Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zigmund Luka is active.

Publication


Featured researches published by Zigmund Luka.


Hepatology | 2007

Loss of the glycine N‐methyltransferase gene leads to steatosis and hepatocellular carcinoma in mice

M. Luz Martínez-Chantar; Mercedes Vazquez-Chantada; Usue Ariz; Nuria Martínez; Marta Varela; Zigmund Luka; Antonieta Capdevila; Juan Rodríguez; Ana M. Aransay; Rune Matthiesen; Heping Yang; Diego F. Calvisi; Manel Esteller; Mario F. Fraga; Shelly C. Lu; Conrad Wagner; José M. Mato

Glycine N‐methyltransferase (GNMT) is the main enzyme responsible for catabolism of excess hepatic S‐adenosylmethionine (SAMe). GNMT is absent in hepatocellular carcinoma (HCC), messenger RNA (mRNA) levels are significantly lower in livers of patients at risk of developing HCC, and GNMT has been proposed to be a tumor‐susceptibility gene for liver cancer. The identification of several children with liver disease as having mutations of the GNMT gene further suggests that this enzyme plays an important role in liver function. In the current study we studied development of liver pathologies including HCC in GNMT‐knockout (GNMT‐KO) mice. GNMT‐KO mice have elevated serum aminotransferase, methionine, and SAMe levels and develop liver steatosis, fibrosis, and HCC. We found that activation of the Ras and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways was increased in liver tumors from GNMT‐KO mice coincidently with the suppression of the Ras inhibitors Ras‐association domain family/tumor suppressor (RASSF) 1 and 4 and the JAK/STAT inhibitors suppressor of cytokine signaling (SOCS) 1–3 and cytokine‐inducible SH2‐protein. Finally, we found that methylation of RASSF1 and SOCS2 promoters and the binding of trimethylated lysine 27 in histone 3 to these 2 genes was increased in HCC from GNMT‐KO mice. Conclusion: These data demonstrate that loss of GNMT induces aberrant methylation of DNA and histones, resulting in epigenetic modulation of critical carcinogenic pathways in mice. (HEPATOLOGY 2008.)


Journal of Inherited Metabolic Disease | 2001

Glycine N-methyltransferase deficiency: A novel inborn error causing persistent isolated hypermethioninaemia

S. H. Mudd; Roberto Cerone; M. C. Schiaffino; A. R. Fantasia; G. Minniti; U. Caruso; R. Lorini; David Watkins; N. Matiaszuk; David S. Rosenblatt; B. Schwahn; Rima Rozen; L. LeGros; M. Kotb; Antonieta Capdevila; Zigmund Luka; James D. Finkelstein; Albert Tangerman; Sally P. Stabler; Robert H. Allen; Conrad Wagner

This paper reports clinical and metabolic studies of two Italian siblings with a novel form of persistent isolated hypermethioninaemia, i.e. abnormally elevated plasma methionine that lasted beyond the first months of life and is not due to cystathionine β-synthase deficiency, tyrosinaemia I or liver disease. Abnormal elevations of their plasma S-adenosylmethionine (AdoMet) concentrations proved they do not have deficient activity of methionine adenosyltransferase I/III. A variety of studies provided evidence that the elevations of methionine and AdoMet are not caused by defects in the methionine transamination pathway, deficient activity of methionine adenosyltransferase II, a mutation in methylenetetrahydrofolate reductase rendering this activity resistant to inhibition by AdoMet, or deficient activity of guanidinoacetate methyltransferase. Plasma sarcosine (N-methylglycine) is elevated, together with elevated plasma AdoMet in normal subjects following oral methionine loads and in association with increased plasma levels of both methionine and AdoMet in cystathionine β-synthase-deficient individuals. However, plasma sarcosine is not elevated in these siblings. The latter result provides evidence they are deficient in activity of glycine N-methyltransferase (GNMT). The only clinical abnormalities in these siblings are mild hepatomegaly and chronic elevation of serum transaminases not attributable to conventional causes of liver disease. A possible causative connection between GNMT deficiency and these hepatitis-like manifestations is discussed. Further studies are required to evaluate whether dietary methionine restriction will be useful in this situation.


Journal of Biological Chemistry | 2009

Glycine N-methyltransferase and regulation of S-adenosylmethionine levels

Zigmund Luka; S. Harvey Mudd; Conrad Wagner

Methylation is a major biological process. It has been shown to be important in formation of compounds such as phosphatidylcholine, creatine, and many others and also participates in epigenetic effects through methylation of histones and DNA. The donor of methyl groups for almost all cellular methylation reactions is S-adenosylmethionine. It seems that the level of S-adenosylmethionine must be regulated in response to developmental stages and metabolic changes, and the enzyme glycine N-methyltransferase has been shown to play a major role in such regulation in mammals. This minireview will focus on the latest discoveries in the elucidation of the mechanism of that regulation.


Transgenic Research | 2006

A glycine N-methyltransferase knockout mouse model for humans with deficiency of this enzyme.

Zigmund Luka; Antonieta Capdevila; José M. Mato; Conrad Wagner

Three human cases having mutations in the glycine N-methyltransferase (GNMT) gene have been reported. This enzyme transfers a methyl group from S-adenosylmethionine (SAM) to glycine to form S-adenosylhomocysteine (SAH) and N-methylglycine (sarcosine) and is believed to be involved in the regulation of methylation. All three cases have mild liver disease but they seem otherwise unaffected. To study this further, gnmt deficient mice were generated for the first time. This resulted in the complete absence of GNMT protein and its activity in livers of homozygous mice. Compared to WT animals the absence of GNMT resulted in up to a 7-fold increase of free methionine and up to a 35-fold increase of SAM. The amount of SAH was significantly decreased (3 fold) in the homozygotes compared to WT. The ratio of SAM/SAH increased from 3 in WT to 300 in livers of homozygous transgenic mice. This suggests a possible significant change in methylation in the liver and other organs where GNMT is expressed.


Proteomics Clinical Applications | 2010

Candidate biomarkers in exosome-like vesicles purified from rat and mouse urine samples

Javier Conde-Vancells; Eva Rodríguez-Suárez; Esperanza Gonzalez; Agustin Berisa; David Gil; Nieves Embade; Mikel Valle; Zigmund Luka; Felix Elortza; Conrad Wagner; Shelly C. Lu; José M. Mato; Juan M. Falcon-Perez

Purpose: There is a compelling clinical imperative to identify discerning molecular biomarkers of hepatic disease in order to inform the diagnosis, prognosis and treatment.


Journal of Inherited Metabolic Disease | 2003

Glycine N-methyltransferase deficiency: A new patient with a novel mutation

P. Augoustides-Savvopoulou; Zigmund Luka; S. Karyda; Sally P. Stabler; Robert H. Allen; K. Patsiaoura; Conrad Wagner; S. H. Mudd

Summary: We report studies of a Greek boy of gypsy origin that show that he has severe deficiency of glycine N-methyltransferase (GNMT) activity due to apparent homozygosity for a novel mutation in the gene encoding this enzyme that changes asparagine-140 to serine. At age 2 years he was found to have mildly elevated serum liver transaminases that have persisted to his present age of 5 years. At age 4 years, hypermethioninaemia was discovered. Plasma methionine concentrations have ranged from 508 to 1049 µmol/L. Several known causes of hypermethioninaemia were ruled out by studies of plasma metabolites: tyrosinaemia type I by a normal plasma tyrosine and urine succinylacetone; cystathionine β-synthase deficiency by total homocysteine of 9.4–12.1 µmol/L; methionine adenosyltransferase I/III deficiency by S-adenosylmethionine (AdoMet) levels elevated to 1643–2222 nmol/L; and S-adenosylhomocysteine (AdoHcy) hydrolase deficiency by normal AdoHcy levels. A normal plasma N-methylglycine concentration in spite of elevated AdoMet strongly suggested GNMT deficiency. Molecular genetic studies identified a missense mutation in the coding region of the boys GNMT gene, which, upon expression, retained only barely detectable catalytic activity. The mild hepatitis-like manifestations in this boy are similar to those in the only two previously reported children with GNMT deficiency, strengthening the likelihood of a causative association. Although his deficiency of GNMT activity may well be more extreme, his metabolic abnormalities are not strikingly greater. Also discussed is the metabolic role of GNMT; several additional metabolite abnormalities found in these patients; and remaining questions about human GNMT deficiency, such as the long-term prognosis, whether other individuals with this defect are currently going undetected, and means to search for such persons.


Hepatology | 2013

Excess S‐adenosylmethionine reroutes phosphatidylethanolamine towards phosphatidylcholine and triglyceride synthesis

Maite Martínez-Uña; Marta Varela-Rey; Ainara Cano; Larraitz Fernández‐Ares; Naiara Beraza; Igor Aurrekoetxea; Ibon Martínez-Arranz; Juan L. García-Rodríguez; Xabier Buqué; Daniela Mestre; Zigmund Luka; Conrad Wagner; Cristina Alonso; Richard H. Finnell; Shelly C. Lu; M. Luz Martínez-Chantar; Patricia Aspichueta; José M. Mato

Methionine adenosyltransferase 1A (MAT1A) and glycine N‐methyltransferase (GNMT) are the primary genes involved in hepatic S‐adenosylmethionine (SAMe) synthesis and degradation, respectively. Mat1a ablation in mice induces a decrease in hepatic SAMe, activation of lipogenesis, inhibition of triglyceride (TG) release, and steatosis. Gnmt‐deficient mice, despite showing a large increase in hepatic SAMe, also develop steatosis. We hypothesized that as an adaptive response to hepatic SAMe accumulation, phosphatidylcholine (PC) synthesis by way of the phosphatidylethanolamine (PE) N‐methyltransferase (PEMT) pathway is stimulated in Gnmt−/− mice. We also propose that the excess PC thus generated is catabolized, leading to TG synthesis and steatosis by way of diglyceride (DG) generation. We observed that Gnmt−/− mice present with normal hepatic lipogenesis and increased TG release. We also observed that the flux from PE to PC is stimulated in the liver of Gnmt−/− mice and that this results in a reduction in PE content and a marked increase in DG and TG. Conversely, reduction of hepatic SAMe following the administration of a methionine‐deficient diet reverted the flux from PE to PC of Gnmt−/− mice to that of wildtype animals and normalized DG and TG content preventing the development of steatosis. Gnmt−/− mice with an additional deletion of perilipin2, the predominant lipid droplet protein, maintain high SAMe levels, with a concurrent increased flux from PE to PC, but do not develop liver steatosis. Conclusion: These findings indicate that excess SAMe reroutes PE towards PC and TG synthesis and lipid sequestration. (Hepatology 2013;58:1296–1305)


Hepatology | 2010

Fatty liver and fibrosis in glycine N-methyltransferase knockout mice is prevented by nicotinamide

Marta Varela-Rey; Nuria Martinez-Lopez; David Fernández-Ramos; Nieves Embade; Diego F. Calvisi; Aswhin Woodhoo; Juan Rodríguez; Mario F. Fraga; Josep Julve; Elisabeth Rodríguez-Millán; Itziar Frades; Luis Torres; Zigmund Luka; Conrad Wagner; Manel Esteller; Shelly C. Lu; M. Luz Martínez-Chantar; José M. Mato

Deletion of glycine N‐methyltransferase (GNMT), the main gene involved in liver S‐adenosylmethionine (SAM) catabolism, leads to the hepatic accumulation of this molecule and the development of fatty liver and fibrosis in mice. To demonstrate that the excess of hepatic SAM is the main agent contributing to liver disease in GNMT knockout (KO) mice, we treated 1.5‐month‐old GNMT‐KO mice for 6 weeks with nicotinamide (NAM), a substrate of the enzyme NAM N‐methyltransferase. NAM administration markedly reduced hepatic SAM content, prevented DNA hypermethylation, and normalized the expression of critical genes involved in fatty acid metabolism, oxidative stress, inflammation, cell proliferation, and apoptosis. More importantly, NAM treatment prevented the development of fatty liver and fibrosis in GNMT‐KO mice. Because GNMT expression is down‐regulated in patients with cirrhosis, and because some subjects with GNMT mutations have spontaneous liver disease, the clinical implications of the present findings are obvious, at least with respect to these latter individuals. Because NAM has been used for many years to treat a broad spectrum of diseases (including pellagra and diabetes) without significant side effects, it should be considered in subjects with GNMT mutations. Conclusion: The findings of this study indicate that the anomalous accumulation of SAM in GNMT‐KO mice can be corrected by NAM treatment leading to the normalization of the expression of many genes involved in fatty acid metabolism, oxidative stress, inflammation, cell proliferation, and apoptosis, as well as reversion of the appearance of the pathologic phenotype. (HEPATOLOGY 2010)


PLOS ONE | 2011

Serum methionine metabolites are risk factors for metastatic prostate cancer progression.

Sally P. Stabler; Tatsuki Koyama; Zhiguo Zhao; Magaly Martinez-Ferrer; Robert H. Allen; Zigmund Luka; Lioudmila V. Loukachevitch; Peter E. Clark; Conrad Wagner; Neil A. Bhowmick

Background Clinical decision for primary treatment for prostate cancer is dictated by variables with insufficient specificity. Early detection of prostate cancer likely to develop rapid recurrence could support neo-adjuvant therapeutics and adjuvant options prior to frank biochemical recurrence. This study compared markers in serum and urine of patients with rapidly recurrent prostate cancer to recurrence-free patients after radical prostatectomy. Based on previous identification of urinary sarcosine as a metastatic marker, we tested whether methionine metabolites in urine and serum could serve as pre-surgical markers for aggressive disease. Methodology/Principal Findings Urine and serum samples (n = 54 and 58, respectively), collected at the time of prostatectomy were divided into subjects who developed biochemical recurrence within 2 years and those who remained recurrence-free after 5 years. Multiple methionine metabolites were measured in urine and serum by GC-MS. The role of serum metabolites and clinical variables (biopsy Gleason grade, clinical stage, serum prostate specific antigen [PSA]) on biochemical recurrence prediction were evaluated. Urinary sarcosine and cysteine levels were significantly higher (p = 0.03 and p = 0.007 respectively) in the recurrent group. However, in serum, concentrations of homocysteine (p = 0.003), cystathionine (p = 0.007) and cysteine (p<0.001) were more abundant in the recurrent population. The inclusion of serum cysteine to a model with PSA and biopsy Gleason grade improved prediction over the clinical variables alone (p<0.001). Conclusions Higher serum homocysteine, cystathionine, and cysteine concentrations independently predicted risk of early biochemical recurrence and aggressiveness of disease in a nested case control study. The methionine metabolites further supplemented known clinical variables to provide superior sensitivity and specificity in multivariable prediction models for rapid biochemical recurrence following prostatectomy.


Laboratory Investigation | 2008

Changes in S-adenosylmethionine and GSH homeostasis during endotoxemia in mice

Kwangsuk Ko; Heping Yang; Mazen Noureddin; Ainhoa Iglesia-Ara; Meng Xia; Conrad Wagner; Zigmund Luka; José M. Mato; Shelly C. Lu

Endotoxemia participates in the pathogenesis of many liver injuries. Lipopolysaccharide (LPS) was shown to inactivate hepatic methionine adenosyltransferase (MAT), the enzyme responsible for S-adenosylmethionine (SAMe) biosynthesis. SAMe treatment was shown to prevent the LPS-induced increase in tumor necrosis factor-α, which may be one of its beneficial effects. SAMe is also an important precursor of glutathione (GSH) and GSH was shown to ameliorate LPS-induced hepatotoxicity. The aims of this work were to examine changes in SAMe and GSH homeostasis during endotoxemia and the effect of SAMe. Mice received SAMe or vehicle pretreatment followed by LPS and were killed up to18 h afterward. Unexpectedly, we found hepatic SAMe level increased 67% following LPS treatment while S-adenosylhomocysteine level fell by 26%, suggesting an increase in SAMe biosynthesis and/or block in transmethylation. The mRNA and protein levels of MAT1A and MAT2A were increased following LPS. However, despite increased MAT1A expression, MAT activity remained inhibited 18 h after LPS. The major methyltransferase that catabolizes hepatic SAMe is glycine N-methyltransferase, whose expression fell by 65% following LPS. Hepatic GSH level fell more than 50% following LPS, coinciding with a comparable fall in the mRNA and protein levels of glutamate-cysteine ligase (GCL) catalytic (GCLC) and modifier subunits (GCLM). SAMe pretreatment prevented the fall in GCLC and attenuated the fall in GCLM expression and GSH level. SAMe pretreatment prevented the LPS-induced increase in plasma alanine transaminases levels but not the LPS-induced increase in hepatic mRNA levels of proinflammatory cytokines. It further enhanced LPS-induced increase in interleukin-10 mRNA level. Taken together, the hepatic response to LPS is to upregulate MAT expression and inhibit SAMe utilization. GSH is markedly depleted largely due to lower expression of GCL. Interestingly, SAMe treatment prevented the fall in GCL and helped to preserve the GSH store and prevent liver injury.

Collaboration


Dive into the Zigmund Luka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shelly C. Lu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

José M. Mato

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Luz Martínez-Chantar

University of the Basque Country

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge