonic white matter and germinal matrix relative to the neocortex in both infants with and without IVH. Pharmacological blockade of AMPA-kainate receptors with systemic NBQX, or selective AMPA receptor inhibition by intramuscular perampanel restored myelination and neurologic recovery in rabbits with IVH. NBQX administration also reduced the population of apoptotic OPCs, levels of several cytokines (TNFα, IL-ß, IL-6, LIF), and the density of Iba1(+) microglia in pups with IVH. Additionally, NBQX treatment inhibited STAT-3 phosphorylation, but not astrogliosis or transcription factors regulating gliosis. Our data suggest that AMPA-kainate receptor inhibition alleviates OPC loss and IVH-induced inflammation and restores myelination and neurologic recovery in preterm rabbits with IVH. Therapeutic use of FDA-approved perampanel treatment might enhance neurologic outcome in premature infants with IVH. SIGNIFICANCE STATEMENT: Intraventricular hemorrhage (IVH) is a major complication of prematurity and a large number of survivors with IVH develop cerebral palsy and cognitive deficits. The development of IVH leads to inflammation of the periventricular white matter, apoptosis and arrested maturation of oligodendrocyte precursor cells, and hypomyelination. Here, we show that AMPA-kainate receptor inhibition by NBQX suppresses inflammation, attenuates apoptosis of oligodendrocyte precursor cells, and promotes myelination as well as clinical recovery in preterm rabbits with IVH. Importantly, AMPA-specific inhibition by the FDA-approved perampanel, which unlike NBQX has a low side-effect profile, also enhances myelination and neurological recovery in rabbits with IVH. Hence, the present study highlights the role of AMPA-kainate receptor in IVH-induced white matter injury and identifies a novel strategy of neuroprotection, which might improve the neurological outcome for premature infants with IVH.


Subject(s)
Brain/metabolism , Hemorrhage/complications , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Receptors, AMPA/metabolism , Recovery of Function/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Calcium Signaling/drug effects , Cerebral Ventricles/physiopathology , Cerebral Ventricles/ultrastructure , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Female , Glycerol/toxicity , Hemorrhage/chemically induced , Hemorrhage/pathology , Humans , Leukoencephalopathies/drug therapy , Leukoencephalopathies/etiology , Male , Nervous System Diseases/drug therapy , Nitriles , Pregnancy , Pyridones/pharmacology , Pyridones/therapeutic use , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , Rabbits , Receptors, AMPA/genetics , Recovery of Function/drug effects
7.
J Neurosci ; 36(3): 872-89, 2016 01 20.
Article in English | MEDLINE | ID: mdl-26791217

ABSTRACT

Intraventricular hemorrhage (IVH) in premature infants results in inflammation, arrested oligodendrocyte progenitor cell (OPC) maturation, and reduced myelination of the white matter. Hyaluronan (HA) inhibits OPC maturation and complexes with the heavy chain (HC) of glycoprotein inter-α-inhibitor to form pathological HA (HC-HA complex), which exacerbates inflammation. Therefore, we hypothesized that IVH would result in accumulation of HA, and that either degradation of HA by hyaluronidase treatment or elimination of HCs from pathological HA by HA oligosaccharide administration would restore OPC maturation, myelination, and neurological function in survivors with IVH. To test these hypotheses, we used the preterm rabbit model of glycerol-induced IVH and analyzed autopsy samples from premature infants. We found that total HA levels were comparable in both preterm rabbit pups and human infants with and without IVH, but HA receptors--CD44, TLR2, TLR4--were elevated in the forebrain of both humans and rabbits with IVH. Hyaluronidase treatment of rabbits with IVH reduced CD44 and TLR4 expression, proinflammatory cytokine levels, and microglia infiltration. It also promoted OPC maturation, myelination, and neurological recovery. HC-HA and tumor necrosis factor-stimulated gene-6 were elevated in newborns with IVH; and depletion of HC-HA levels by HA oligosaccharide treatment reduced inflammation and enhanced myelination and neurological recovery in rabbits with IVH. Hence, hyaluronidase or HA oligosaccharide treatment represses inflammation, promotes OPC maturation, and restores myelination and neurological function in rabbits with IVH. These therapeutic strategies might improve the neurological outcome of premature infants with IVH. Significance statement: Approximately 12,000 premature infants develop IVH every year in the United States, and a large number of survivors with IVH develop cerebral palsy and cognitive deficits. The onset of IVH induces inflammation of the periventricular white matter, which results in arrested maturation of OPCs and myelination failure. HA is a major component of the extracellular matrix of the brain, which regulates inflammation through CD44 and TLR2/4 receptors. Here, we show two mechanism-based strategies that effectively enhanced myelination and neurological recovery in preterm rabbit model of IVH. First, degrading HA by hyaluronidase treatment reduced CD44 and TLR4 expression, proinflammatory cytokines, and microglial infiltration, as well as promoted oligodendrocyte maturation and myelination. Second, intraventricular injection of HA oligosaccharide reduced inflammation and enhanced myelination, conceivably by depleting HC-HA levels.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Ventricles/metabolism , Hyaluronic Acid/biosynthesis , Hyaluronoglucosaminidase/biosynthesis , Oligosaccharides/biosynthesis , Recovery of Function/physiology , Animals , Animals, Newborn , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Cerebral Ventricles/drug effects , Cerebral Ventricles/pathology , Female , Humans , Hyaluronic Acid/administration & dosage , Infant, Newborn , Injections, Intraventricular , Male , Oligosaccharides/administration & dosage , Pregnancy , Rabbits , Recovery of Function/drug effects
8.
J Neurosci ; 33(44): 17232-46, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24174657

ABSTRACT

Intraventricular hemorrhage (IVH) remains a major cause of white matter injury in preterm infants with no viable therapeutic strategy to restore myelination. Maturation of oligodendrocytes and myelination is influenced by thyroid hormone (TH) signaling, which is mediated by TH receptor α (TRα) and TRß. In the brain, cellular levels of TH are regulated by deiodinases, with deiodinase-2 mediating TH activation and deiodinase-3 TH inactivation. Therefore, we hypothesized that IVH would decrease TH signaling via changes in the expression of deiodinases and/or TRs, and normalization of TH signaling would enhance maturation of oligodendrocytes and myelination in preterm infants with IVH. These hypotheses were tested using both autopsy materials from human preterm infants and a rabbit model of IVH. We found that deiodinase-2 levels were reduced, whereas deiodinase-3 levels were increased in brain samples of both humans and rabbits with IVH compared with controls without IVH. TRα expression was also increased in human infants with IVH. Importantly, treatment with TH accelerated the proliferation and maturation of oligodendrocytes, increased transcription of Olig2 and Sox10 genes, augmented myelination, and restored neurological function in pups with IVH. Consistent with these findings, the density of myelinating oligodendrocytes was almost doubled in TH-treated human preterm infants compared with controls. Thus, in infants with IVH the combined elevation in deiodinase-3 and reduction in deiodinase-2 decreases TH signaling that can be worsened by an increase in unliganded TRα. Given that TH promotes neurological recovery in IVH, TH treatment might improve the neurodevelopmental outcome of preterm infants with IVH.


Subject(s)
Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/physiopathology , Cerebral Ventricles/physiopathology , Myelin Sheath/physiology , Recovery of Function/physiology , Thyroxine/physiology , Animals , Animals, Newborn , Cerebral Ventricles/physiology , Disease Models, Animal , Double-Blind Method , Female , Humans , Infant, Newborn , Infant, Premature , Male , Myelin Sheath/pathology , Rabbits , Thyroxine/therapeutic use , Treatment Outcome
9.
Anal Quant Cytopathol Histpathol ; 35(6): 306-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24617036

ABSTRACT

OBJECTIVE: To analyze whether immunogold labeling density for basic fibroblastic growth factor in granules is compatible with the activation stage of mast cells. STUDY DESIGN: Cytoplasmic features and granules of 46 mast cells were examined at the ultrastructural level. The cells were classified according to their activation stage, namely, whether resting, initially activated, fully degranulated or piecemeal degranulated. Granules were classified as electron lucent, moderate or dense granules. Gold particles per secretory granules in the cells were counted. Recently described quantitative analysis techniques were used for evaluation. RESULTS: There is a statistically meaningful relationship between the activation stage of mast cells and their immunogold labeling density. The number of different types of granules encountered in a cell depends on the type of the cell. The distribution of gold particles among the secretory granules depends upon the cell. The type of granule does not have an individual effect on the number of particles, as indicated by an overall statistical analysis of granules, cells and their interaction effects. CONCLUSION: A count of gold particles in the cells can be used as a strong biological indicator. Therefore the number of gold particles might be very useful for comparative studies related to the secretion of this growth factor under different conditions.


Subject(s)
Cell Degranulation/physiology , Fibroblast Growth Factor 2/metabolism , Mast Cells/metabolism , Mast Cells/ultrastructure , Animals , Cytoplasmic Granules/ultrastructure , Immunohistochemistry , Rats
10.
J Neurosci ; 31(34): 12068-82, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865450

ABSTRACT

Intraventricular hemorrhage (IVH) results in neural cell death and white matter injury in premature infants. No therapeutic strategy is currently available against this disorder. Bone morphogenetic protein (BMP) signaling suppresses oligodendrocyte development through basic-helix-loop-helix (bHLH) transcription factors and promotes astrocytosis. Therefore, we hypothesized that IVH in premature newborns initiates degeneration and maturation arrest of oligodendrocyte lineage and that BMP inhibition alleviates hypomyelination, gliosis, and motor impairment in the survivors of IVH. To test the hypotheses, a rabbit model of IVH was used in which premature rabbit pups (E29) are treated with intraperitoneal glycerol at 2 h of age to induce IVH; and the pups with IVH exhibit hypomyelination and gliosis at 2 weeks of postnatal age. Maturation of oligodendrocyte lineage was evaluated by specific markers, and the expression of bHLH transcription factors was assessed. BMP levels were measured in both premature rabbit pups and autopsy materials from premature infants. Recombinant human noggin was used to suppress BMP action; and neurobehavioral performance, myelination and gliosis were assessed in noggin-treated pups compared with untreated controls. We found that IVH resulted in apoptosis and reduced proliferation of oligodendrocyte progenitors, as well as arrested maturation of preoligodendrocytes in rabbits. BMP4 levels were significantly elevated in both rabbit pups and human premature infants with IVH compared with controls. Importantly, BMP inhibition by recombinant human noggin restored the levels of phospho-Smad1/5/8, Olig2 transcription factor, oligodendrocyte maturation, myelination, astrocyte morphology, and motor function in premature pups with IVH. Hence, BMP inhibition might enhance neurological recovery in premature infants with IVH.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/physiology , Cerebral Hemorrhage/drug therapy , Recovery of Function/drug effects , Animals , Animals, Newborn , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Disease Models, Animal , Female , Humans , Infant, Newborn , Infant, Premature/physiology , Lateral Ventricles/blood supply , Lateral Ventricles/drug effects , Lateral Ventricles/physiopathology , Male , Pregnancy , Rabbits , Recovery of Function/physiology
11.
J Mol Histol ; 42(5): 417-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21830143

ABSTRACT

Basic fibroblast growth factor (bFGF) is one of the most potent angiogenic factors. Unlike many other growth factors, bFGF lacks a classic peptide sequence for its secretion. Recent studies suggest that there is an unconventional secretory pathway for this growth factor. The aim of this study was to identify the specific location of bFGF in endothelial cells and to find morphologic evidences concerning its synthesis, storage and release from endothelial cells. The capillaries in hippocampus, adrenal gland, kidney, peripheral nerves as well as the vessels in connective tissues were analysed by using immunogold labeling techniques at electron microscope level. Results show that endogenous bFGF is mainly located in the nuclei of endothelial cells. Slight immunoreactivity is found in the cytoplasm. Immunolabeling is notably absent in pinocytotic vesicles, Golgi complexes, endoplasmic reticulum, nuclear membrane and intercellular junctions. These results provide morphologic evidence suggesting that endothelial cells might export bFGF via unique cellular pathways that are clearly distinct from classical signal peptide mediated secretion and/or release of this protein could be directly through mechanically induced disruptions of these cells. The current study support the recent hypothesis related with unconventional secretory pathway for bFGF as some other "cargo" proteins.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/ultrastructure , Animals , Endothelial Cells/cytology , Organ Specificity , Protein Transport , Rats , Reproducibility of Results , Staining and Labeling
12.
Food Chem Toxicol ; 49(3): 571-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20538033

ABSTRACT

Konzo is a self-limiting central motor-system disease associated with food dependency on cassava and low dietary intake of sulfur amino acids (SAA). Under conditions of SAA-deficiency, ingested cassava cyanogens yield metabolites that include thiocyanate and cyanate, a protein-carbamoylating agent. We studied the physical and biochemical modifications of rat serum and spinal cord proteins arising from intoxication of young adult rats with 50-200mg/kg linamarin, or 200mg/kg sodium cyanate (NaOCN), or vehicle (saline) and fed either a normal amino acid- or SAA-deficient diet for up to 2 weeks. Animals under SAA-deficient diet and treatment with linamarin or NaOCN developed hind limb tremors or motor weakness, respectively. LC/MS-MS analysis revealed differential albumin carbamoylation in animals treated with NaOCN, vs. linamarin/SAA-deficient diet, or vehicle. 2D-DIGE and MALDI-TOF/MS-MS analysis of the spinal cord proteome showed differential expression of proteins involved in oxidative mechanisms (e.g. peroxiredoxin 6), endocytic vesicular trafficking (e.g. dynamin 1), protein folding (e.g. protein disulfide isomerase), and maintenance of the cytoskeleton integrity (e.g. α-spectrin). Studies are needed to elucidate the role of the aformentioned modifications in the pathogenesis of cassava-associated motor-system disease.


Subject(s)
Manihot/chemistry , Motor Neuron Disease/chemically induced , Motor Neuron Disease/metabolism , Nitriles/toxicity , Amino Acid Sequence , Amino Acids, Sulfur/deficiency , Animals , Biomarkers , Cyanates/analysis , Diet , Male , Motor Neuron Disease/physiopathology , Proteome/analysis , Rats , Rats, Nude , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thiocyanates/analysis , Two-Dimensional Difference Gel Electrophoresis
13.
Toxicol Sci ; 107(2): 482-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19033394

ABSTRACT

Neuroprotein changes in the spinal cord of rodents with aliphatic gamma-diketone axonopathy induced by 2,5-hexanedione (2,5-HD) are compared with those reported previously in aromatic gamma-diketone-like axonopathy induced by 1,2-diacetylbenzene (1,2-DAB). Sprague-Dawley rats were treated intraperitoneally with 500 mg/kg/day 2,5-HD, equimolar doses of 2,3-hexanedione (negative control), or an equivalent amount of saline containing 50% dimethyl sulfoxide (vehicle), 5 days a week, for 3 weeks. Analysis of the lumbosacral proteome by 2-dimensional differential in-gel electrophoresis and matrix-assisted laser desorption ionization time-of-flight/tandem mass spectrometry revealed 34 proteins markedly modified by 2,5-HD of which neurofilament triplet L, gelsolin, protein disulfide isomerase, glutathione S-transferase, nicotinamide adenine dinucleotide (reduced) dehydrogenase 1 alpha, pyruvate kinase, and fatty acid synthase were also modified by 1,2-DAB. The expression of proteins involved in maintaining the physical integrity of the cytoskeleton or controlling the redox and protein-folding mechanisms was reduced, whereas that of proteins supporting energy metabolism was mainly increased. The similarity of the neuroproteomic patterns of 2,5-HD and 1,2-DAB axonopathy suggests common biomarkers and/or mechanisms of neurotoxicity associated with exposure to their parent chemicals, namely the industrial solvents n-hexane and 1,2-diethylbenzene, respectively.


Subject(s)
Axons/pathology , Hexanes/toxicity , Hexanones/toxicity , Nerve Tissue Proteins/drug effects , Neurotoxicity Syndromes/pathology , Neurotoxins/toxicity , Acetophenones/toxicity , Amino Acids/metabolism , Animals , Biomarkers , Cell Shape/drug effects , Electrophoresis, Polyacrylamide Gel , Hexanes/pharmacokinetics , Isoelectric Focusing , Male , Neurofilament Proteins/metabolism , Proteomics , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
14.
Toxicol Sci ; 105(1): 134-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18502740

ABSTRACT

Motor neuron axonopathy in diseases such as amyotrophic lateral sclerosis can be modeled and probed with neurotoxic chemicals that induce similar patterns of pathology, such as axonal spheroids that represent focal accumulation of anterogradely transported neurofilaments (NFs). The aromatic gamma-diketone-like 1,2-diacetylbenzene (1,2-DAB), but not its 1,3-DAB isomer, reacts with epsilon-amino- or sulfyhydryl groups of (neuro)proteins, forms adducts, and causes NFs to accumulate at proximal sites of elongate motor axons. We exploit the protein-reactive properties of neurotoxic 1,2-DAB versus the nonprotein-reactive properties of non-neurotoxic 1,3-DAB to unveil proteomic changes associated with this type of pathology. We used two-dimensional differential in-gel electrophoresis (2D-DIGE), matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometry to analyze the lumbosacral spinal cord proteome of adult Sprague-Dawley rats treated systemically with 20 mg/kg/day 1,2-DAB, equimolar dose of 1,3-DAB, or equivalent volume of vehicle (saline containing 2% acetone), 5 days a week, for 2 weeks. 1,2-DAB significantly altered the expression of protein disulfide isomerase, an enzyme involved in protein folding, and gelsolin, an actin-capping and -severing protein. Modifications of these two proteins have been incriminated in the pathogenesis of nerve fiber degeneration. Protein-reactive and neurotoxic 1,2-DAB appears to be excellent tool to dissect mechanisms of nerve fiber (axon) degeneration.


Subject(s)
Acetophenones/toxicity , Axons/drug effects , Benzene Derivatives/metabolism , Gelsolin/analysis , Protein Disulfide-Isomerases/analysis , Spinal Cord/drug effects , Animals , Axons/pathology , Body Weight/drug effects , Electrophoresis, Gel, Two-Dimensional , Male , Proteomics , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spinal Cord/chemistry , Spinal Cord/pathology
15.
Toxicol Sci ; 99(1): 326-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17562736

ABSTRACT

Gene expression profiling is a widely used technique with data from the majority of published microarray studies being publicly available. These data are being used for meta-analyses and in silico discovery; however, the comparability of toxicogenomic data generated in multiple laboratories has not been critically evaluated. Using the power of prospective multilaboratory investigations, seven centers individually conducted a common toxicogenomics experiment designed to advance understanding of molecular pathways perturbed in liver by an acute toxic dose of N-acetyl-p-aminophenol (APAP) and to uncover reproducible genomic signatures of APAP-induced toxicity. The nonhepatotoxic APAP isomer N-acetyl-m-aminophenol was used to identify gene expression changes unique to APAP. Our data show that c-Myc is induced by APAP and that c-Myc-centered interactomes are the most significant networks of proteins associated with liver injury. Furthermore, sources of error and data variability among Centers and methods to accommodate this variability were identified by coupling gene expression with extensive toxicological evaluation of the toxic responses. We show that phenotypic anchoring of gene expression data is required for biologically meaningful analysis of toxicogenomic experiments.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Gene Expression Profiling/methods , Gene Expression/drug effects , Genomics/methods , Liver/drug effects , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Endpoint Determination , Genomic Islands , Isomerism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Reproducibility of Results , Salivary alpha-Amylases , Transcription Factors/biosynthesis , Transcription Factors/genetics
16.
J Cereb Blood Flow Metab ; 27(2): 334-47, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16757980

ABSTRACT

Ischemia is implicated in periventricular white matter injury (PWMI), a lesion associated with cerebral palsy. PWMI features selective damage to early cells of the oligodendrocyte lineage, a phenomenon associated with glutamate receptor activation. We have investigated the distribution of glutamate in rat periventricular white matter at post-natal day 7. Immuno-electron microcopy was used to identify O4(+) oligodendroglia in control rats, and a similar approach was employed to stain glutamate in these cells before and after 90 mins of hypoxia-ischemia. This relatively brief period of hypoxia-ischemia produced mild cell injury, corresponding to the early stages of PWMI. Glutamate-like reactivity was higher in oligodendrocytes than in other cell types (2.13+/-0.25 counts/microm(2)), and declined significantly during hypoxia-ischemia (0.93+/-0.15 counts/microm(2): P<0.001). Astrocytes had lower glutamate levels (0.7+/-0.07 counts/microm(2)), and showed a relatively small decline during hypoxia-ischemia. Axonal regions contained high levels of glutamate (1.84+/-0.20 counts/microm(2)), much of which was lost during hypoxia-ischemia (0.72+/-0.20 counts/microm(2): P>0.001). These findings suggest that oligodendroglia and axons are the major source of extracellular glutamate in developing white matter during hypoxia-ischemia, and that astrocytes fail to accumulate the glutamate lost from these sources. We also examined glutamate levels in the choroid plexus. Control glutamate levels were high in both choroid epithelial (1.90+/-0.20 counts/microm(2)), and ependymal cells (2.20+/-0.28 counts/microm(2)), and hypoxia-ischemia produced a large fall in ependymal glutamate (0.97+/-0.08 counts/microm(2): P>0.001). The ependymal cells were damaged by the insult and represent a further potential source of glutamate during ischemia.


Subject(s)
Animals, Newborn/physiology , Axons/metabolism , Brain Chemistry/physiology , Glutamic Acid/metabolism , Hypoxia-Ischemia, Brain/metabolism , Oligodendroglia/metabolism , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Axons/ultrastructure , Cell Lineage/physiology , Choroid/metabolism , Ependyma/metabolism , Microscopy, Immunoelectron , Oligodendroglia/ultrastructure , Rats , Rats, Sprague-Dawley , Terminology as Topic
17.
Acta Neuropathol ; 112(3): 317-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16874529

ABSTRACT

The chromogenic and neurotoxic gamma-diketone 1,2-diacetylbenzene (1,2-DAB), but not its isomer 1,3-DAB, induces blue discoloration of tissues and urine, clustering of axonal microtubules and proximal neurofilament-filled axonal swellings in rodents. The remarkable chromogenic property of 1,2-DAB, a monocyclic aromatic hydrocarbon, arises from reaction with lysine residues of proteins and formation of dimeric and polymeric derivatives. Tetralin, a dicyclic solvent structurally related to acetyl ethyl tetramethyl tetralin, a chromogenic and neurotoxic agent, reportedly induces excretion of green urine, and causes neurological disturbances in humans. Monocyclic aromatic 1,2,4-triethylbenzene (1,2,4-TEB), but not its isomer 1,3,5-TEB, is also reportedly chromogenic and induces neurophysiological deficits in rodents consistent with axonal neuropathy, but without neuropathological confirmation. We treated 12-week-old C57Bl/6 mice by gavage with 300, 600, or 900 mg/kg/day 1,2,4-TEB, or equivalent doses of 1,3,5-TEB, 3 days/week, for up to 12 weeks, or intraperitoneally with 400 mg/kg/day tetralin, or 50 or 100 mg/kg/day of its alpha-tetralol analogue, 5 days/week, for up to 5 weeks. Animals treated with 1,2,4-TEB, but not 1,3,5-TEB, tetralin or alpha-tetralol, developed hind limb weakness, excreted greenish urine, and showed 1,2-DAB-like neuropathology. These findings support the hypothesis that 1,2-spaced ethyl (or acetyl) moieties on a benzene ring of hydrocarbons are required for hydrocarbons to induce chromogenic changes and proximal giant neurofilamentous axonopathy. Key molecular targets of these compounds likely reside in the axon where they serve to maintain normal cytoskeletal organization.


Subject(s)
Axons/pathology , Hydrocarbons, Aromatic/chemistry , Hydrocarbons, Aromatic/toxicity , Neurotoxicity Syndromes/pathology , Polycyclic Compounds/chemistry , Polycyclic Compounds/toxicity , Animals , Behavior, Animal/drug effects , Benzene Derivatives/toxicity , Body Weight , Isomerism , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Motor Activity/drug effects , Neurotoxicity Syndromes/psychology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/pathology , Species Specificity , Spinal Cord/pathology , Structure-Activity Relationship , Tetrahydronaphthalenes/toxicity , Tetralones/toxicity
18.
Exp Eye Res ; 80(6): 877-84, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15939045

ABSTRACT

The effect of age on the number and morphology of optic nerve axons in adult Brown Norway rats (5-31 months old) (n=29) was examined using transmission electron microscopy (TEM). By manually counting every axon in areas representing 60% of the optic nerve cross-section, we found a significant negative correlation between age and axon count (R(2)=0.18, P<0.05). However, when the oldest animals were omitted, the relationship was no longer statistically significant. Simultaneously, the proportion of spontaneously degenerating axons increased at an exponential rate (R(2)=0.79, P<0.05), with significantly more degeneration in the 31-month group than in 5-month-old animals (ANOVA, P<0.05). This study demonstrates, using quantitative TEM methods, that optic nerve axonal numbers are relatively constant throughout the majority of the adult life of the Brown Norway rat, an increasingly popular strain for glaucoma research. Total axonal loss with aging is substantially less than that reported for other strains. The reduction in axonal numbers and the rate of axonal degeneration do not appear significantly altered until the last few months of life, failing to support some studies that have concluded that optic nerve axon loss in adult rats is linear. However, they do agree with other studies in the rat, and a similar study performed in non-human primate eyes, that concluded that aging changes in the optic nerve and retina follow a complex pattern. Therefore, the impact of animal age must be considered when modeling the course and pathophysiology of experimental glaucomatous optic nerve damage in rats.


Subject(s)
Aging/physiology , Axons/physiology , Optic Nerve/physiopathology , Animals , Cell Count , Intraocular Pressure/physiology , Microscopy, Electron/methods , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neuroglia/physiology , Optic Nerve/pathology , Rats , Rats, Inbred BN
SELECTION OF CITATIONS
SEARCH DETAIL
...