Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neurosci Bull ; 34(1): 98-108, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28585113

ABSTRACT

Increasing evidence suggests that spinal microglia regulate pathological pain in males. In this study, we investigated the effects of several microglial and astroglial modulators on inflammatory and neuropathic pain following intrathecal injection in male and female mice. These modulators were the microglial inhibitors minocycline and ZVEID (a caspase-6 inhibitor) and the astroglial inhibitors L-α-aminoadipate (L-AA, an astroglial toxin) and carbenoxolone (a connexin 43 inhibitor), as well as U0126 (an ERK kinase inhibitor) and D-JNKI-1 (a c-Jun N-terminal kinase inhibitor). We found that spinal administration of minocycline or ZVEID, or Caspase6 deletion, reduced formalin-induced inflammatory and nerve injury-induced neuropathic pain primarily in male mice. In contrast, intrathecal L-AA reduced neuropathic pain but not inflammatory pain in both sexes. Intrathecal U0126 and D-JNKI-1 reduced neuropathic pain in both sexes. Nerve injury caused spinal upregulation of the astroglial markers GFAP and Connexin 43 in both sexes. Collectively, our data confirmed male-dominant microglial signaling but also revealed sex-independent astroglial signaling in the spinal cord in inflammatory and neuropathic pain.


Subject(s)
Astrocytes/pathology , Microglia/pathology , Neuralgia/pathology , Sex Characteristics , Spinal Cord/pathology , 2-Aminoadipic Acid/toxicity , Animals , Anti-Inflammatory Agents/therapeutic use , Carbenoxolone/pharmacology , Caspase 6/deficiency , Caspase 6/metabolism , Connexin 43/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Mice, Transgenic , Minocycline/therapeutic use , Neuralgia/chemically induced , Neuralgia/drug therapy , Pain Measurement , Phenylurea Compounds/pharmacology , Time Factors
2.
J Neurosci ; 32(22): 7454-65, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649225

ABSTRACT

Huntington's disease (HD) is caused by a mutation in the huntingtin (htt) gene encoding an expansion of glutamine repeats at the N terminus of the Htt protein. Proteolysis of Htt has been identified as a critical pathological event in HD models. In particular, it has been postulated that proteolysis of Htt at the putative caspase-6 cleavage site (at amino acid Asp-586) plays a critical role in disease progression and pathogenesis. However, whether caspase-6 is indeed the essential enzyme that cleaves Htt at this site in vivo has not been determined. To evaluate, we crossed the BACHD mouse model with a caspase-6 knock-out mouse (Casp6(-/-)). Western blot and immunocytochemistry confirmed the lack of caspase-6 protein in Casp6(-/-) mice, regardless of HD genotype. We predicted the Casp6(-/-) mouse would have reduced levels of caspase-6 Htt fragments and increased levels of full-length Htt protein. In contrast, we found a significant reduction of full-length mutant Htt (mHtt) and fragments in the striatum of BACHD Casp6(-/-) mice. Importantly, we detected the presence of Htt fragments consistent with cleavage at amino acid Asp-586 of Htt in the BACHD Casp6(-/-) mouse, indicating that caspase-6 activity cannot fully account for the generation of the Htt 586 fragment in vivo. Our data are not consistent with the hypothesis that caspase-6 activity is critical in generating a potentially toxic 586 aa Htt fragment in vivo. However, our studies do suggest a role for caspase-6 activity in clearance pathways for mHtt protein.


Subject(s)
Aspartic Acid/metabolism , Caspase 6/metabolism , Gene Expression Regulation/genetics , Huntington Disease/metabolism , Huntington Disease/physiopathology , Nerve Tissue Proteins/metabolism , Age Factors , Amino Acids/genetics , Amino Acids/metabolism , Animals , Aspartic Acid/genetics , Body Weight/genetics , Brain/metabolism , Brain/pathology , Caspase 6/deficiency , Cells, Cultured , Corpus Striatum/cytology , Disease Models, Animal , Embryo, Mammalian , Exploratory Behavior/physiology , Female , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Neurons , Proteolysis , RNA, Small Interfering/metabolism , Rotarod Performance Test , Trinucleotide Repeat Expansion/genetics , Ubiquitination/genetics
3.
Hum Mol Genet ; 21(9): 1954-67, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22262731

ABSTRACT

Apoptosis, or programmed cell death, is a cellular pathway involved in normal cell turnover, developmental tissue remodeling, embryonic development, cellular homeostasis maintenance and chemical-induced cell death. Caspases are a family of intracellular proteases that play a key role in apoptosis. Aberrant activation of caspases has been implicated in human diseases. In particular, numerous findings implicate Caspase-6 (Casp6) in neurodegenerative diseases, including Alzheimer disease (AD) and Huntington disease (HD), highlighting the need for a deeper understanding of Casp6 biology and its role in brain development. The use of targeted caspase-deficient mice has been instrumental for studying the involvement of caspases in apoptosis. The goal of this study was to perform an in-depth neuroanatomical and behavioral characterization of constitutive Casp6-deficient (Casp6-/-) mice in order to understand the physiological function of Casp6 in brain development, structure and function. We demonstrate that Casp6-/- neurons are protected against excitotoxicity, nerve growth factor deprivation and myelin-induced axonal degeneration. Furthermore, Casp6-deficient mice show an age-dependent increase in cortical and striatal volume. In addition, these mice show a hypoactive phenotype and display learning deficits. The age-dependent behavioral and region-specific neuroanatomical changes observed in the Casp6-/- mice suggest that Casp6 deficiency has a more pronounced effect in brain regions that are involved in neurodegenerative diseases, such as the striatum in HD and the cortex in AD.


Subject(s)
Caspase 6/physiology , Nerve Degeneration/enzymology , Aging/pathology , Aging/physiology , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Animals , Apoptosis/physiology , Base Sequence , Behavior, Animal/physiology , Brain/enzymology , Brain/pathology , Caspase 6/deficiency , Caspase 6/genetics , Humans , Huntington Disease/enzymology , Huntington Disease/pathology , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/physiology
4.
Biochemistry ; 50(42): 9046-55, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21936563

ABSTRACT

Telomerase is a ribonucleoprotein complex that is essential for persistent cellular proliferation. The catalytic subunit of human telomerase, hTERT, functions as a reverse transcriptase and promotes vitality by maintaining telomeric DNA length. hTERT is tightly regulated with complex but poorly understood positive and negative regulation at several levels including transcription, protein-protein interactions, and post-translation modifications. Because evidence implicates hTERT as an apoptosis inhibitor and because telomerase activity tends to decrease during apoptosis, we hypothesized that hTERT is a caspase substrate leading to down regulation during apoptosis. Caspases are proteases that initiate and execute apoptosis by cleaving target proteins. Indeed, we found that caspases-6 and -7 cleave hTERT during apoptosis in cultured cells. Caspase-6 cleaves at residues D129 and D637, and caspase-7 cleaves at E286 and D628. Three of the caspase cleavage sites are unique motifs. All four caspase motifs appear conserved in TERTs from Old World monkeys and apes, and the caspase-6 sites appear conserved in all primates. The caspase site that cleaves at D129 appears conserved in amniotes. hTERT fragments generated by cleavage were remarkably persistent, lasting hours after caspase activation. These results reveal a new biologically relevant mechanism for telomerase down regulation through caspase-mediated cleavage of hTERT and expand the list of known caspase motifs.


Subject(s)
Caspase 6/chemistry , Caspase 7/chemistry , Catalytic Domain , Telomerase/chemistry , Amino Acid Sequence , Animals , Apoptosis/genetics , Aspartic Acid/genetics , Caspase 6/deficiency , Caspase 6/genetics , Caspase 7/deficiency , Caspase 7/genetics , Down-Regulation/genetics , Glutamic Acid/genetics , HEK293 Cells , Humans , Jurkat Cells , K562 Cells , Molecular Sequence Data , Mutagenesis, Site-Directed , Rabbits , Signal Transduction/genetics , Substrate Specificity/genetics , Telomerase/antagonists & inhibitors , Telomerase/biosynthesis
5.
J Neurosci ; 31(24): 8894-904, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21677173

ABSTRACT

Despite extensive research to develop an effective neuroprotective strategy for the treatment of ischemic stroke, therapeutic options remain limited. Although caspase-dependent death is thought to play a prominent role in neuronal injury, direct evidence of active initiator caspases in stroke and the functional relevance of this activity have not previously been shown. Using an unbiased caspase-trapping technique in vivo, we isolated active caspase-9 from ischemic rat brain within 1 h of reperfusion. Pathogenic relevance of active caspase-9 was shown by intranasal delivery of a novel cell membrane-penetrating highly specific inhibitor for active caspase-9 at 4 h postreperfusion (hpr). Caspase-9 inhibition provided neurofunctional protection and established caspase-6 as its downstream target. The temporal and spatial pattern of expression demonstrates that neuronal caspase-9 activity induces caspase-6 activation, mediating axonal loss by 12 hpr followed by neuronal death within 24 hpr. Collectively, these results support selective inhibition of these specific caspases as an effective therapeutic strategy for stroke.


Subject(s)
Caspase 6/physiology , Enzyme Inhibitors/therapeutic use , Infarction, Middle Cerebral Artery , Inhibitor of Apoptosis Proteins/therapeutic use , Nervous System Diseases , Neurons/pathology , Administration, Intranasal , Aldehydes/pharmacology , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Caspase 6/deficiency , Caspase 9/metabolism , Caspase Inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , In Vitro Techniques , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Nervous System Diseases/pathology , PTEN Phosphohydrolase/chemistry , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/therapeutic use , Rats , Rats, Wistar , Time Factors
6.
J Immunol ; 186(1): 403-10, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098228

ABSTRACT

Alveolar macrophages (AMs) are exposed to respirable microbial particles. Similar to phagocytes in the gastrointestinal tract, AMs can suppress inflammation after exposure to nonpathogenic organisms. IL-1R-associated kinase-M (IRAK-M) is one inhibitor of innate immunity, normally suppressing pulmonary inflammation. During pneumonia, polymorphonuclear neutrophils (PMNs) are recruited by chemotactic factors released by AMs to produce an intense inflammation. We report that intact IRAK-M is strongly expressed in resting human AMs but is cleaved in patients with pneumonia via PMN-mediated induction of caspase-6 (CASP-6) activity. PMN contact is necessary and PMN membranes are sufficient for CASP-6 induction in macrophages. PMNs fail to induce TNF-α fully in macrophages expressing CASP-6 cleavage-resistant IRAK-M. Without CASP-6 expression, PMN stimulation fails to cleave IRAK-M, degrade IκBα, or induce TNF-α. CASP-6(-/-) mice subjected to cecal ligation and puncture have impaired TNF-α production in the lung and decreased mortality. LPS did not induce or require CASP-6 activity demonstrating that TLR2/4 signaling is independent from the CASP-6 regulated pathway. These data define a central role for CASP-6 in PMN-driven macrophage activation and identify IRAK-M as an important target for CASP-6. PMNs de-repress AMs via CASP-6-mediated IRAK-M cleavage. This regulatory system will blunt lung inflammation unless PMNs infiltrate the alveolar spaces.


Subject(s)
Caspase 6/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophage Activation/immunology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/immunology , Neutrophils/enzymology , Neutrophils/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Caspase 6/biosynthesis , Caspase 6/deficiency , Cell Line , Cell Line, Tumor , Coculture Techniques , Female , Humans , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Interleukin-1 Receptor-Associated Kinases/chemistry , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophage Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutagenesis, Site-Directed , Peritonitis/enzymology , Peritonitis/immunology , Peritonitis/mortality , Pneumonia/enzymology , Pneumonia/genetics , Pneumonia/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...