Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Genet ; 20(3): 251-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9806543

ABSTRACT

Loss-of-function mutations in the gene (CSTB) encoding human cystatin B, a widely expressed cysteine protease inhibitor, are responsible for a severe neurological disorder known as Unverricht-Lundborg disease (EPM1). The primary cellular events and mechanisms underlying the disease are unknown. We found that mice lacking cystatin B develop myoclonic seizures and ataxia, similar to symptoms seen in the human disease. The principal cytopathology appears to be a loss of cerebellar granule cells, which frequently display condensed nuclei, fragmented DNA and other cellular changes characteristic of apoptosis. This mouse model of EPM1 provides evidence that cystatin B, a non-caspase cysteine protease inhibitor, has a role in preventing cerebellar apoptosis.


Subject(s)
Apoptosis/genetics , Ataxia/genetics , Cerebellum/pathology , Cystatins/deficiency , Cystatins/genetics , Cysteine Proteinase Inhibitors/deficiency , Cysteine Proteinase Inhibitors/genetics , Epilepsies, Myoclonic/genetics , Amino Acid Sequence , Animals , Ataxia/pathology , Base Sequence , Corneal Opacity/genetics , Cystatin B , Cystatins/physiology , Cysteine Proteinase Inhibitors/physiology , DNA Primers/genetics , Disease Models, Animal , Epilepsies, Myoclonic/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Models, Genetic , Mutation , Phenotype
2.
J Exp Med ; 203(7): 1817-25, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16818671

ABSTRACT

Most memory phenotype (MP) CD44(hi) CD8(+) cells are resting interleukin (IL)-15-dependent cells characterized by high expression of the IL-2/IL-15 receptor beta (CD122). However, some MP CD8(+) cells have a CD122(lo) phenotype and are IL-15 independent. Here, evidence is presented that the CD122(lo) subset of MP CD8(+) cells is controlled largely by major histocompatibility complex (MHC) class I molecules. Many of these cells display surface markers typical of recently activated T cells (CD62L(lo), CD69(hi), CD43(hi), and CD127(lo)) and show a high rate of background proliferation. Cells with this phenotype are highly enriched in common gamma chain-deficient mice and absent from MHC-I(-/-) mice. Unlike CD122(hi) CD8(+) cells, CD122(lo) MP CD8(+) cells survive poorly after transfer to MHC-I(-/-) hosts and cease to proliferate. Although distinctly different from typical antigen-specific memory cells, CD122(lo) MP CD8(+) cells closely resemble the antigen-dependent memory CD8(+) cells found in chronic viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/physiology , Immunologic Memory/physiology , Immunophenotyping , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cystatin C , Cystatins/deficiency , Cystatins/genetics , Histocompatibility Antigens Class I/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/metabolism
3.
J Neurosci Res ; 89(5): 639-49, 2011 May.
Article in English | MEDLINE | ID: mdl-21344476

ABSTRACT

Demyelination coincides with numerous changes of gene expression in the central nervous system (CNS). Cystatin F, which is a papain-like lysosomal cysteine proteinase inhibitor that is normally expressed by immune cells and not in the brain, is massively induced in the CNS during acute demyelination. We found that microglia, which are monocyte/macrophage-lineage cells in the CNS, express cystatin F only during demyelination. By using several demyelinating animal models and the spinal cord tissues from multiple sclerosis (MS) patients, we examined spatiotemporal expression pattern of cystatin F by in situ hybridization and immunohistochemistry. We found that the timing of cystatin F induction matches with ongoing demyelination, and the places with cystatin F expression overlapped with the remyelinating area. Most interestingly, cystatin F induction ceased in chronic demyelination, in which remyelinating ability was lost. These findings demonstrate that the expression of cystatin F indicates the occurrence of ongoing demyelination/remyelination and the absence of cystatin F expression indicates the cessation of remyelination in the demyelinating area.


Subject(s)
Cystatins/biosynthesis , Demyelinating Autoimmune Diseases, CNS/metabolism , Microglia/metabolism , Nerve Fibers, Myelinated/metabolism , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/metabolism , Cells, Cultured , Chronic Disease , Cystatins/deficiency , Cystatins/metabolism , Demyelinating Autoimmune Diseases, CNS/genetics , Demyelinating Autoimmune Diseases, CNS/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Neurologic Mutants , Mice, Transgenic , Microglia/pathology , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/genetics , Recovery of Function/genetics
4.
J Clin Invest ; 104(9): 1191-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545518

ABSTRACT

The pathogenesis of atherosclerosis and abdominal aortic aneurysm involves breakdown of the elastic laminae. Elastolytic cysteine proteases, including cathepsins S and K, are overexpressed at sites of arterial elastin damage, but whether endogenous local inhibitors counterbalance these proteases is unknown. We show here that, whereas cystatin C is normally expressed in vascular wall smooth muscle cells (SMCs), this cysteine protease inhibitor is severely reduced in both atherosclerotic and aneurysmal aortic lesions. Furthermore, increased abdominal aortic diameter among 122 patients screened by ultrasonography correlated inversely with serum cystatin C levels. In vitro, cytokine-stimulated vascular SMCs secrete cathepsins, whose elastolytic activity could be blocked when cystatin C secretion was induced by treatment with TGF-beta(1). The findings highlight a potentially important role for imbalance between cysteine proteases and cystatin C in arterial wall remodeling and establish that cystatin C deficiency occurs in vascular disease.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Arteriosclerosis/metabolism , Cystatins/deficiency , Cysteine Proteinase Inhibitors/deficiency , Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Arteries/metabolism , Arteries/pathology , Arteriosclerosis/pathology , Cells, Cultured , Cystatin C , Cystatins/blood , Cysteine Proteinase Inhibitors/blood , Dose-Response Relationship, Drug , Humans , Immunoblotting , Immunohistochemistry , Interferon-gamma/metabolism , Muscle, Smooth/metabolism , Transforming Growth Factor beta/metabolism
5.
Circ Res ; 96(3): 368-75, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15653570

ABSTRACT

The pathogenesis of atherosclerosis and abdominal aortic aneurysm involves substantial proteolysis of the arterial extracellular matrix. The lysosomal cysteine proteases can exert potent elastolytic and collagenolytic activity. Human atherosclerotic plaques have increased cysteine protease content and decreased levels of the endogenous inhibitor cystatin C, suggesting an imbalance that would favor matrix degradation in the arterial wall. This study tested directly the hypothesis that impaired expression of cystatin C alters arterial structure. Cystatin C-deficient mice (Cyst C-/-) were crossbred with apolipoprotein E-deficient mice (ApoE-/-) to generate cystatin C and apolipoprotein E-double deficient mice (Cyst C-/-ApoE-/-). After 12 weeks on an atherogenic diet, cystatin C deficiency yielded significantly increased tunica media elastic lamina fragmentation, decreased medial size, and increased smooth muscle cell and collagen content in aortic lesions of ApoE-/- mice. Cyst C-/-ApoE-/- mice also showed dilated thoracic and abdominal aortae compared with control ApoE-/- mice, although atheroma lesion size, intimal macrophage accumulation, and lipid core size did not differ between these mice. These findings demonstrate directly the importance of cysteine protease/protease inhibitor balance in dysregulated arterial integrity and remodeling during experimental atherogenesis.


Subject(s)
Aorta, Abdominal/chemistry , Aorta, Abdominal/metabolism , Aorta, Thoracic/chemistry , Aorta, Thoracic/metabolism , Apolipoproteins E/deficiency , Basement Membrane/metabolism , Cystatins/deficiency , Dilatation, Pathologic/genetics , Animals , Aorta, Abdominal/pathology , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/genetics , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Cystatin C , Cysteine Endopeptidases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Tunica Intima/metabolism , Tunica Intima/pathology , Tunica Media/pathology
6.
Retrovirology ; 3: 76, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17078891

ABSTRACT

BACKGROUND: The currently well-established humanized mouse models, namely the hu-PBL-SCID and SCID-hu systems played an important role in HIV pathogenesis studies. However, despite many notable successes, several limitations still exist. They lack multi-lineage human hematopoiesis and a functional human immune system. These models primarily reflect an acute HIV infection with rapid CD4 T cell loss thus limiting pathogenesis studies to a short-term period. The new humanized Rag2-/-gamma c-/- mouse model (RAG-hu) created by intrahepatic injection of CD34 hematopoietic stem cells sustains long-term multi-lineage human hematopoiesis and is capable of mounting immune responses. Thus, this model shows considerable promise to study long-term in vivo HIV infection and pathogenesis. RESULTS: Here we demonstrate that RAG-hu mice produce human cell types permissive to HIV-1 infection and that they can be productively infected by HIV-1 ex vivo. To assess the capacity of these mice to sustain long-term infection in vivo, they were infected by either X4-tropic or R5-tropic HIV-1. Viral infection was assessed by PCR, co-culture, and in situ hybridization. Our results show that both X4 and R5 viruses are capable of infecting RAG-hu mice and that viremia lasts for at least 30 weeks. Moreover, HIV-1 infection leads to CD4 T cell depletion in peripheral blood and thymus, thus mimicking key aspects of HIV-1 pathogenesis. Additionally, a chimeric HIV-1 NL4-3 virus expressing a GFP reporter, although capable of causing viremia, failed to show CD4 T cell depletion possibly due to attenuation. CONCLUSION: The humanized RAG-hu mouse model, characterized by its capacity for sustained multi-lineage human hematopoiesis and immune response, can support productive HIV-1 infection. Both T cell and macrophage tropic HIV-1 strains can cause persistent infection of RAG-hu mice resulting in CD4 T cell loss. Prolonged viremia in the context of CD4 T cell depletion seen in this model mirrors the main features of HIV infection in the human. Thus, the RAG-hu mouse model of HIV-1 infection shows great promise for future in vivo pathogenesis studies, evaluation of new drug treatments, vaccines and novel gene therapy strategies.


Subject(s)
CD4-Positive T-Lymphocytes , Cystatins/deficiency , DNA-Binding Proteins/deficiency , Disease Models, Animal , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Animals , Antigens, CD34/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Cystatin C , HIV Infections/pathology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans , Liver/cytology , Mice , Mice, Inbred BALB C , Transplantation, Heterologous , Viremia/immunology , Viremia/pathology
8.
Ann N Y Acad Sci ; 1044: 236-43, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15958717

ABSTRACT

Although many biologic principles are conserved in mice and humans, species-specific differences exist, for example, in susceptibility and response to pathogens, that often do not allow direct implementation of findings in experimental mice to humans. Research in humans, however, for ethical and practical reasons, is largely restricted to in vitro assays that lack components and the complexity of a living organism. To nevertheless study the human hematopoietic and immune system in vivo, xenotransplantation assays have been developed that substitute human components to small animals. Here, we summarize our recent findings that transplantation of human cord blood CD34(+) cells to newborn Rag2(-/-)gamma(c)(-/-) mice leads to de novo development of major functional components of the human adaptive immune system. These human adaptive immune system Rag2(-/-)gamma(c)(-/-) (huAIS-RG) mice can now be used as a technically straightforward preclinical model to evaluate in vivo human adaptive immune system development as well as immune responses, for example, to vaccines or live infectious pathogens.


Subject(s)
Cystatins/physiology , Immune System/physiology , Animals , Antigens, CD34/immunology , Cell Transplantation , Cystatin C , Cystatins/deficiency , Cystatins/genetics , DNA-Binding Proteins , Fetal Blood/cytology , Humans , Infant, Newborn , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Mutant Strains , Nuclear Proteins , Transplantation, Heterologous
9.
Arterioscler Thromb Vasc Biol ; 24(8): 1359-66, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15178558

ABSTRACT

Atherosclerosis is an inflammatory disease characterized by extensive remodeling of the extracellular matrix architecture of the arterial wall. Although matrix metalloproteinases and serine proteases participate in these pathologic events, recent data from atherosclerotic patients and animals suggest the participation of lysosomal cysteine proteases in atherogenesis. Atherosclerotic lesions in humans overexpress the elastolytic and collagenolytic cathepsins S, K, and L but show relatively reduced expression of cystatin C, their endogenous inhibitor, suggesting a shift in the balance between cysteine proteases and their inhibitor that favors remodeling of the vascular wall. Extracts of human atheromatous tissue show greater elastolytic activity in vitro than do those from healthy donors. The cysteinyl protease inhibitor E64d limits this increased elastolysis, indicating involvement of cysteine proteases in elastin degradation during atherogenesis. Furthermore, inflammatory cytokines augment expression and secretion of active cysteine proteases from cultured monocyte-derived macrophages, vascular smooth muscle cells, and endothelial cells and increase degradation of extracellular elastin and collagen. Cathepsin S-deficient cells or those treated with E64d show significantly impaired elastolytic or collagenolytic activity. Additionally, recent in vivo studies of atherosclerosis-prone, LDL receptor-null mice lacking cathepsin S show participation of this enzyme in the initial infiltration of leukocytes, medial elastic lamina degradation, endothelial cell invasion, and neovascularization, illustrating an important role for cysteine proteases in arterial remodeling and atherogenesis.


Subject(s)
Arteriosclerosis/enzymology , Cystatins/physiology , Cysteine Endopeptidases/physiology , Lysosomes/enzymology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/complications , Arteriosclerosis/pathology , Cell Adhesion , Chemotaxis, Leukocyte/physiology , Cystatin C , Cystatins/deficiency , Cystatins/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/physiology , Cytokines/physiology , Disease Progression , Endothelium, Vascular/enzymology , Enzyme Induction , Extracellular Matrix Proteins/metabolism , Humans , Leukocytes/pathology , Macrophages/enzymology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/physiology , Thromboembolism/etiology
10.
J Neuropathol Exp Neurol ; 61(12): 1085-91, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12484571

ABSTRACT

Progressive myoclonus epilepsy of the Unverricht-Lundborg type (EPM1) is a recessively inherited neurodegenerative disease caused by loss-of-function mutations in the gene encoding cystatin B, a cysteine protease inhibitor. Mice with disruptions in this gene display myoclonic seizures, progressive ataxia, and cerebellar pathology closely paralleling EPMI in humans. To provide further insight into our understanding of EPM1, we report pathological findings in brains from cystatin B-deficient mice. In addition to confirming the loss of cerebellar granular cell neurons by apoptosis, we identified additional neuronal apoptosis in young mutant mice (3-4 months old) within the hippocampal formation and entorhinal cortex. In older mutant mice (16-18 months old), there was also gliosis most marked in the presubiculum and parasubiculum of the hippocampal formation, as well as the entorhinal cortex, neocortex, and striatum. Furthermore, widespread white matter gliosis was also noted, which may be a secondary phenomenon. Within the cerebral cortex, cellular atrophy was a prominent finding in the superficial neurons of the prosubiculum. Finally, we show that mutant mice in either a "seizure-prone" or "seizure-resistant" genetic background display similar neuropathological changes. These findings indicate that neuronal atrophy is an important consequence of cystatin-B deficiency independent of seizure events, suggesting a physiological role for this protein in the maintenance of normal neuronal structure.


Subject(s)
Cystatins/deficiency , Cysteine Proteinase Inhibitors/deficiency , Unverricht-Lundborg Syndrome/pathology , Unverricht-Lundborg Syndrome/physiopathology , Animals , Body Weight , Brain/cytology , Brain/metabolism , Brain/pathology , Cystatin B , Cystatins/physiology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Inbred Strains , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Organ Size , Phenotype
12.
Epilepsia ; 49(4): 557-63, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18028412

ABSTRACT

Unverricht-Lundborg disease (EPM1) is an autosomal recessively inherited neurodegenerative disorder and the most common single cause of progressive myoclonus epilepsy worldwide. Mutations in the gene encoding cystatin B (CSTB), a cysteine protease inhibitor, are responsible for the primary defect underlying EPM1. Here, progress toward understanding the molecular mechanisms in EPM1 is reviewed. We summarize the current knowledge about the CSTB gene and mutations as well as the cellular biology of the CSTB protein with emphasis on data emerging from analysis of EPM1 patients. We shed light on the disease mechanisms of EPM1 based on characterization of the CSTB-deficient mouse model.


Subject(s)
Cystatins/genetics , Unverricht-Lundborg Syndrome/genetics , Animals , Cystatin B , Cystatins/deficiency , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation, Missense/genetics , Promoter Regions, Genetic/genetics
13.
Sex Dev ; 1(6): 363-72, 2007.
Article in English | MEDLINE | ID: mdl-18391548

ABSTRACT

Testatin has been implicated in fetal testis development due to its restricted expression in pre-Sertoli cells immediately after the onset of Sry gene expression. However, testatin knockout mice showed normal testis development and fertility. We investigated the spatial and temporal expression pattern of the Cres/testatin subgroup of genes, including the novel gene Cstl1/Cres4, in fetal mouse gonads and in adult testis, epididymis and ovary. The genes are related to the family 2 cystatins of protease inhibitors. Using real-time PCR and in situ hybridization we could show that 4 subgroup genes, testatin, CstSC, CstTE-1/Cres3 and Cres are expressed in fetal testis. We also confirmed the expression of testatin, CstE2, CstSC, CstTE-1/Cres3, Cres, CstT and Cstl1/Cres4 in adult testis and CstE2, CstTE-1/Cres3, Cres and CstE1/Cres2 in adult epididymis. In testatin knockout animals, the expression of CstE2 was heavily downregulated in adult testis, but not in adult epididymis, compared to wildtype controls. In conclusion, an explanation for the lack of phenotype in testatin knockout mice could be functional redundancy with another member of the Cres/testatin subgroup. The most likely candidate/s would be CstSC, CstTE-1/Cres3 or Cres as they are expressed in the fetal testicular tubules in early testis differentiation together with testatin.


Subject(s)
Cystatins/genetics , Gene Expression , Reproduction/genetics , Testis/growth & development , Amino Acid Sequence , Animals , Cystatins/chemistry , Cystatins/deficiency , Epididymis/chemistry , Female , In Situ Hybridization , Male , Mice , Mice, Knockout , Molecular Sequence Data , Ovary/chemistry , Ovary/growth & development , Polymerase Chain Reaction , RNA, Messenger/analysis , Testis/chemistry , Testis/embryology
14.
Biol Chem ; 388(8): 847-52, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17655504

ABSTRACT

The myristoylated alanine-rich C kinase substrate (MARCKS) is a substrate of protein kinase C (PKC). Besides regulation at the level of gene transcription, MARCKS concentrations within the cell are also regulated by proteolytic cleavage by cathepsins and calpains, which are cysteine proteinases. Stefin B (cystatin B) is an endogenous inhibitor of lysosomal cysteine cathepsins, but not calpains. We have observed increased cleavage of MARCKS in brain and macrophages, but not in liver and kidney extracts of stefin B-deficient mice compared to wild-type mice. Processing of cathepsin B was unaltered in the brain of stefin B-deficient mice and we conclude that increased cleavage of MARCKS could be attributed to the lack of inhibitor.


Subject(s)
Cathepsins/metabolism , Cystatins/deficiency , Cysteine/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Processing, Post-Translational , Animals , Cystatin B , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myristoylated Alanine-Rich C Kinase Substrate , Organ Specificity , Protein Transport , Tissue Extracts/metabolism
15.
Epilepsia ; 48(4): 752-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17319918

ABSTRACT

Unverricht-Lundborg disease (EPM1) has been considered to be an autosomal-recessive disease related with loss of function mutations in the gene encoding cystatin B. Although heterozygous carriers are generally asymptomatic, earlier studies in Finnish EPM1 families have reported minor symptoms together with slight changes in the EEG recordings also in near relatives of patients. Here we tested the hypothesis that EPM1 phenotype is expressed also in heterozygous subjects using 17-month-old cystatin B deficient mice as a model of disease. Western blot analysis demonstrated a 50% decrease in cystatin B expression in the cerebellum of these animals. Heterozygous mice showed significantly impaired rotarod performance and were weaker in the grid test. Also the total seizure-rating score of heterozygous animals was higher than in wild-type mice. The stereological analysis revealed a significant decrease in the number of neurons in cerebral cortex and the granule cell layer of cerebellum. These results suggest that partial decrease in cystatin B expression in heterozygous mice could lead to the development of mild EPM1 phenotype.


Subject(s)
Cystatins/deficiency , Cystatins/genetics , Heterozygote , Unverricht-Lundborg Syndrome/genetics , Animals , Ataxia/diagnosis , Ataxia/genetics , Behavior, Animal/physiology , Brain/pathology , Cell Count , Cell Death/genetics , Cerebellum/pathology , Cerebral Cortex/pathology , Cystatin B , Disease Models, Animal , Female , Finland/epidemiology , Handling, Psychological , Male , Mice , Mice, Mutant Strains , Mutation , Neurons/pathology , Phenotype , Rotarod Performance Test , Seizures/diagnosis , Seizures/genetics , Unverricht-Lundborg Syndrome/epidemiology , Unverricht-Lundborg Syndrome/pathology
16.
Cancer Res ; 66(16): 7899-909, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912163

ABSTRACT

Cystatin M is a secreted inhibitor of lysosomal cysteine proteases. Several lines of evidence indicate that cystatin M is a tumor suppressor important in breast malignancy; however, the mechanism(s) that leads to inactivation of cystatin M during cancer progression is unknown. Inspection of the human cystatin M locus uncovered a large and dense CpG island within the 5' region of this gene (termed CST6). Analysis of cultured human breast tumor lines indicated that cystatin M expression is either undetectable or in low abundance in several lines; however, enhanced gene expression was measured in cells cultured on the DNA demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC). Increased cystatin M expression does not correlate with a cytotoxic response to 5-aza-dC; rather, various molecular approaches indicated that the CST6 gene was aberrantly methylated in these tumor lines as well as in primary breast tumors. Moreover, 60% (12 of 20) of primary tumors analyzed displayed CST6 hypermethylation, indicating that this aberrant characteristic is common in breast malignancies. Finally, preinvasive and invasive breast tumor cells were microdissected from nine archival breast cancer specimens. Of the five tumors displaying CST6 gene methylation, four tumors displayed methylation in both ductal carcinoma in situ and invasive breast carcinoma lesions and reduced expression of cystatin M in these tumors was confirmed by immunohistochemistry. In summary, this study establishes that the tumor suppressor cystatin M is a novel target for epigenetic silencing during mammary tumorigenesis and that this aberrant event can occur before development of invasive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cystatins/genetics , Gene Silencing , 5' Untranslated Regions , Breast Neoplasms/pathology , Cell Line, Tumor , Cystatin M , Cystatins/deficiency , DNA Methylation , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Disease Progression , Female , Humans , Neoplasm Invasiveness , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
17.
Epilepsia ; 47(10): 1650-4, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17054687

ABSTRACT

PURPOSE: Progressive myoclonus epilepsy of the Unverricht-Lundborg type (EPM1) is a rare neurologic disorder, associated with mutations in the Cystatin B (Cstb) gene. Mice lacking Cstb, a cysteine protease inhibitor of the cathepsine family of proteases, provide a mammalian model for EPM1 by displaying similarly progressive ataxia, myoclonic seizures, and neurodegeneration. However, the linkage of Cstb deficit on the molecular level to pathologic features like myoclonic jerks or tonic-clonic seizures has remained unclear. We examined the tryptophan (TRP) metabolism, along the serotonin (5HT) and kynurenine (KYN) pathway in the brain of Cstb-deficient mice, in relation to their possible involvement in the seizure phenotype. METHODS: TRP and its metabolites, along the 5HT and KYN pathways, were assayed in brain tissue by high-pressure liquid chromatography (HPLC) with electrochemical detection. The inverted wire grid and mild handling tests were used for evaluation of ataxia and myoclonic activity. RESULTS: The Cstb-deficient mice had constitutively increased TRP, 5HT, and 5-hydroxyindole acetic acid (5HIAA) levels in the cerebral cortex and cerebellum and increased levels of KYN in the cerebellum. These neurochemical changes were accompanied with ataxia and an apparent myoclonic phenotype among the Cstb-deficient mice. CONCLUSIONS: Our findings suggest that secondary processes (i.e., overstimulation of serotoninergic transmission) on the cellular level, initiated by Cstb deficiency in specific brain regions, may be responsible for the myoclonic/seizure phenotype in EPM1.


Subject(s)
Brain/metabolism , Cystatins/deficiency , Disease Models, Animal , Mutation/genetics , Tryptophan/metabolism , Unverricht-Lundborg Syndrome/genetics , Unverricht-Lundborg Syndrome/metabolism , Animals , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chromatography, High Pressure Liquid , Cystatin B , Cystatins/metabolism , Female , Hydroxyindoleacetic Acid/metabolism , Kynurenine/metabolism , Male , Mice , Phenotype , Serotonin/metabolism , Serotonin/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Tryptophan/genetics
18.
Epilepsia ; 46 Suppl 5: 49-51, 2005.
Article in English | MEDLINE | ID: mdl-15987253

ABSTRACT

PURPOSE: To evaluate the levels of tryptophan and its metabolites along serotonin (5-HT) and kynurenine (KYN) pathways in serum of progressive myoclonus epilepsy (EPM1) patients and cystatin B (CSTB)-deficient mice, a model system for EPM1. METHODS: Tryptophan and its metabolites along serotonin (5-HT) and KYN pathways were determined in serum of EPM1 patients and CSTB-deficient mice by reverse-phase high-pressure liquid chromatography (HPLC) with electrochemical detection. RESULTS: Reduced levels of 5-HT and KYN intermediate metabolite 3-hydroxyanthranilic acid were found in serum of CSTB-deficient mice. A similar trend was found in EPM1 patients. Although tryptophan concentration was reduced in serum of EPM1 patients, no such decrease was observed in CSTB-deficient mice. CONCLUSIONS: The present study demonstrates that tryptophan metabolism along 5-HT and KYN pathways are disrupted in EPM1. Further studies are needed to elucidate the role of KYN pathway in pathogenesis of EPM1.


Subject(s)
3-Hydroxyanthranilic Acid/analysis , Cystatins/deficiency , Disease Models, Animal , Kynurenine/metabolism , Myoclonic Epilepsies, Progressive/blood , Serotonin/blood , Tryptophan/metabolism , 3-Hydroxyanthranilic Acid/metabolism , Adult , Animals , Chromatography, High Pressure Liquid , Cystatin B , Cystatins/metabolism , Female , Humans , Male , Mice , Myoclonic Epilepsies, Progressive/metabolism
19.
Biochem Biophys Res Commun ; 334(4): 1336-42, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16040003

ABSTRACT

In addition to being the conduit for pathogens into hosts, tick saliva contains a broad array of secretory products that facilitate prolonged tick attachment and blood feeding. Proteins found in tick saliva modulate host hemostasis and immune responses. However, it is not clear whether ticks manipulate the immune responses of their hosts by disrupting the antigen-processing pathways of the hosts. Protein secretion into tick saliva from the salivary glands is due to exocytosis of vesicular membrane-bound granular material regulated by SNARE complex proteins. Proteins associated with vesicles (v-SNAREs) are essential components of the exocytotic process. In this study, we assessed the functional significance of synaptobrevin, a SNARE protein, and cystatin, a cysteine protease inhibitor to blood feeding success, in the lone star tick, Amblyomma americanum, using in vivo RNA interference. In separate experiments, tick salivary cystatin and synpatobrevin genes were silenced by injecting adult ticks with 500 ng of dsRNA complementing each gene sequence. Silencing was demonstrated by reduced transcript in midguts and salivary glands. Additionally, disrupting expression of cystatin and synaptobrevin by RNAi reduced the ability of ticks to feed successfully, as demonstrated by feeding inhibition and reduced engorgement weights. Moreover, normal ticks exposed to a rabbit previously exposed to cystatin-silenced ticks exhibited significant resistance to tick feeding. Based on these findings, ticks appear to skillfully evade the host immune system by secreting cystatin, which disrupts normal antigen processing in antigen-presenting cells of hosts.


Subject(s)
Cystatins/metabolism , Feeding Behavior/physiology , Gene Silencing , Membrane Proteins/metabolism , Rabbits/blood , Rabbits/immunology , Ticks/physiology , Animals , Cystatins/deficiency , Cystatins/genetics , Female , Membrane Proteins/deficiency , Membrane Proteins/genetics , Protein Engineering/methods , R-SNARE Proteins , RNA Interference , RNA, Double-Stranded/genetics
20.
Br J Surg ; 88(11): 1472-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11683743

ABSTRACT

BACKGROUND: The cysteine protease inhibitor cystatin C may play a role in the development and progression of abdominal aortic aneurysms (AAAs). METHODS: From a mass screening trial of men aged 65-73 years, 151 small AAAs were followed for a mean of 2.9 years. Of these patients, 142 had serum samples taken to determine the levels of cystatin C, creatinine and C-reactive protein (CRP). RESULTS: Serum cystatin C concentration correlated negatively with AAA size (r = - 0.22 (95 per cent confidence interval (c.i.) - 0.59 to - 0.02)) and annual expansion rate (r = - 0.24 (95 per cent c.i. - 0.75 to - 0.05)), persisting after adjustment for renal function, smoking, diastolic blood pressure, CRP, age and AAA size. Creatinine clearance and CRP did not correlate with size or expansion rate. Thirty-one AAAs had expanded to over 50 mm, when operation was recommended. The serum level of cystatin C was a significant predictor of this occurrence, with a sensitivity and specificity of 61 and 57 per cent respectively. However, initial AAA size had the optimal sensitivity and specificity (both 81 per cent) in this regard. CONCLUSION: Deficiency of cystatin C was associated with increased aneurysm size and expansion rate, possibly due to lack of inhibition of cysteine proteases.


Subject(s)
Aortic Aneurysm, Abdominal/blood , Cystatins/deficiency , Aged , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/physiopathology , Biomarkers/blood , Blood Pressure , Cystatin C , Cystatins/blood , Disease Progression , Humans , Male , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL