Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Ann Afr Med ; 20(2): 138-140, 2021.
Article in English | MEDLINE | ID: mdl-34213482

ABSTRACT

Restrictive cardiomyopathy is an unusual form of cardiomyopathy accounting only for 2%-5% of all pediatric cardiomyopathies. It is mostly idiopathic. Gaucher disease in association with restrictive cardiomyopathy is extremely rare. We herein report a case of cardiac failure in an 8-year-old male child caused by restrictive cardiomyopathy. Pathogenesis of which was attributed to Gaucher disease. In any case of restrictive cardiomyopathy, Gaucher disease should be included in differential diagnosis and investigated accordingly.


Résumé La cardiomyopathie restrictive est une forme inhabituelle de cardiomyopathie qui ne représente que 2 à 5 % de toutes les cardiomyopathies pédiatriques. C'est surtout idiopathique. La maladie de Gaucher associée à une cardiomyopathie restrictive est extrêmement rare. Nous rapportons ici un cas d'insuffisance cardiaque dans un Enfant de sexe masculin de 8 ans causé par une cardiomyopathie restrictive. dont la pathogenèse a été attribuée à la maladie de Gaucher. En tout cas de restriction cardiomyopathie, la maladie de Gaucher doivent être incluses dans le diagnostic différentiel et étudiées en conséquence.


Subject(s)
Cardiomyopathy, Restrictive/complications , Gaucher Disease/complications , Heart Failure/etiology , Cardiomyopathy, Restrictive/diagnostic imaging , Cardiomyopathy, Restrictive/pathology , Child , Echocardiography , Electrocardiography , Enzyme Replacement Therapy , Humans
2.
Cell Rep ; 32(2): 107897, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668248

ABSTRACT

Glioblastoma (GBM) is a malignant brain tumor with few therapeutic options. The disease presents with a complex spectrum of genomic aberrations, but the pharmacological consequences of these aberrations are partly unknown. Here, we report an integrated pharmacogenomic analysis of 100 patient-derived GBM cell cultures from the human glioma cell culture (HGCC) cohort. Exploring 1,544 drugs, we find that GBM has two main pharmacological subgroups, marked by differential response to proteasome inhibitors and mutually exclusive aberrations in TP53 and CDKN2A/B. We confirm this trend in cell and in xenotransplantation models, and identify both Bcl-2 family inhibitors and p53 activators as potentiators of proteasome inhibitors in GBM cells. We can further predict the responses of individual cell cultures to several existing drug classes, presenting opportunities for drug repurposing and design of stratified trials. Our functionally profiled biobank provides a valuable resource for the discovery of new treatments for GBM.


Subject(s)
Glioblastoma/drug therapy , Glioblastoma/pathology , Molecular Targeted Therapy , Precision Medicine , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bortezomib/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Regulatory Networks/drug effects , Genetic Heterogeneity , Genome, Human , Glioblastoma/genetics , Humans , Mice, Inbred BALB C , Mutation/genetics , Proteasome Inhibitors/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
3.
Cancers (Basel) ; 11(4)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31027305

ABSTRACT

New, effective treatment strategies for glioblastomas (GBMs), the most malignant and invasive brain tumors in adults, are highly needed. In this study, we investigated the potential of integrin α10ß1 as a therapeutic target in GBMs. Expression levels and the role of integrin α10ß1 were studied in patient-derived GBM tissues and cell lines. The effect of an antibody-drug conjugate (ADC), an integrin α10 antibody conjugated to saporin, on GBM cells and in a xenograft mouse model was studied. We found that integrin α10ß1 was strongly expressed in both GBM tissues and cells, whereas morphologically unaffected brain tissues showed only minor expression. Partial or no overlap was seen with integrins α3, α6, and α7, known to be expressed in GBM. Further analysis of a subpopulation of GBM cells selected for high integrin α10 expression demonstrated increased proliferation and sphere formation. Additionally, siRNA-mediated knockdown of integrin α10 in GBM cells led to decreased migration and increased cell death. Furthermore, the ADC reduced viability and sphere formation of GBM cells and induced cell death both in vitro and in vivo. Our results demonstrate that integrin α10ß1 has a functional role in GBM cells and is a novel, potential therapeutic target for the treatment of GBM.

5.
Mol Cancer Ther ; 16(8): 1705-1716, 2017 08.
Article in English | MEDLINE | ID: mdl-28716813

ABSTRACT

Curative therapy for medulloblastoma and other pediatric embryonal brain tumors has improved, but the outcome still remains poor and current treatment causes long-term complications. Malignant brain tumors infiltrate the healthy brain tissue and, thus despite resection, cells that have already migrated cause rapid tumor regrowth. Heparan sulfate proteoglycans (HSPG), major components of the extracellular matrix (ECM), modulate the activities of a variety of proteins. The major enzyme that degrades HS, heparanase (HPSE), is an important regulator of the ECM. Here, we report that the levels of HPSE in pediatric brain tumors are higher than in healthy brain tissue and that treatment of pediatric brain tumor cells with HPSE stimulated their growth. In addition, the latent, 65 kDa form of HPSE (that requires intracellular enzymatic processing for activation) enhanced cell viability and rapidly activated the ERK and AKT signaling pathways, before enzymatically active HPSE was detected. The HPSE inhibitor PG545 efficiently killed pediatric brain tumor cells, but not normal human astrocytes, and this compound also reduced tumor cell invasion in vitro and potently reduced the size of flank tumors in vivo Our findings indicate that HPSE in malignant brain tumors affects both the tumor cells themselves and their ECM. In conclusion, HPSE plays a substantial role in childhood brain tumors, by contributing to tumor aggressiveness and thereby represents a potential therapeutic target. Mol Cancer Ther; 16(8); 1705-16. ©2017 AACR.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Glucuronidase/antagonists & inhibitors , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Child , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glucuronidase/metabolism , Humans , Neoplasm Invasiveness , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Protein Kinases/metabolism , RNA, Small Interfering/metabolism , Saponins/pharmacology , Saponins/therapeutic use , Signal Transduction/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
6.
Matrix Biol ; 62: 92-104, 2017 10.
Article in English | MEDLINE | ID: mdl-27890389

ABSTRACT

Heparan sulfate proteoglycans (HSPGs), ubiquitous components of mammalian cells, play important roles in development and homeostasis. These molecules are located primarily on the cell surface and in the pericellular matrix, where they interact with a multitude of macromolecules, including many growth factors. Manipulation of the enzymes involved in biosynthesis and modification of HSPG structures alters the properties of stem cells. Here, we focus on the involvement of heparanase (HPSE), the sole endo-glucuronidase capable of cleaving of HS, in differentiation of embryonic stem cells into the cells of the neural lineage. Embryonic stem (ES) cells overexpressing HPSE (Hpse-Tg) proliferated more rapidly than WT ES cells in culture and formed larger teratomas in vivo. In addition, differentiating Hpse-Tg ES cells also had a higher growth rate, and overexpression of HPSE in NSPCs enhanced Erk and Akt phosphorylation. Employing a two-step, monolayer differentiation, we observed an increase in HPSE as wild-type (WT) ES cells differentiated into neural stem and progenitor cells followed by down-regulation of HPSE as these NSPCs differentiated into mature cells of the neural lineage. Furthermore, NSPCs overexpressing HPSE gave rise to more oligodendrocytes than WT cultures, with a concomitant reduction in the number of neurons. Our present findings emphasize the importance of HS, in neural differentiation and suggest that by regulating the availability of growth factors and, or other macromolecules, HPSE promotes differentiation into oligodendrocytes.


Subject(s)
Glucuronidase/genetics , Glucuronidase/metabolism , Mouse Embryonic Stem Cells/cytology , Oligodendroglia/cytology , Teratoma/pathology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/transplantation , Neurons/cytology , Neurons/metabolism , Oligodendroglia/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Teratoma/genetics , Teratoma/metabolism
7.
Mol Cancer Res ; 14(12): 1243-1253, 2016 12.
Article in English | MEDLINE | ID: mdl-27565180

ABSTRACT

Malignant glioma continues to be fatal, despite improved insight into its underlying molecular mechanisms. The most malignant form, glioblastoma (GBM), is characterized by aberrant activation of receptor tyrosine kinases (RTK) and infiltrative growth. Heparan sulfate proteoglycans (HSPG), integral components of the extracellular matrix of brain tumors, can regulate activation of many RTK pathways. This prompted us to investigate heparanase (HPSE), which cleaves HSPGs, for its role in glioma. This hypothesis was evaluated using tissue microarrays, GBM cells derived from patients, murine in vitro and in vivo models of glioma, and public databases. Downregulation of HPSE attenuated glioma cell proliferation, whereas addition of HPSE stimulated growth and activated ERK and AKT signaling. Using HPSE transgenic and knockout mice, it was demonstrated that tumor development in vivo was positively correlated to HPSE levels in the brain. HPSE also modified the tumor microenvironment, influencing reactive astrocytes, microglia/monocytes, and tumor angiogenesis. Furthermore, inhibition of HPSE reduces tumor cell numbers, both in vitro and in vivo HPSE was highly expressed in human glioma and GBM cell lines, compared with normal brain tissue. Indeed, a correlation was observed between high levels of HPSE and shorter survival of patients with high-grade glioma. In conclusion, these data provide proof-of-concept for anti-HPSE treatment of malignant glioma, as well as novel insights for the development of HPSE as a therapeutic target. IMPLICATIONS: This study aims to target both the malignant brain tumor cells per se and their microenvironment by changing the level of an enzyme, HPSE, that breaks down modified sugar chains on cell surfaces and in the extracellular space. Mol Cancer Res; 14(12); 1243-53. ©2016 AACR.


Subject(s)
Brain Neoplasms/pathology , Cell Nucleus/metabolism , Glioblastoma/pathology , Glucuronidase/metabolism , Up-Regulation , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Progression , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Mice , Prognosis , Signal Transduction , Survival Analysis
8.
Sci Signal ; 8(406): ra125, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26645582

ABSTRACT

Glioblastomas are aggressive astrocytomas characterized by endothelial cell proliferation and abnormal vasculature, which can cause brain edema and increase patient morbidity. We identified the heparin-binding cytokine pleiotrophin as a driver of vascular abnormalization in glioma. Pleiotrophin abundance was greater in high-grade human astrocytomas and correlated with poor survival. Anaplastic lymphoma kinase (ALK), which is a receptor that is activated by pleiotrophin, was present in mural cells associated with abnormal vessels. Orthotopically implanted gliomas formed from GL261 cells that were engineered to produce pleiotrophin showed increased microvessel density and enhanced tumor growth compared with gliomas formed from control GL261 cells. The survival of mice with pleiotrophin-producing gliomas was shorter than that of mice with gliomas that did not produce pleiotrophin. Vessels in pleiotrophin-producing gliomas were poorly perfused and abnormal, a phenotype that was associated with increased deposition of vascular endothelial growth factor (VEGF) in direct proximity to the vasculature. The growth of pleiotrophin-producing GL261 gliomas was inhibited by treatment with the ALK inhibitor crizotinib, the ALK inhibitor ceritinib, or the VEGF receptor inhibitor cediranib, whereas control GL261 tumors did not respond to either inhibitor. Our findings link pleiotrophin abundance in gliomas with survival in humans and mice, and show that pleiotrophin promotes glioma progression through increased VEGF deposition and vascular abnormalization.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/mortality , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Carrier Proteins/metabolism , Cytokines/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/mortality , Anaplastic Lymphoma Kinase , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cytokines/genetics , Disease-Free Survival , Female , Humans , Male , Mice , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Survival Rate , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
FEBS J ; 281(22): 4993-5008, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25284049

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are the main components of the extracellular matrix, where they interact with a large number of physiologically important macromolecules. The sulfation pattern of heparan sulfate (HS) chains determines the interaction potential of the proteoglycans. Enzymes of the biosynthetic and degradation pathways for HS chains are thus important regulators in processes ranging from embryonic development to tissue homeostasis, but also for tumor development. Formation of the nervous system is also critically dependent on intact HSPGs, and several studies have outlined the role of HS in neural induction from embryonic stem cells. High-grade glioma is the most common malignant primary brain tumor among adults, and the outcome is poor. Neural stem cells and glioma stem cells have several common traits, such as sustained proliferation and a highly efficient migratory capacity in the brain. There are also similarities between the neurogenic niche where adult neural stem cells reside, and the tumorigenic niche. These include interactions with the extracellular matrix, and many of the matrix components are deregulated in glioma, e.g. HSPGs and enzymes implementing the biosynthesis and modification of HS. In this article, we will present how HS-regulated pathways are involved in neural differentiation, and discuss their impact on brain development. We will also review and critically discuss the important role of structural modifications of HS in glioma growth and invasion. We propose that targeting invasive mechanisms of glioma cells through modulation of HS structure and HS-mediated pathways may be an attractive alternative to other therapeutic attempts, which so far have only marginally increased survival for glioma patients.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Heparitin Sulfate/physiology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Carcinogenesis/metabolism , Glioma/blood supply , Glioma/pathology , Humans , Neovascularization, Pathologic/metabolism , Neural Stem Cells/physiology , Neurogenesis , Prognosis , Signal Transduction
10.
Cell ; 157(2): 313-328, 2014 04 10.
Article in English | MEDLINE | ID: mdl-24656405

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Piperidines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Heterografts , Humans , Hydroxyquinolines/pharmacology , MAP Kinase Kinase 4/metabolism , Mice , Neoplasm Transplantation , Pinocytosis/drug effects , Vacuoles/metabolism , Zebrafish
11.
J Biol Chem ; 287(14): 10853-62, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22298785

ABSTRACT

Heparan sulfate proteoglycans, present on cell surfaces and in the extracellular matrix, interact with growth factors and morphogens to influence growth and differentiation of cells. The sulfation pattern of the heparan sulfate chains formed during biosynthesis in the Golgi compartment will determine the interaction potential of the proteoglycan. The glucosaminyl N-deacetylase/N-sulfotransferase (NDST) enzymes have a key role during biosynthesis, greatly influencing total sulfation of the heparan sulfate chains. The differentiation potential of mouse embryonic stem cells lacking both NDST1 and NDST2 was studied using in vitro differentiation protocols, expression of differentiation markers, and assessment of the ability of the cells to respond to growth factors. The results show that NDST1 and NDST2 are dispensable for mesodermal differentiation into osteoblasts but necessary for induction of adipocytes and neural cells. Gene expression analysis suggested a differentiation block at the primitive ectoderm stage. Also, GATA4, a primitive endoderm marker, was expressed by these cells. The addition of FGF4 or FGF2 together with heparin rescued the differentiation potential to neural progenitors and further to mature neurons and glia. Our results suggest that the embryonic stem cells lacking both NDST1 and NDST2, expressing a very low sulfated heparan sulfate, can take the initial step toward differentiation into all three germ layers. Except for their potential for mesodermal differentiation into osteoblasts, the cells are then arrested in a primitive ectoderm and/or endoderm stage.


Subject(s)
Amidohydrolases/deficiency , Amidohydrolases/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Heparitin Sulfate/metabolism , Sulfotransferases/deficiency , Sulfotransferases/metabolism , Adipocytes/cytology , Amidohydrolases/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Ectoderm/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/pharmacology , Gene Knockout Techniques , Heparin/pharmacology , Mesoderm/cytology , Mice , Mutation , Osteoblasts/cytology , Signal Transduction/drug effects , Sulfotransferases/genetics
12.
Hear Res ; 284(1-2): 42-51, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22222235

ABSTRACT

More than 29 million adults in the United States have been diagnosed with hearing loss. Interestingly, elevated homocysteine (Hcy) levels, known as hyperhomocysteinemia (HHcy), are also associated with impaired hearing. However, the associated mechanism remains obscure. The collagen receptor such as discoidin domain receptor 1 and matrix metalloproteinase (MMP) play a significant role in inner ear structure and function. We hypothesize that HHcy increases hearing thresholds by compromise in inner ear vasculature resulted from impaired Hcy metabolism, increased oxidative stress, collagen IVa and collagen Ia turnover. The treatment with folic acid (FA) protects elevated hearing thresholds and prevents reduction in vessel density by lowering abundant collagen deposition and oxidative stress in inner ear. To test this hypothesis we employed 8 weeks old male wild type (WT), cystathionine-beta-synthase heterozygote knockout (CBS+/-) mice, WT + FA (0.0057 µg/g/day, equivalent to a 400 µg/70 kg/day human dose in drinking water); and CBS(+/-) +FA. The mice were treated for four weeks. The hearing thresholds were determined by recording the auditory brainstem responses. Integrity of vessels was analyzed by perfusion of horseradish peroxidase (HRP) tracer. Endothelial permeability was assessed, which indicated restoration of HRP leakage by FA treatment. A total Hcy level was increased in stria vascularis (SV) and spiral ligament (SL) of CBS+/- mice which was lowered by FA. Interestingly, FA treatment lowered Col IVa Immunostaining by affecting its turnover. The levels of MMP-2, -9, methylenetetrahydrofolate reductase (MTHFR) and cystathione gamma lyase (CSE) were measured by Western blot analysis. The oxidative stress was high in SV and SL of CBS+/- compared to WT however the treatment with FA lowered oxidative stress in CBS+/- mice. These data suggested that hearing loss in CBS+/- mice was primarily due to leakage in inner ear circulation, also partly by induced collagen imbalance, increase in Hcy and oxidative stress in inner ear.


Subject(s)
Ear, Inner/blood supply , Ear, Inner/drug effects , Folic Acid/pharmacology , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/physiopathology , Adult , Animals , Auditory Threshold/drug effects , Auditory Threshold/physiology , Cochlea/blood supply , Cochlea/drug effects , Cochlea/physiopathology , Collagen Type IV/genetics , Collagen Type IV/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Ear, Inner/physiopathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/etiology , Hearing Loss/genetics , Hearing Loss/physiopathology , Heterozygote , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/genetics , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metabolic Networks and Pathways , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Neurochem Int ; 56(2): 301-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19913585

ABSTRACT

An elevated level of homocysteine (Hcy), known as hyperhomocysteinemia (HHcy), was associated with neurovascular diseases. At physiological levels, hydrogen sulfide (H(2)S) protected the neurovascular system. Because Hcy was also a precursor of hydrogen sulfide (H(2)S), we sought to test whether the H(2)S protected the brain during HHcy. Cystathionine-beta-synthase heterozygous (CBS+/-) and wild type (WT) mice were supplemented with or without NaHS (30 microM/L, H(2)S donor) in drinking water. Blood flow and cerebral microvascular permeability in pial vessels were measured by intravital microscopy in WT, WT+NaHS, CBS-/+ and (CBS-/+)+NaHS-treated mice. The brain tissues were analyzed for matrix metalloproteinase (MMP) and tissue inhibitor of metalloproteinase (TIMP) by Western blot and RT-PCR. The mRNA levels of CBS and cystathionine gamma lyase (CSE, enzyme responsible for conversion of Hcy to H(2)S) genes were measured by RT-PCR. The results showed a significant increase in MMP-2, MMP-9, TIMP-3 protein and mRNA in CBS (-/+) mice, while H(2)S treatment mitigated this increase. Interstitial localization of MMPs was also apparent through immunohistochemistry. A decrease in protein and mRNA expression of TIMP-4 was observed in CBS (-/+) mice. Microscopy data revealed increase in permeability in CBS (-/+) mice. These effects were ameliorated by H(2)S and suggested that physiological levels of H(2)S supplementation may have therapeutic potential against HHcy-induced microvascular permeability, in part, by normalizing the MMP/TIMP ratio in the brain.


Subject(s)
Capillary Permeability/drug effects , Hydrogen Sulfide/pharmacology , Hyperhomocysteinemia/physiopathology , Matrix Metalloproteinase 9/metabolism , Animals , Hyperhomocysteinemia/enzymology , Mice , Reverse Transcriptase Polymerase Chain Reaction
14.
Cell Biochem Biophys ; 55(3): 153-62, 2009.
Article in English | MEDLINE | ID: mdl-19669742

ABSTRACT

Elevated level of homocysteine (Hcy) called hyperhomocysteinemia (HHcy) is one of the major risk factors for chronic heart failure. Although the role of Hcy in cardiac remodeling is documented, the regulatory mechanism involved therein is still nebulous. MicroRNAs (miRNAs) and dicer have been implicated in regulation of cardiovascular diseases. Dicer is the only known enzyme involved in miRNA maturation. We investigated the involvement of dicer and miRNA in Hcy-induced cardiac remodeling. HL-1 cardiomyocytes were cultured in different doses of Hcy. Total RNA was isolated and RT-PCR and real-time PCR was performed for dicer, MMP-2,-9, TIMP-1,-3, and NOX-4. MiRNA microarray was used for analyzing the differential expression of miRNAs. Individual miRNA assay was also done. Western blotting was used to assess the MMP-9 expression in HHcy cardiomyocytes. The RT-PCR results suggest that dicer expression is enhanced in HHcy cardiomyocytes suggesting its involvement in cardiac remodeling caused due to high dose of Hcy. On the other hand, high dose of Hcy increased NOX-4 expression, a marker for oxidative stress. Additionally, HHcy cardiomyocytes showed elevated levels of MMP-2,-9 and TIMP-1,-3, and reduced expression of TIMP-4, suggesting cardiac remodeling due to oxidative stress. The miRNA microarray assay revealed differential expression of 11 miRNAs and among them miR-188 show dramatic downregulation. These findings suggest that dicer and miRNAs especially miR-188 are involved in Hcy-induced cardiac remodeling.


Subject(s)
Homocysteine/pharmacology , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , MicroRNAs , Myocytes, Cardiac/physiology , Regeneration/drug effects , Animals , Blotting, Western , Cell Line , Gene Expression Profiling , Homocysteine/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Ribonuclease III , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
15.
Mol Cell Biochem ; 332(1-2): 215-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19590937

ABSTRACT

Clinical study reports hearing loss in patients with low folic acid (FA) and elevated homocysteine (Hcy). We hypothesize that elevated Hcy induces imbalance in matrix turnover and oxidative stress in cochlea. Cystathione beta-synthase heterozygous knockout mice were used as model for hyperhomocysteinemia. Matrix remodeling induced by Hcy resulted from elevated MMP-2, -9, and -14. MMP-2 and -9 showed elevated gelatinase activity in CBS (+/-) cochlea. Tissue inhibitors of matrix metalloproteinase were significantly lower in CBS (+/-) cochlea. The expression analyses for MMPs and TIMPs were equally represented at protein and mRNA levels. Cochlea of CBS mice showed following structural changes; (1) detachment of tectorial membrane lying on hair cells (2) thinner s. vascularis (3) large fibroblast in spiral ligament. Hcy induced higher protein nitrotyrosination and cytosolic NADPHoxidase subunit p22(phox) in cochlea. It is thus suggested that Hcy induced matrix imbalance, structural changes and oxidative stress in cochlea.


Subject(s)
Cochlea/enzymology , Cochlea/pathology , Extracellular Matrix/enzymology , Hyperhomocysteinemia/enzymology , Hyperhomocysteinemia/pathology , Matrix Metalloproteinases/metabolism , Oxidative Stress , Animals , Blotting, Western , Cystathionine beta-Synthase/physiology , Extracellular Matrix/pathology , Homocysteine/metabolism , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
16.
J Cell Biochem ; 106(1): 119-26, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19021146

ABSTRACT

Increased levels of homocysteine (Hcy), recognized as hyperhomocysteinemia (HHcy), were associated with cardiovascular diseases. There was controversy regarding the detrimental versus cardio protective role of inducible nitric oxide synthase (iNOS) in ischemic heart disease. The aim of this study was to test the hypothesis that the Hcy generated nitrotyrosine by inducing the endothelial nitric oxide synthase, causing endothelial-myocyte (E-M) coupling. To differentiate the role of iNOS versus constitutive nitric oxide synthase (eNOS and nNOS) in Hcy-mediated nitrotyrosine generation and matrix remodeling in cardiac dysfunction, left ventricular (LV) tissue was analyzed from cystathionine beta synthase (CBS) heterozygote knockout, iNOS homozygote knockout, CBS-/+/iNOS-/- double knockout, and wild-type (WT) mice. The levels of nitrotyrosine, MMP-2 and -9 (zymographic analysis), and fibrosis (by trichrome stain) were measured. The endothelial-myocyte function was determined in cardiac rings. In CBS-/+ mice, homocysteine was elevated and in iNOS-/- mice, nitric oxide was significantly reduced. The nitrotyrosine and matrix metalloproteinase-9 (MMP-9) levels were elevated in double knockout and CBS-/+ as compared to WT mice. Although MMP-2 levels were similar in CBS-/+, iNOS-/-, and CBS-/+/iNOS-/-, the levels were three- to fourfold higher than WT. The levels of collagen were similar in CBS-/+ and iNOS-/-, but they were threefold higher than WT. Interesting, the levels of collagen increased sixfold in double knockouts, compared to WT, suggesting synergism between high Hcy and lack of iNOS. Left ventricular hypertrophy was exaggerated in the iNOS-/- and double knockout, and mildly increased in the CBS-/+, compared to WT mice. The endothelial-dependent relaxation was attenuated to the same extent in the CBS-/+ and iNOS-/-, compared to WT, but it was robustly blunted in double knockouts. The results concluded that homocysteine generated nitrotyrosine in the vicinity of endothelium, caused MMP activation and endothelium-myocyte uncoupling. The generation of nitrotyrosine was independent of iNOS.


Subject(s)
Cystathionine beta-Synthase/genetics , Endothelium, Vascular/metabolism , Muscle Cells/metabolism , Nitric Oxide Synthase Type II/genetics , Tyrosine/analogs & derivatives , Animals , Collagen/metabolism , Cystathionine beta-Synthase/metabolism , Genotype , Homocysteine/metabolism , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Phenotype , Tyrosine/metabolism , Ventricular Remodeling
17.
Antioxid Redox Signal ; 11(1): 25-33, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18837652

ABSTRACT

Homocysteine (Hcy) causes cerebrovascular dysfunction by inducing oxidative stress. However, to date, there are no strategies to prevent Hcy-induced oxidative damage. Hcy is an H2S precursor formed from methionine (Met) metabolism. We aimed to investigate whether H2S ameliorated Met-induced oxidative stress in mouse brain endothelial cells (bEnd3). The bEnd3 cells were exposed to Met treatment in the presence or absence of NaHS (donor of H2S). Met-induced cell toxicity increased the levels of free radicals in a concentration-dependent manner. Met increased NADPH-oxidase-4 (NOX-4) expression and mitigated thioredxion-1(Trx-1) expression. Pretreatment of bEnd3 with NaHS (0.05 mM) attenuated the production of free radicals in the presence of Met and protected the cells from oxidative damage. Furthermore, NaHS enhanced inhibitory effects of apocynin, N-acetyl-l-cysteine (NAC), reduced glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), Nomega-nitro-l-arginine methyl ester (L-NAME) on ROS production and redox enzymes levels induced by Met. In conclusion, the administration of H2S protected the cells from oxidative stress induced by hyperhomocysteinemia (HHcy), which suggested that NaHS/H2S may have therapeutic potential against Met-induced oxidative stress.


Subject(s)
Brain/cytology , Endothelial Cells/metabolism , Hydrogen Sulfide/therapeutic use , Methionine/toxicity , Oxidative Stress/drug effects , Acetophenones/metabolism , Acetylcysteine/metabolism , Animals , Catalase/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Formazans/metabolism , Glutathione/metabolism , Homocysteine/biosynthesis , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Mice , Models, Biological , NG-Nitroarginine Methyl Ester/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Tetrazolium Salts/metabolism , Time Factors
18.
Am J Physiol Endocrinol Metab ; 295(5): E1205-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18780770

ABSTRACT

Diabetes and hyperhomocysteinemia (HHcy) are two independent risk factors for glomeruloslerosis and renal insufficiency. Although PPARgamma agonists such as ciglitazone (CZ) are known to modulate diabetic nephropathy, the role of CZ in diabetes-associated HHcy and renopathy is incompletely defined. We tested the hypothesis that induction of PPARgamma by CZ decreases tissue Hcy level; this provides a protective role against diabetic nephropathy. C57BL/6J mice were administered alloxan to create diabetes. Mice were grouped to 0, 1, 10, 12, and 16 wk of treatment; only 12- and 16-wk animals received CZ in drinking water after a 10-wk alloxan treatment. In diabetes, PPARgamma cDNA, mRNA, and protein expression were repressed, whereas an increase in plasma and glomerular Hcy levels was observed. CZ normalized PPARgamma mRNA and protein expression and glomerular level of Hcy, whereas plasma level of Hcy remained unchanged. GFR was dramatically increased at 1-wk diabetic induction, followed by hypofiltration at 10 wk, and was normalized by CZ treatment. This result corroborated with glomerular and preglomerular arteriole histology. A steady-state increase of RVR in diabetic mice became normal with CZ treatment. CZ ameliorated decrease bioavailability of NO in the diabetic animal. Glomerular MMP-2 and MMP-9 activities as well as TIMP-1 expression were increased robustly in diabetic mice and normalized with CZ treatment. Interestingly, TIMP-4 expression was opposite to that of TIMP-1 in diabetic and CZ-treated groups. These results suggested that diabetic nephropathy exacerbated glomerular tissue level of Hcy, and this caused further deterioration of glomerulus. CZ, however, protected diabetic nephropathy in part by activating PPARgamma and clearing glomerular tissue Hcy.


Subject(s)
Diabetic Nephropathies/drug therapy , Homocysteine/metabolism , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/blood , Diabetic Nephropathies/physiopathology , Epinephrine/analogs & derivatives , Epinephrine/pharmacology , Gene Expression/drug effects , Glomerular Filtration Rate/drug effects , Homocysteine/blood , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , Renal Artery/drug effects , Renal Artery/physiology , Renal Circulation/drug effects , Renal Circulation/physiology , Thiazolidinediones/therapeutic use , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Vascular Resistance/drug effects , Vascular Resistance/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Tissue Inhibitor of Metalloproteinase-4
19.
Neurochem Int ; 53(6-8): 214-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18725259

ABSTRACT

An elevated level of Homocysteine (Hcy) is a risk factor for vascular dementia and stroke. Cysthathionine beta Synthase (CBS) gene is involved in the clearance of Hcy. Homozygous individuals for (CBS-/-) die early, but heterozygous for (CBS-/+) survive with high levels of Hcy. The gamma-Amino Butyric Acid (GABA) presents in the central nervous system (CNS) and functions as an inhibitory neurotransmitter. Hcy competes with GABA at the GABA(A) receptor and affects the CNS function. We hypothesize that Hcy causes a decrease in blood flow to the brain due to increase in vascular resistance (VR) because of arterial remodeling in the carotid artery (CA). Blood pressure and blood flow in CA of wild type (WT), CBS-/+, CBS-/+ GABA(A)-/- double knockout, and GABA(A)-/- were measured. CA was stained with trichrome, and the brain permeability was measured. Matrix Metalloproteinases (MMP-2 and MMP-9), tissue inhibitor of metalloproteinase (TIMP-3, TIMP-4), elastin, and collagen-III expression were measured by real-time polymerase chain reaction (RT-PCR). Results showed an increase in VR in CBS-/+/GABA(A)-/-double knockout>CBS-/+/>GABA(A)-/- compared to WT mice. Increased MMP-2, MMP-9, collagen-III and TIMP-3 mRNA levels were found in GABA(A)-/-, CBS-/+, CBS-/+/GABA(A) double knockout compared to WT. The levels of TIMP-4 and elastin were decreased, whereas the levels of MMP-2, MMP-9 and TIMP-3 increased, which indirectly reflected the arterial resistance. These results suggested that Hcy caused arterial remodeling in part, by increase in collagen/elastin ratio thereby increasing VR leading to the decrease in CA blood flow.


Subject(s)
Brain Ischemia/physiopathology , Carotid Arteries/physiopathology , Cerebrovascular Circulation/physiology , Collagen/metabolism , Elastin/metabolism , Homocysteine/metabolism , Animals , Brain Ischemia/chemically induced , Brain Ischemia/metabolism , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cerebrovascular Circulation/drug effects , Collagen Type III/genetics , Collagen Type III/metabolism , Cystathionine beta-Synthase/genetics , Down-Regulation/drug effects , Down-Regulation/physiology , Elastin/genetics , Homocysteine/toxicity , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , Vascular Resistance , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...