422 results on '"Kruger, Warren D."'
Search Results
152. Hyperhomocysteinemia Potentiates Hyperglycemia-Induced Inflammatory Monocyte Differentiation and Atherosclerosis.
- Author
-
Pu Fang, Daqing Zhang, Zhongjian Cheng, Chenghui Yan, Xiaohua Jiang, Kruger, Warren D., Shu Meng, Arning, Erland, Bottiglieri, Teodoro, Choi, Eric T., Yaling Han, Xiao-feng Yang, and Hong Wang
- Subjects
HYPERHOMOCYSTEINEMIA ,PEOPLE with diabetes ,CARDIOVASCULAR diseases ,HYPERGLYCEMIA ,ATHEROSCLEROSIS ,LABORATORY mice ,STREPTOZOTOCIN ,MONOCYTES - Abstract
Hyperhomocysteinemia (HHcy) is associated with increased diabetic cardiovascular diseases. However, the role of HHcy in atherogenesis associated with hyperglycemia (HG) remains unknown. To examine the role and mechanisms by which HHcy accelerates HG-induced atherosclerosis, we established an atherosclerosis-susceptible HHcy and HG mouse model. HHcy was established in mice deficient in cystathionine β-synthase (Cbs) in which the homocysteine (Hcy) level could be lowered by inducing transgenic human CBS (Tg-hCBS) using Zn supplementation. HG was induced by streptozotocin injection. Atherosclerosis was induced by crossing Tg-hCBS Cbs mice with apolipoprotein E-deficient (ApoE
-/- ) mice and feeding them a high-fat diet for 2 weeks. We demonstrated that HHcy and HG accelerated atherosclerosis and increased lesion monocytes (MCs) and macrophages (MØs) and further increased inflammatory MC and MØ levels in peripheral tissues. Furthermore, Hcy-lowering reversed circulating mononuclear cells, MC, and inflammatory MC and MC-derived MØ levels. In addition, inflammatory MC correlated positively with plasma Hcy levels and negatively with plasma s-adenosylmethionine-to-s-adenosylhomocysteine ratios. Finally, l-Hcy and d-glucose promoted inflammatory MC differentiation in primary mouse splenocytes, which was reversed by adenoviral DNA methyltransferase-1. HHcy and HG, individually and synergistically, accelerated atherosclerosis and inflammatory MC and MØ differentiation, at least in part, via DNA hypomethylation. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
153. Functional characterization of human methylenetetra- hydrofolate reductase in Saccharomyces cerevisiae.
- Author
-
Shan, Xiaoyin, primary, Wang, Liqun, additional, Hoffmaster, Roselle, additional, and Kruger, Warren D., additional
- Published
- 1999
- Full Text
- View/download PDF
154. Reduction of homocysteine levels in coronary artery disease by low-dose folic acid combined with vitamins B6 and B12
- Author
-
Lobo, Arlene, primary, Naso, Arabi, additional, Arheart, Kristopher, additional, Kruger, Warren D, additional, Abou-Ghazala, Tariq, additional, Alsous, Fadi, additional, Nahlawi, Maher, additional, Gupta, Anjan, additional, Moustapha, Ali, additional, van Lente, Frederick, additional, Jacobsen, Donald W, additional, and Robinson, Killian, additional
- Published
- 1999
- Full Text
- View/download PDF
155. Cystathionine β-synthase mutations in homocystinuria
- Author
-
Kraus, Jan P., primary, Janošík, Miroslav, additional, Kožich, Viktor, additional, Mandell, Roseann, additional, Shih, Vivian, additional, Sperandeo, M.P., additional, Sebastio, Gianfranco, additional, de Franchis, Raffaella, additional, Andria, Generoso, additional, Kluijtmans, Leo A.J., additional, Blom, Henk, additional, Boers, Godfried H.J., additional, Gordon, Ross B., additional, Kamoun, Pierre, additional, Tsai, Michael Y., additional, Kruger, Warren D., additional, Koch, Hans G., additional, Ohura, Toshihiro, additional, and Gaustadnes, Mette, additional
- Published
- 1999
- Full Text
- View/download PDF
156. Cystathionine ?-synthase mutations in homocystinuria
- Author
-
Kraus, Jan P., primary, Jano?�k, Miroslav, additional, Ko?ich, Viktor, additional, Mandell, Roseann, additional, Shih, Vivian, additional, Sperandeo, M.P., additional, Sebastio, Gianfranco, additional, de Franchis, Raffaella, additional, Andria, Generoso, additional, Kluijtmans, Leo A.J., additional, Blom, Henk, additional, Boers, Godfried H.J., additional, Gordon, Ross B., additional, Kamoun, Pierre, additional, Tsai, Michael Y., additional, Kruger, Warren D., additional, Koch, Hans G., additional, Ohura, Toshihiro, additional, and Gaustadnes, Mette, additional
- Published
- 1999
- Full Text
- View/download PDF
157. Evidence for Heme-mediated Redox Regulation of Human Cystathionine β-Synthase Activity
- Author
-
Taoka, Shinichi, primary, Ohja, Sunil, additional, Shan, Xiaoyin, additional, Kruger, Warren D., additional, and Banerjee, Ruma, additional
- Published
- 1998
- Full Text
- View/download PDF
158. Correction of disease-causing CBS mutations in yeast
- Author
-
Shan, Xiaoyin, primary and Kruger, Warren D., additional
- Published
- 1998
- Full Text
- View/download PDF
159. Cystathionine β-synthase-deficient mice thrive on a low-methionine diet.
- Author
-
Gupta, Sapna, Melnyk, Stepan B., and Kruger, Warren D.
- Subjects
CYSTATHIONINE beta-synthase ,HOMOCYSTEINE ,LABORATORY mice ,OSTEOPOROSIS genetics ,GENETIC polymorphism research ,GENE expression ,BONE density - Abstract
Cystathionine β-synthase (CBS) deficiency is a recessive inborn error of metabolism characterized by elevated serum total homocysteine (tHcy). Previously, our laboratory developed a mouse model of CBS deficiency, TgI278T Cbs
-/- (abbreviated as Cbs-/- ), characterized by low weight, low adiposity, decreased Scd-1 expression, facial alopecia, and osteoporosis. To determine the potential benefit of a methionine-restricted diet (MRD), we fed Cbs-/- and Cbs+/- control mice either an MRD or a regular diet (RD) from weaning till 240 d of age. Cbs-/- mice fed the MRD had a 77% decrease in tHcy, 28% increase in weight, 130% increase in fat mass, 82% increase in Scd-1 expression, and 10.6% increase in bone density and entirely lacked the alopecia phenotype observed in age-matched Cbs-/- mice fed the RD. At the end of the study, Cbs-/- mice fed the MRD were phenotypically indistinguishable from Cbs+/- mice fed the RD. Notably, whereas the MRD diet was highly beneficial to Cbs-/- mice, it had nearly opposite effect on Cbs+/- mice. These studies show that a low-methionine diet can correct the phenotypic consequences of loss of CBS and provide a striking example of how genotype and diet can interact to influence phenotype in mammals. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
160. Germline Mutations in Mtap Cooperate with Myc to Accelerate Tumorigenesis in Mice.
- Author
-
Kadariya, Yuwaraj, Tang, Baiqing, Wang, Liqun, Al-Saleem, Tahseen, Hayakawa, Kyoko, Slifker, Michael J., and Kruger, Warren D.
- Subjects
GERM cells ,GENETIC mutation ,NEOPLASTIC cell transformation ,LABORATORY mice ,GENE expression ,METHYLTHIOADENOSINE ,PHOSPHORYLASES ,LYMPHOMAS - Abstract
Objective: The gene encoding the methionine salvage pathway methylthioadenosine phosphorylase (MTAP) is a tumor suppressor gene that is frequently inactivated in a wide variety of human cancers. In this study, we have examined if heterozygosity for a null mutation in Mtap (Mtap
lacZ ) could accelerate tumorigenesis development in two different mouse cancer models, Eμ-myc transgenic and Pten+/− . Methods: Mtap Eμ-myc and Mtap Pten mice were generated and tumor-free survival was monitored over time. Tumors were also examined for a variety of histological and protein markers. In addition, microarray analysis was performed on the livers of MtaplacZ/+ and Mtap+/+ mice. Results: Survival in both models was significantly decreased in MtaplacZ/+ compared to Mtap+/+ mice. In Eµ-myc mice, Mtap mutations accelerated the formation of lymphomas from cells in the early pre-B stage, and these tumors tended to be of higher grade and have higher expression levels of ornithine decarboxylase compared to those observed in control Eµ-myc Mtap+/+ mice. Surprisingly, examination of Mtap status in lymphomas in Eµ-myc MtaplacZ/+ and Eµ-myc Mtap+/+ animals did not reveal significant differences in the frequency of loss of Mtap protein expression, despite having shorter latency times, suggesting that haploinsufficiency of Mtap may be playing a direct role in accelerating tumorigenesis. Consistent with this idea, microarray analysis on liver tissue from age and sex matched Mtap+/+ and MtaplacZ/+ animals found 363 transcripts whose expression changed at least 1.5-fold (P<0.01). Functional categorization of these genes reveals enrichments in several pathways involved in growth control and cancer. Conclusion: Our findings show that germline inactivation of a single Mtap allele alters gene expression and enhances lymphomagenesis in Eµ-myc mice. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
161. Increasing the therapeutic index of 5-fluorouracil and 6-thioguanine by targeting loss of MTAP in tumor cells.
- Author
-
Baiqing Tang, Testa, Joseph R., and Kruger, Warren D.
- Published
- 2012
- Full Text
- View/download PDF
162. The Herp Protein Pathway is Not Involved in the Pro-Amyloidogenic Effect of Hyperhomocysteinemia.
- Author
-
Jia-Min Zhuo, Kruger, Warren D., and Praticò, Domenico
- Subjects
- *
HOMOCYSTEINE , *ALZHEIMER'S disease , *PRESENILINS , *AMYLOID beta-protein , *METABOLISM , *HEAT shock proteins , *MESSENGER RNA - Abstract
Diet-induced high circulating levels of homocysteine, also known as hyper-homocysteinemia (HHcy), is associated with an acceleration of Alzheimer's disease-like amyloidosis. Herp is a homocysteine-responsive stress protein, which has been shown to increase the formation of amyloid-β (Aβ) via interaction with presenilins in vitro. The aim of our paper was to investigate the functional role that Herp plays in HHcy-induced amyloidosis. Amyloidosis secondary to diet-induced HHcy in Tg2576 mice is associated with an increase of Herp protein and mRNA levels. By contrast, no other stress-related proteins are altered by the same diet regimen. Compared to wild type animals, brains from a genetically induced HHcy mouse model did not manifest any significant change in Herp levels. Cells stably over-expressing human AβPP Swedish mutant incubated with high levels of homocysteine had an increase in Aβ formation, but no change in Herp level. Finally, over-expression of Herp did not result in any significant modification of Aβ levels. We conclude that the Herp protein pathway is unlikely to be directly involved in the pro-amyloidotic effect of HHcy. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
163. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthase-deficient mice.
- Author
-
Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, Schafer AI, Durante W, Kruger WD, Yang X, Wang H, Zhang, Daqing, Jiang, Xiaohua, Fang, Pu, Yan, Yan, Song, Jian, Gupta, Sapna, Schafer, Andrew I, Durante, William, and Kruger, Warren D
- Published
- 2009
- Full Text
- View/download PDF
164. Genetic or nutritional disorders in homocysteine or folate metabolism increase protein N-homocysteinylation in mice.
- Author
-
Jakubowski, Hieronim, Perla-Kaján, Joanna, Finnell, Richard H., Cabrera, Robert M., Hong Wang, Gupta, Sapna, Kruger, Warren D., Kraus, Jan P., and Shih, Diana M.
- Subjects
GENETIC disorders ,HOMOCYSTEINE ,VITAMIN B complex ,METHYLENETETRAHYDROFOLATE reductase ,ATHEROSCLEROSIS ,LABORATORY mice - Abstract
Genetic disorders of homocysteine (Hcy) or folate metabolism or high-methionine diets elevate plasma Hcy and its atherogenic metabolite Hcy-thiolactone. In humans, severe hyperhomocysteinemia due to genetic alterations in cystathionine β-synthase (Cbs) or methylenetetrahydrofolate reductase (Mthfr) results in neurological abnormalities and premature death from vascular complications. In mouse models, dietary or genetic hyperhomocysteinemia results in liver or brain pathological changes and accelerates atherosclerosis. Hcy-thiolactone has the ability to form isopeptide bonds with protein lysine residues, which generates modified proteins (N-Hcy-protein) with autoimmunogenic and prothrombotic properties. Our aim was to determine how N-Hcy-protein levels are affected by genetic or nutritional disorders in Hcy or folate metabolism in mice. We found that plasma N-Hcy-protein was elevated 10-fold in mice fed a high-methionine diet compared with the animals fed a normal commercial diet. We also found that inactivation of Cbs, Mthfr, or the proton-coupled folate transporter (Pcft) gene resulted in a 10- to 30-fold increase in plasma or serum N-Hcy-protein levels. Liver N-Hcy-protein was elevated 3.4-fold in severely and 11-fold in extremely hyperhomocysteinemic Cbs-deficient mice, 3.6-fold in severely hyperhomocysteinemic Pcft mice, but was not elevated in mildly hyperhomocysteinemic Mthfr-deficient animals, suggesting that mice have a capacity to prevent accumulation of N-Hcy-protein in their organs. These findings provide evidence that N-Hcy-protein is an important metabolite associated with Hcy pathophysiology in the mouse. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
165. Mouse models of cystathionine β-synthase deficiency reveal significant threshold effects of hyperhomocysteinemia.
- Author
-
Gupta, Sapna, Kühnisch, Jirko, Mustafa, Aladdin, Lhotak, Sarka, Schlachterman, Alexander, Slifker, Michael J., Klein-Szanto, Andres, High, Katherine A., Austin, Richard C., and Kruger, Warren D.
- Subjects
METABOLIC disorders ,HOMOCYSTEINE ,THROMBOSIS ,PHENOTYPES ,ANIMAL disease models ,LABORATORY mice - Abstract
Untreated cystathionine β-synthase (CBS) deficiency in humans is characterized by extremely elevated plasma total homocysteine (tHcy>200 µM), with thrombosis as the major cause of morbidity. Treatment with vitamins and diet leads to a dramatic reduction in thrombotic events, even though patients often still have severe elevations in tHcy (>80 µM). To understand the difference between extreme and severe hyperhomocysteinemia, we have examined two mouse models of CBS deficiency: Tg-hCBS Cbs
-/- mice, with a mean serum tHcy of 169 µM, and Tg-1278T Cbs-/- mice, with a mean tHcy of 296 µM. Only Tg-1278T Cbs-/- animals exhibited strong biological phenotypes, including facial alopecia, osteoporosis, endoplasmic reticulum (ER) stress in the liver and kidney, and a 20% reduction in mean survival rime. Metabolic profiling of serum and liver reveals that Tg-1278T Cbs-/- mice have significantly elevated levels of free oxidized homocysteine but not protein-bound homocysteine in serum and elevation of all forms of homocysteine and S-adenosyl homocysteine in the liver compared to Tg-hCBS Cbs-/- mice. RNA profiling of livers indicate that Tg-1278T Cbs-/- and Tg-hCBS Cbs-/- mice have unique gene signatures, with minimal overlap. Our results indicate that there is a clear pathogenic threshold effect for tHcy and bring into question the idea that mild elevations in tHcy are directly pathogenic. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
166. Mutations in the regulatory domain of cystathionine β–synthase can functionally suppress patient-derived mutations in cis.
- Author
-
Shan, Xiaoyin, Dunbrack Jr., Roland L., Christopher, Scott A., and Kruger, Warren D.
- Abstract
Human cystathionine β–synthase (CBS) is an S-adenosylmethionine-regulated enzyme that plays a key role in the metabolism of homocysteine. Mutations in CBS are known to cause homocystinuria, an inborn error in metabolism. We previously developed a yeast functional assay for CBS and used it to characterize mutations found in homocystinuric patients. We discovered that many patient-derived mutations are functionally suppressed by deletion of the C‐terminal 142 amino acids, which contain a 53 amino acid motif known as the CBS domain. This domain is found in a wide variety of proteins of diverse biological function. Here we have used a genetic screen to identify missense mutations in the C-terminal region of CBS that can suppress the most common patient mutation, I278T. Seven suppressor mutations were identified, four of which map to the CBS domain. When combined in cis with another pathogenic mutation, V168M, six of seven of the suppressor mutations rescued the yeast phenotype. Enzyme activity analyses indicate that the suppressors restore activity from <2% to 17–64% of the wild-type levels. Analysis of the suppressor mutations in the absence of the pathogenic mutation shows that six of the seven suppressor alleles have lost enzymatic responsiveness to S-adenosylmethionine. Using homology modeling, we show that the suppressor mutations appear to map on one face of the CBS domain. Our results indicate that subtle changes to the C-terminus of CBS can restore activity to mutant proteins and provide a rationale for screening for compounds that can activate mutant CBS alleles. [ABSTRACT FROM PUBLISHER]
- Published
- 2001
- Full Text
- View/download PDF
167. Functional Modeling of Vitamin Responsiveness in Yeast: A Common Pyridoxine-Responsive Cystathionine β-Synthase Mutation in Homocystinuria.
- Author
-
Kim, Cecilia E., Gallagher, Paula M., Guttormsen, Anne B., Refsum, Helga, Ueland, Per M., Ose, Leiv, Følling, Ivar, Whitehead, Alexander S., Tsai, Michael Y., and Kruger, Warren D.
- Published
- 1997
- Full Text
- View/download PDF
168. Cloning, Mapping and RNA Analysis of the Human Methionine Synthase Gene.
- Author
-
Li, Yunan N., Gulati, Sumedha, Baker, Priscilla J., Brody, Lawrence C., Banerjee, Ruma, and Kruger, Warren D.
- Published
- 1996
169. A yeast assay for functional detection of mutations in the human cystathionine β-synthase gene.
- Author
-
kruger, Warren D. and Cox, David R.
- Published
- 1995
170. Cystathionine ß-synthase deficiency: Effects of betaine supplementation after methionine restriction in B6-nonresponsive homocystinuria
- Author
-
Singh, Rani H, Kruger, Warren D, Wang, Liqun, Pasquali, Marzia, and Elsas, Louis J
- Abstract
Purpose: For treatment of cystathionine ß-synthase (CßS) deficiency, we determined the effect of betaine (N, N, N-trimethylglycine) therapy and examined the genotype-phenotype relationships to betaine.Methods: In five patients with B6-nonresponsive homocystinuria, we defined the CßS genotypes and determined metabolic responses to betaine as an additive to traditional dietary methionine restriction.Results: After betaine therapy, tHcy declined (mean 47.4 µmol/L; range: -21.2 to -104.0 µmol/L; P = 0.02), whereas total plasma cysteine and methionine did not change. Plasma methionine/tHcy ratios increased by 5.45 (range: +1.5 to 15.3; P = 0.05) inpatients with B6-nonresponsive alleles.Conclusion: Betaine improves metabolic control in B6-nonresponsive patients with homocystinuria after optimum dietary control.
- Published
- 2004
- Full Text
- View/download PDF
171. Hepatitis B viremia is associated with increased risk of hepatocellular carcinoma in chronic carriers
- Author
-
Tang, Baiqing, Kruger, Warren D., Chen, Gang, Shen, Fumin, Lin, Wen Yao, Mboup, Souleymane, London, W. Thomas, and Evans, Alison A.
- Abstract
The role of quantitative viral load in development of hepatocellular carcinoma (HCC) among chronic hepatitis B virus (HBV) carriers was evaluated using real‐time PCR (TaqMan PCR), a highly sensitive method for quantitative detection of HBV DNA. Serum samples collected at study entry from HCC cases and matched controls were chosen separately from ongoing prospective cohort studies in Senegal, West Africa, and Haimen City, China. For 14 HCC cases and 28 controls from Senegal, the relative risk (RR, 95% CI) of HCC was 15.6 (2.0–124.3) for those positive by the TaqMan PCR assay. Average length of follow‐up (study entry to death from HCC) among cases was 2.8 (±1.6) years. The paired median difference between cases and controls was 3.8 × 104virions/ml, with cases higher (P= 0.09). In order to clarify the relationship with lower‐titer viremia, we selected 55 cases and 55 matched controls from the Chinese cohort all negative for serum HBV DNA by conventional dot blot hybridization. In this group, the RR associated with HBV DNA positivity by TaqMan PCR was 3.1 (1.1–9.2), with an average duration of follow‐up of 3.3 (±2.1) years. The median difference in quantitative viremia between cases and controls was 6.0×104virions/ml, with cases higher (P< 0.0001). Increased risk appeared to be confined to subjects with viral loads >2.3 × 104virions/ml. In conclusion, HBV viremia, except perhaps at extremely low levels, is associated with increased risk for HCC in prospective studies of chronic carriers in two disparate populations. J. Med. Virol. 72:35–40, 2004. © 2004 Wiley‐Liss, Inc.
- Published
- 2004
- Full Text
- View/download PDF
172. Involvement of GSK3 in the homocysteine-dependent modulation of the Alzheimer-like phenotype in vivo
- Author
-
Pratico, Domenico, Zhuo, Jia-Min, Portugal, George S., Kruger, Warren D., and Gould, Thomas J.
- Published
- 2009
- Full Text
- View/download PDF
173. Disposition of homocysteine in subjects heterozygous for homocystinuria due to cystathionine β-synthase deficiency: Relationship between genotype and phenotype
- Author
-
Guttormsen, Anne B., Ueland, Per M., Kruger, Warren D., Kim, Cecilia E., Ose, Leiv, Følling, Ivar, and Refsum, Helga
- Abstract
We have investigated 31 subjects from five unrelated families with one or more members with cystathionine β-synthase (CBS) deficiency. On the basis of their CBS genotype, the subjects were grouped as normal (n = 11) or heterozygotes (n = 20). Based on pyridoxine effect in the probands, the heterozygotes were further classified as pyridoxine-responsive (n = 9) or non-responsive (n = 11). Heterozygous subjects had normal fasting total plasma homocysteine (tHcy), but median urinary tHcy excretion rate was significantly elevated compared to healthy controls (0.39 μmol/h vs 0.24 μmol/h, P < 0.05). An abnormal tHcy response after methionine loading identified 73% of the pyridoxine non-responsive heterozygotes, but only 33% of the pyridoxine responsive participants. The increase in cystathionine or the change in tHcy relative to cystathionine did not improve diagnostic accuracy of the methionine loading test. After Hcy loading, the maximal increase in tHcy was significantly elevated, whereas t
1/2 was normal in heterozygotes. In conclusion, a single biochemical test cannot discriminate CBS heterozygotes from controls. Abnormal tHcy response after methionine loading was the most sensitive test. Our data suggest that the urinary tHcy excretion rate is a simple, non-invasive approach for studying mild disturbances in Hcy metabolism. © 2001 Wiley-Liss, Inc.- Published
- 2001
- Full Text
- View/download PDF
174. Polymorphisms in the CBSGene Associated with Decreased Risk of Coronary Artery Disease and Increased Responsiveness to Total Homocysteine Lowering by Folic Acid
- Author
-
Kruger, Warren D., Evans, Alison A., Wang, Liqun, Malinow, M.Rene, Duell, P.Barton, Anderson, Peter H., Block, Peter C., Hess, David L., Graf, Eric E., and Upson, Barbara
- Abstract
Elevated total plasma homocysteine (tHcy) is an established risk factor for the development of vascular disease and neural tube defects. Total homocysteine levels can be lowered by folic acid supplements but individual response is highly variable. In this case–control study, involving 142 coronary artery disease (CAD) patients and 102 controls, we have typed six genetic polymorphisms in three homocysteine metabolizing genes and examined their relationship to the incidence of CAD, tHcy levels, and lowering of tHcy levels in response to folic acid supplementation. We found that two single nucleotide polymorphisms in the cystathionine beta synthase (CBS) gene, 699C → T and 1080T → C, are associated with decreased risk of CAD and increased responsiveness to the tHcy lowering effects of folic acid. Individuals homozygous for 699T were significantly underrepresented in CAD patients as compared to controls (4.9% vs 17.3%, P= 0.0015), as were individuals homozygous for the 1080C (29.6% vs 44.2%, P= 0.018). Additionally, 699T and 1080C homozygous individuals were the most responsive to folate supplementation. 699T homozygotes lowered tHcy levels 13.6% on average, compared to 4.8% lowering in 699C homozygotes (P= 0.009), while 1080C homozygotes lowered 12.9% compared to just 2.7% for 1080T homozygotes (P= 0.005). The two polymorphisms in CBSare third codon changes and would not be predicted to affect the underlying protein. However, there is strong linkage disequilibrium between these two positions, suggesting that they may also be linked to other as yet unidentified polymorphisms within the CBSgene. These observations suggest that specific CBSalleles are a risk factor for the development of vascular disease and that genetic information could be predictive of individual response to folic acid supplementation.
- Published
- 2000
- Full Text
- View/download PDF
175. Author Correction: Highly recurrent CBSepimutations in gastric cancer CpG island methylator phenotypes and inflammation
- Author
-
Padmanabhan, Nisha, Kyon, Huang Kie, Boot, Arnoud, Lim, Kevin, Srivastava, Supriya, Chen, Shuwen, Wu, Zhiyuan, Lee, Hyung-Ok, Mukundan, Vineeth T., Chan, Charlene, Chan, Yarn Kit, Xuewen, Ong, Pitt, Jason J., Isa, Zul Fazreen Adam, Xing, Manjie, Lee, Ming Hui, Tan, Angie Lay Keng, Ting, Shamaine Ho Wei, Luftig, Micah A., Kappei, Dennis, Kruger, Warren D., Bian, Jinsong, Ho, Ying Swan, Teh, Ming, Rozen, Steve George, and Tan, Patrick
- Published
- 2021
- Full Text
- View/download PDF
176. Highly recurrent CBSepimutations in gastric cancer CpG island methylator phenotypes and inflammation
- Author
-
Padmanabhan, Nisha, Kyon, Huang Kie, Boot, Arnoud, Lim, Kevin, Srivastava, Supriya, Chen, Shuwen, Wu, Zhiyuan, Lee, Hyung-Ok, Mukundan, Vineeth T., Chan, Charlene, Chan, Yarn Kit, Xuewen, Ong, Pitt, Jason J., Isa, Zul Fazreen Adam, Xing, Manjie, Lee, Ming Hui, Tan, Angie Lay Keng, Ting, Shamaine Ho Wei, Luftig, Micah A., Kappei, Dennis, Kruger, Warren D., Bian, Jinsong, Ho, Ying Swan, Teh, Ming, Rozen, Steve George, and Tan, Patrick
- Abstract
Background: CIMP (CpG island methylator phenotype) is an epigenetic molecular subtype, observed in multiple malignancies and associated with the epigenetic silencing of tumor suppressors. Currently, for most cancers including gastric cancer (GC), mechanisms underlying CIMP remain poorly understood. We sought to discover molecular contributors to CIMP in GC, by performing global DNA methylation, gene expression, and proteomics profiling across 14 gastric cell lines, followed by similar integrative analysis in 50 GC cell lines and 467 primary GCs. Results: We identify the cystathionine beta-synthase enzyme (CBS) as a highly recurrent target of epigenetic silencing in CIMP GC. Likewise, we show that CBSepimutations are significantly associated with CIMP in various other cancers, occurring even in premalignant gastroesophageal conditions and longitudinally linked to clinical persistence. Of note, CRISPR deletion of CBSin normal gastric epithelial cells induces widespread DNA methylation changes that overlap with primary GC CIMP patterns. Reflecting its metabolic role as a gatekeeper interlinking the methionine and homocysteine cycles, CBSloss in vitro also causes reductions in the anti-inflammatory gasotransmitter hydrogen sulfide (H
2 S), with concomitant increase in NF-κB activity. In a murine genetic model of CBSdeficiency, preliminary data indicate upregulated immune-mediated transcriptional signatures in the stomach. Conclusions: Our results implicate CBSas a bi-faceted modifier of aberrant DNA methylation and inflammation in GC and highlights H2 S donors as a potential new therapy for CBS-silenced lesions.- Published
- 2021
- Full Text
- View/download PDF
177. Real-time detection of enzymatically formed hydrogen sulfide by pathogenic variants of cystathionine beta-synthase using hemoglobin I of Lucina pectinata as a biosensor.
- Author
-
Myszkowska, Joanna, Klotz, Katharina, Leandro, Paula, Kruger, Warren D., Froese, D. Sean, Baumgartner, Matthias R., Spiekerkoetter, Ute, and Hannibal, Luciana
- Subjects
- *
BIOLOGICAL specimens , *CHEMICAL properties , *TREATMENT effectiveness , *HYDROGEN sulfide , *CYSTATHIONINE - Abstract
Classical homocystinuria is a rare disease caused by mutations in cystathionine β-synthase (CBS) gene (OMIM 613381). CBS catalyzes the first step of the transsulfuration pathway that converts homocysteine (Hcy) into cystathionine (Cysta) via a number of co-substrates and mechanisms. Formation of Cysta by condensation of Hcy and cysteine (Cys) produces a molar equivalent of hydrogen sulfide (H 2 S). H 2 S plays important roles in cognitive and vascular functions. Clinically, patients with CBS deficiency present with vascular, ocular, neurological and skeletal impairments. Biochemically, CBS deficiency manifests with elevated Hcy and reduced concentration of Cysta in plasma and urine. A number of pathogenic variants of human CBS have been characterized by their residual enzymatic activity, but very few studies have examined H 2 S production by pathogenic CBS variants, possibly due to technical hurdles in H 2 S detection and quantification. We describe a method for the real-time, continuous quantification of H 2 S formed by wild-type and pathogenic variants of human recombinant CBS, as well as by fibroblast extracts from healthy controls and patients diagnosed with CBS deficiency. The method takes advantage of the specificity and high affinity of hemoglobin I of the clam Lucina pectinata toward H 2 S and is based on UV–visible spectrophotometry. Comparison with the gold-standard, end-point H 2 S quantification method employing monobromobimane, as well as correlations with CBS enzymatic activity determined by LC-MS/MS showed agreement and correlation, and permitted the direct, time-resolved determination of H 2 S production rates by purified human recombinant CBS and by CBS present in fibroblast extracts. Rates of H 2 S production were highest for wild-type CBS, and lower for pathogenic variants. This method enables the examination of structural determinants of CBS that are important for H 2 S production and its possible relevance to the clinical outcome of patients. [Display omitted] • Hydrogen sulfide (H 2 S) is produced by transsulfuration enzymes and the microbiome. • Cystathionine beta-synthase (CBS) is the major intracellular source of H 2 S. • The chemical properties of H 2 S challenge its quantitation in biological specimens. • Hemoglobin I of Lucina pectinata serves as a specific biosensor to detect H 2 S. • H 2 S formed by purified CBS and in human fibroblasts are quantifiable in real time. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
178. Sequences expressed sex-specifically in Drosophila melanogaster adults
- Author
-
DiBenedetto, Angela J., primary, Lakich, Delia M., additional, Kruger, Warren D., additional, Belote, John M., additional, Baker, Bruce S., additional, and Wolfner, Mariana F., additional
- Published
- 1987
- Full Text
- View/download PDF
179. Examination of two different proteasome inhibitors in reactivating mutant human cystathionine β-synthase in mice.
- Author
-
Gupta, Sapna, Lee, Hyung-Ok, Wang, Liqun, and Kruger, Warren D.
- Subjects
- *
PROTEASOME inhibitors , *HEAT shock proteins , *CYSTATHIONINE , *LIVER proteins , *MOLECULAR chaperones , *INBORN errors of metabolism , *PROTEASE inhibitors - Abstract
Classic homocystinuria is an inborn error of metabolism caused mainly by missense mutations leading to misfolded and/or unstable human cystathionine β-synthase (CBS) protein, causing the accumulation of excess total homocysteine (tHcy) in tissues. Previously, it has been shown that certain missense containing human CBS proteins can be functionally rescued in mouse models of CBS deficiency by treatment with proteasome inhibitors. The rescue by proteasome inhibitors is thought to work both by inhibiting the degradation of misfolded CBS protein and by inducing the levels of heat-shock chaperone proteins in the liver. Here we examine the effectiveness of two FDA approved protease inhibitors, carfilzomib and bortezomib, on various transgenic mouse models of human CBS deficiency. Our results show that although both drugs are effective in inducing the liver chaperone proteins Hsp70 and Hsp27, and are effective in inhibiting proteasome function, bortezomib was somewhat more robust in restoring the mutant CBS function. Moreover, there was no significant correlation between proteasome inhibition and CBS activity, suggesting that some of bortezomib's effects are via other mechanisms. We also test the use of low-doses of bortezomib and carfilzomib on various mouse models for extended periods of time and find that while low-doses are less toxic, they are also less effective at restoring CBS function. Overall, these results show that while restoration of mutant CBS function is possible with proteasome inhibitors, the exact mechanism is complicated and it will likely be too toxic for long-term patient treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
180. Extracellular 5'-methylthioadenosine inhibits intracellular symmetric dimethylarginine protein methylation of FUSE-binding proteins.
- Author
-
Baiqing Tang, Hyung-Ok Lee, Gupta, Sapna, Liqun Wang, Kurimchak, Alison M., Duncan, James S., and Kruger, Warren D.
- Subjects
- *
ORNITHINE decarboxylase , *PROTEINS , *METHYLATION , *HEMATOLOGIC malignancies , *METHIONINE , *ARGININE - Abstract
Methylthioadenosine phosphorylase (MTAP) is a key enzyme in the methionine salvage pathway that converts the polyamine synthesis byproduct 5'-deoxy-5'-methylthioadenosine (MTA) into methionine. Inactivation of MTAP, often by homozygous deletion, is found in both solid and hematologic malignancies and is one of the most frequently observed genetic alterations in human cancer. Previous work established that MTAP-deleted cells accumulate MTA and contain decreased amounts of proteins with symmetric dimethylarginine (sDMA). These findings led to the hypothesis that accumulation of intracellular MTA inhibits the protein arginine methylase (PRMT5) responsible for bulk protein sDMAylation. Here, we confirm that MTAP-deleted cells have increased MTA accumulation and reduced protein sDMAylation. However, we also show that addition of extracellular MTA can cause a dramatic reduction of the steady-state levels of sDMA-containing proteins in MTAP+ cells, even though no sustained increase in intracellular MTA is found because of catabolism of MTA by MTAP. We determined that inhibition of protein sDMAylation by MTA occurs within 48 h, is reversible, and is specific. In addition, we have identified two enhancer-binding proteins, FUBP1 and FUBP3, that are differentially sDMAylated in response to MTAP and MTA. These proteins work via the far upstream element site located upstream of Myc and other promoters. Using a transcription reporter construct containing the far upstream element site, we demonstrate that MTA addition can reduce transcription, suggesting that the reduction in FUBP1 and FUBP3 sDMAylation has functional consequences. Overall, our findings show that extracellular MTA can inhibit protein sDMAylation and that this inhibition can affect FUBP function. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
181. Hyperhomocysteinemia impairs endothelium-derived hyperpolarizing factor-mediated vasorelaxation in transgenic cystathionine beta synthase-deficient mice.
- Author
-
Zhongjian Cheng, Xiaohua Jiang, Kruger, Warren D., Praticò, Domenico, Gupta, Sapna, Mallilankaraman, Karthik, Madesh, Muniswamy, Schafer, Andrew I., Durante, William, Xiaofeng Yang, and Hong Wang
- Subjects
- *
HOMOCYSTEINE , *LABORATORY mice , *TRANSGENIC mice , *NITRIC oxide , *ENDOTHELIUM , *RELAXATION for health - Abstract
Hyperhomocysteinemia (HHcy) is associated with endothelial dysfunction (ED), but the mechanism is largely unknown. In this study, we investigated the role and mechanism of HHcy-induced ED in microvasculature in our newly established mouse model of severe HHcy (plasma total homocysteine, 169.5μM). We found that severe HHcy impaired nitric oxide (NO)- and endothelium-derived hyperpolarizing factor (EDHF)-mediated, endothelium-dependent relaxations of small mesenteric arteries (SMAs). Endothelium-independent and prostacyclin-mediated endothelium-dependent relaxations were not changed. A nonselective Ca2+-activated potassium channel (KCa) inhibitor completely blocked EDHF-mediated relaxation. Selective blockers for small-conductance KCa (SK) or intermediate-conductance KCa (IK) failed to inhibit EDHF-mediated relaxation in HHcy mice. HHcy increased the levels of SK3 and IK1 protein, superoxide (O2-), and 3-nitrotyrosine in the endothelium of SMAs. Preincubation with antioxidants and peroxynitrite (ONOO-) inhibitors improved endothelium-dependent and EDHF-mediated relaxations and decreased O2- production in SMAs from HHcy mice. Further, EDHF-mediated relaxation was inhibited by ONOO- and prevented by catalase in the control mice. Finally, L-homocysteine stimulated O2- production, which was reversed by antioxidants, and increased SK/IK protein levels and tyrosine nitration in cultured human cardiac microvascular endothelial cells. Our results suggest that HHcy impairs EDHF relaxation in SMAs by inhibiting SK/IK activities via oxidation- and tyrosine nitration-related mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
182. Inhibition of betaine-homocysteine S-methyltransferase in rats causes hyperhomocysteinemia and reduces liver cystathionine β-synthase activity and methylation capacity.
- Author
-
Strakova, Jana, Gupta, Sapna, Kruger, Warren D., Dilger, Ryan N., Tryon, Katherine, Li, Lucas, and Garrow, Timothy A.
- Published
- 2011
- Full Text
- View/download PDF
183. Analysis of differential neonatal lethality in cystathionine β-synthase deficient mouse models using metabolic profiling.
- Author
-
Gupta, Sapna, Liqun Wang, Slifker, Michael J., Cai, Kathy Q., Maclean, Kenneth N., Wasek, Brandi, Bottiglieri, Teodoro, and Kruger, Warren D.
- Abstract
Cystathionine beta-synthase (CBS) is a key enzyme of the trans-sulfuration pathway that converts homocysteine to cystathionine. Loss of CBS activity due to mutation results in CBS deficiency, an inborn error of metabolism characterized by extreme elevation of plasma total homocysteine (tHcy). C57BL6 mice containing either a homozygous null mutation in the cystathionine β-synthase (Cbs−/−) gene or an inactive human CBS protein (Tg-G307S Cbs−/−) are born in mendelian numbers, but the vast majority die between 18 and 21 days of age due to liver failure. However, adult Cbs null mice that express a hypomorphic allele of human CBS as a transgene (Tg-I278T Cbs−/−) show almost no neonatal lethality despite having serum tHcy levels similar to mice with no CBS activity. Here, we characterize liver and serum metabolites in neonatal Cbs+/−, Tg-G307S Cbs−/−, and Tg-I278T Cbs−/− mice at 6, 10, and 17 days of age to understand this difference. In serum, we observe similar elevations in tHcy in both Tg-G307S Cbs−/− and Tg-I278T Cbs−/− compared to control animals, but methionine is much more severely elevated in Tg-G307S Cbs−/− mice. Large scale metabolomic analysis of liver tissue confirms that both methionine and methioninesulfoxide are significantly more elevated in Tg-G307S Cbs−/− animals, along with significant differences in several other metabolites including hexoses, amino acids, other amines, lipids, and carboxylic acids. Our data are consistent with a model that the neonatal lethality observed in CBS-null mice is driven by excess methionine resulting in increased stress on a variety of related pathways including the urea cycle, TCA cycle, gluconeogenesis, and phosphatidylcholine biosynthesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
184. CRY1‐CBS binding regulates circadian clock function and metabolism.
- Author
-
Cal‐Kayitmazbatir, Sibel, Kulkoyluoglu‐Cotul, Eylem, Growe, Jacqueline, Selby, Christopher P., Rhoades, Seth D., Malik, Dania, Oner, Hasimcan, Asimgil, Hande, Francey, Lauren J., Sancar, Aziz, Kruger, Warren D., Hogenesch, John B., Weljie, Aalim, Anafi, Ron C., and Kavakli, Ibrahim Halil
- Subjects
- *
METABOLIC regulation , *METABOLISM , *MUTANT proteins , *CIRCADIAN rhythms , *ENZYME metabolism , *MELANOPSIN - Abstract
Circadian disruption influences metabolic health. Metabolism modulates circadian function. However, the mechanisms coupling circadian rhythms and metabolism remain poorly understood. Here, we report that cystathionine β‐synthase (CBS), a central enzyme in one‐carbon metabolism, functionally interacts with the core circadian protein cryptochrome 1 (CRY1). In cells, CBS augments CRY1‐mediated repression of the CLOCK/BMAL1 complex and shortens circadian period. Notably, we find that mutant CBS‐I278T protein, the most common cause of homocystinuria, does not bind CRY1 or regulate its repressor activity. Transgenic CbsZn/Zn mice, while maintaining circadian locomotor activity period, exhibit reduced circadian power and increased expression of E‐BOX outputs. CBS function is reciprocally influenced by CRY1 binding. CRY1 modulates enzymatic activity of the CBS. Liver extracts from Cry1−/− mice show reduced CBS activity that normalizes after the addition of exogenous wild‐type (WT) CRY1. Metabolomic analysis of WT, CbsZn/Zn, Cry1−/−, and Cry2−/− samples highlights the metabolic importance of endogenous CRY1. We observed temporal variation in one‐carbon and transsulfuration pathways attributable to CRY1‐induced CBS activation. CBS‐CRY1 binding provides a post‐translational switch to modulate cellular circadian physiology and metabolic control. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
185. Mouse modeling and structural analysis of the p.G307S mutation in human cystathionine β-synthase (CBS) reveal effects on CBS activity but not stability.
- Author
-
Gupta, Sapna, Kelow, Simon, Liqun Wang, Andrake, Mark D., Dunbrack Jr., Roland L., and Kruger, Warren D.
- Subjects
- *
CYSTATHIONINE beta-synthase , *GENETIC mutation , *STRUCTURAL stability , *SULFUR metabolism , *MOLECULAR dynamics - Abstract
Mutations in the cystathionine β-synthase (CBS) gene are the cause of classical homocystinuria, the most common inborn error in sulfur metabolism. The p.G307S mutation is the most frequent cause of CBS deficiency in Ireland, which has the highest prevalence of CBS deficiency in Europe. Individuals homozygous for this mutation tend to be severely affected and are pyridoxine nonresponsive, but the molecular basis for the strong effects of this mutation is unclear. Here, we characterized a transgenic mouse model lacking endogenous Cbs and expressing human p.G307S CBS protein from a zinc-inducible metallothionein promoter (Tg-G307S Cbs-/-). Unlike mice expressing other mutant CBS alleles, the Tg-G307S transgene could not efficiently rescue neonatal lethality of Cbs-/- in a C57BL/6J background. In a C3H/HeJ background, zinc-induced Tg-G307S Cbs-/- mice expressed high levels of p.G307S CBS in the liver, and this protein variant forms multimers, similarly to mice expressingWThuman CBS. However, the p.G307S enzyme had no detectable residual activity. Moreover, treating mice with proteasome inhibitors failed to significantly increase CBS-specific activity. These findings indicated that the G307S substitution likely affects catalytic function as opposed to causing a folding defect. Using molecular dynamics simulation techniques, we found that the G307S substitution likely impairs catalytic function by limiting the ability of the tyrosine at position 308 to assume the proper conformational state(s) required for the formation of the pyridoxal-cystathionine intermediate. These results indicate that the p.G307S CBS is stable but enzymatically inert and therefore unlikely to respond to chaperone-based therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
186. An orally administered enzyme therapeutic for homocystinuria that suppresses homocysteine by metabolizing methionine in the gastrointestinal tract.
- Author
-
Skvorak, Kristen, Mitchell, Vesna, Teadt, Leann, Franklin, Kierra A., Lee, Hyung-Ok, Kruse, Nikki, Huitt-Roehl, Callie, Hang, Julie, Du, Faye, Galanie, Stephanie, Guan, Steven, Aijaz, Hera, Zhang, Nianliu, Rajkovic, Gabriel, Kruger, Warren D., Ismaili, Moulay Hicham Alaoui, Huisman, Gjalt, McCluskie, Kerryn, and Silverman, Adam P.
- Subjects
- *
GASTROINTESTINAL system , *HOMOCYSTEINE , *AMINO acid metabolism disorders , *METHIONINE , *ORAL drug administration , *METHIONINE metabolism - Abstract
Classical homocystinuria (HCU) is a rare inborn error of amino acid metabolism characterized by accumulation of homocysteine, an intermediate product of methionine metabolism, leading to significant systemic toxicities, particularly within the vascular, skeletal, and ocular systems. Most patients require lifelong dietary therapy with severe restriction of natural protein to minimize methionine intake, and many patients still struggle to maintain healthy homocysteine levels. Since eliminating methionine from the diet reduces homocysteine levels, we hypothesized that an enzyme that can degrade methionine within the gastrointestinal (GI) tract could help HCU patients maintain healthy levels while easing natural protein restrictions. We describe the preclinical development of CDX-6512, a methionine gamma lyase (MGL) enzyme that was engineered for stability and activity within the GI tract for oral administration to locally degrade methionine. CDX-6512 is stable to low pH and intestinal proteases, enabling it to survive the harsh GI environment without enteric coating and to degrade methionine freed from dietary protein within the small intestine. Administering CDX-6512 to healthy non-human primates following a high protein meal led to a dose-dependent suppression of plasma methionine. In Tg - I278T Cbs −/− mice, an animal model that recapitulates aspects of HCU disease including highly elevated serum homocysteine levels, oral dosing of CDX-6512 after a high protein meal led to suppression in serum levels of both methionine and homocysteine. When animals received a daily dose of CDX-6512 with a high protein meal for two weeks, the Tg - I278T Cbs −/− mice maintained baseline homocysteine levels, whereas homocysteine levels in untreated animals increased by 39%. These preclinical data demonstrate the potential of CDX-6512 as an oral enzyme therapy for HCU. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
187. Analysis of the Qatari R336C cystathionine β‐synthase protein in mice
- Author
-
Gheyath K. Nasrallah, Tawfeg Ben-Omran, Sapna Gupta, Warren D. Kruger, Liqun Wang, Lorena Gallego-Villar, Johannes Häberle, Beat Thöny, Hyung-Ok Lee, Henk J. Blom, Nader Al-Dewik, Internal medicine, University of Zurich, and Kruger, Warren D
- Subjects
Male ,Homocysteine ,DNA Mutational Analysis ,Bortezomib ,Mice ,chemistry.chemical_compound ,Pyridoxal phosphate ,Genetics (clinical) ,Mice, Knockout ,Mice, Inbred C3H ,0303 health sciences ,biology ,030305 genetics & heredity ,Pyridoxine ,Female ,Homocystinuria ,Proteasome Inhibitors ,inorganic chemicals ,Genetically modified mouse ,2716 Genetics (clinical) ,congenital, hereditary, and neonatal diseases and abnormalities ,mouse model ,Transgene ,Cystathionine beta-Synthase ,610 Medicine & health ,Article ,inborn error ,03 medical and health sciences ,1311 Genetics ,Genetics ,medicine ,Animals ,Alleles ,030304 developmental biology ,methionine ,Cofactor binding ,Methionine ,missense mutation ,organic chemicals ,nutritional and metabolic diseases ,homocysteine ,medicine.disease ,Cystathionine beta synthase ,Molecular biology ,Mice, Inbred C57BL ,chemistry ,10036 Medical Clinic ,Mutation ,biology.protein ,metabolism - Abstract
Classical homocystinuria is a recessive inborn error of metabolism caused by mutations in the cystathionine beta-synthase (CBS) gene. The highest incidence of CBS deficiency in the world is found in the country of Qatar due to the combination of high rates of consanguinity and the presence of a founder mutation, c.1006C>T (p.R336C). This mutation does not respond to pyridoxine and is considered severe. Here we describe the creation of a mouse that is null for the mouse Cbs gene and expresses human p.R336C CBS from a zinc-inducible transgene (Tg-R336C Cbs ). Zinc treated Tg-R336C Cbs mice have extreme elevation in both serum tHcy and liver tHcy compared to control transgenic mice. Both the steady-state protein levels and CBS enzyme activity levels in liver lysates from Tg-R336C Cbs mice are significantly reduced compared to that found in Tg-hCBS Cbs mice expressing wild-type human CBS. Treatment of Tg-R336C Cbs mice with the proteasome inhibitor bortezomib results in stabilization of liver CBS protein and an increase in activity to levels found in corresponding Tg-hCBS Cbs wild type mice. Surprisingly, serum tHcy did not fully correct even though liver enzyme activity was as high as control animals. This discrepancy is explained by in vitro enzymatic studies of mouse liver extracts showing that p.R336C causes reduced binding affinity for the substrate serine by almost seven-fold and significantly increased dependence on pyridoxal phosphate in the reaction buffer. These studies demonstrate that the p.R336C alteration effects both protein stability and substrate/cofactor binding. National Institute of Diabetes and Digestive and Kidney Diseases, Grant/Award Number: DK101404; Qatar Foundation, Grant/Award Number: NPR7‐355‐3‐088; NPRP grant, Grant/Award Number: NPR7‐355‐3‐088; National Institutes of Health, Grant/Award Numbers: P30 CA006927, R01 DK101404
- Published
- 2019
- Full Text
- View/download PDF
188. Paradoxical absence of a prothrombotic phenotype in a mouse model of severe hyperhomocysteinemia.
- Author
-
Dayal, Sanjana, Chauhan, Anil K., Jensen, Melissa, Leo, Lorie, Lynch, Cynthia M., Farad, Frank M., Kruger, Warren D., and Lentz, Steven R.
- Subjects
- *
CARDIOVASCULAR diseases , *PHENOTYPES , *HYPERHOMOCYSTEINEMIA , *HOMOCYSTEINE , *HEALTH outcome assessment , *DISEASE susceptibility , *LABORATORY mice - Abstract
Hyperhomocysteinemia confers a high risk for thrombotic vascular events, but homocysteine-lowering therapies have been ineffective in reducing the incidence of secondary vascular outcomes, raising questions regarding the role of homocysteine as a mediator of cardiovascular disease. Therefore, to determine the contribution of elevated homocysteine to thrombosis susceptibility, we studied Cbs-/- mice conditionally expressing a zinc-inducible mutated human CBS (I278T) transgene. Tg-I278T Cbs-/- mice exhibited severe hyperhomocysteinemia and endothelial dysfunction in cerebral arterioles. Surprisingly, however, these mice did not display increased susceptibility to arterial or venous thrombosis as measured by photochemical injury in the carotid artery, chemical injury in the carotid artery or mesenteric arterioles, or ligation of the inferior vena cava. A survey of hemostatic and hemodynamic parameters revealed no detectible differences between control and Tg-I278T Cbs-/- mice. Our data demonstrate that severe elevation in homocysteine leads to the development of vascular endothelial dysfunction but is not sufficient to promote thrombosis. These findings may provide insights into the failure of homocysteine-lowering trials in secondary prevention from thrombotic vascular events. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
189. Methionine-deficient diet induces post-transcriptional downregulation of cystathionine β-synthase
- Author
-
Tang, Baiqing, Mustafa, Aladdin, Gupta, Sapna, Melnyk, Stepan, James, S. Jill, and Kruger, Warren D.
- Subjects
- *
DISEASE risk factors , *BLOOD plasma , *HOMOCYSTEINE , *METHIONINE , *METABOLIC regulation , *PROTEIN analysis , *ANALYSIS of variance , *ANIMAL experimentation , *BIOLOGICAL models , *COMPUTER software , *ENZYMES , *MICE , *POLYMERASE chain reaction , *RESEARCH funding , *STATISTICS , *T-test (Statistics) , *DATA analysis - Abstract
Objective: Elevated plasma total homocysteine (tHcy) is a risk factor for a variety of human diseases. Homocysteine is formed from methionine and has two primary metabolic fates: remethylation to form methionine or commitment to the transsulfuration pathway by the action of cystathionine β-synthase (CBS). We have examined the metabolic response in mice of a shift from a methionine-replete to a methionine-free diet. Methods and results: We found that shifting 3-mo-old C57BL6 mice to a methionine-free diet caused a transient increase in tHcy and an increase in the tHcy/methionine ratio. Because CBS is a key regulator of tHcy, we examined CBS protein levels and found that within 3 d on the methionine-deficient diet, animals had a 50% reduction in the levels of liver CBS protein and enzyme activity. Examination of CBS mRNA and studies of transgenic animals that express CBS from a heterologous promoter indicated that this reduction is occurring post-transcriptionally. Loss of CBS protein was unrelated to intracellular levels of S-adenosylmethionine, a known regulator of CBS activity and stability. Conclusion: Our results imply that methionine deprivation induces a metabolic state in which methionine is effectively conserved in tissue by shutdown of the transsulfuration pathway by an S-adenosylmethionine–independent mechanism that signals a rapid downregulation of CBS protein. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
190. Dietary intake of S-(α-carboxybutyl)-dl-homocysteine induces hyperhomocysteinemia in rats
- Author
-
Strakova, Jana, Williams, Kelly T., Gupta, Sapna, Schalinske, Kevin L., Kruger, Warren D., Rozen, Rima, Jiracek, Jiri, Li, Lucas, and Garrow, Timothy A.
- Subjects
- *
ADENOSYLMETHIONINE , *SULFUR amino acids , *METHYLTRANSFERASES , *LABORATORY rats , *HOMOCYSTEINE , *METHYLENETETRAHYDROFOLATE reductase , *OLIGOPEPTIDES , *METHIONINE - Abstract
Abstract: Betaine homocysteine S-methyltransferase (BHMT) catalyzes the transfer of a methyl group from betaine to homocysteine (Hcy), forming dimethylglycine and methionine. We previously showed that inhibiting BHMT in mice by intraperitoneal injection of S-(α-carboxybutyl)-dl-homocysteine (CBHcy) results in hyperhomocysteinemia. In the present study, CBHcy was fed to rats to determine whether it could be absorbed and cause hyperhomocysteinemia as observed in the intraperitoneal administration of the compound in mice. We hypothesized that dietary administered CBHcy will be absorbed and will result in the inhibition of BHMT and cause hyperhomocysteinemia. Rats were meal-fed every 8 hours an l-amino acid–defined diet either containing or devoid of CBHcy (5 mg per meal) for 3 days. The treatment decreased liver BHMT activity by 90% and had no effect on methionine synthase, methylenetetrahydrofolate reductase, phosphatidylethanolamine N-methyltransferase, and CTP:phosphocholine cytidylyltransferase activities. In contrast, cystathionine β-synthase activity and immunodetectable protein decreased (56% and 26%, respectively) and glycine N-methyltransferase activity increased (52%) in CBHcy-treated rats. Liver S-adenosylmethionine levels decreased by 25% in CBHcy-treated rats, and S-adenosylhomocysteine levels did not change. Furthermore, plasma choline decreased (22%) and plasma betaine increased (15-fold) in CBHcy-treated rats. The treatment had no effect on global DNA and CpG island methylation, liver histology, and plasma markers of liver damage. We conclude that CBHcy-mediated BHMT inhibition causes an elevation in total plasma Hcy that is not normalized by the folate-dependent conversion of Hcy to methionine. Furthermore, metabolic changes caused by BHMT inhibition affect cystathionine β-synthase and glycine N-methyltransferase activities, which further deteriorate plasma Hcy levels. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
191. Active Cystathionine β-Synthase Can Be Expressed in Heme-free Systems in the Presence of Metal-substituted Porphyrins or a Chemical Chaperone.
- Author
-
Majtan, Tomas, Singh, Laishram R., Liqun Wang, Kruger, Warren D., and Kraus, Jan P.
- Subjects
- *
SACCHAROMYCES cerevisiae , *CYSTATHIONINE gamma-lyase , *HOMOCYSTEINE , *METABOLISM , *PORPHYRINS , *METHIONINE , *ESCHERICHIA coli - Abstract
Cystathionine β-synthase (CBS), a key enzyme in the metabolism of homocysteine, has previously been shown to require a heme co-factor for maximal activity. However, the biochemical function of the CBS heme is not well defined. Here, we show that expression of human CBS in heme-deficient strains of Saccharomyces cerevisiae and Escherichia coli results in production of an enzyme that is misfolded and degraded. Addition of exogenous heme, porphyrins with non-iron metal, or porphyrin lacking metal entirely produced stable and active CBS enzyme. Purification of recombinant CBS enzyme expressed in the presence of various metalloporphyrins confirmed that Mn(III) and Co(III) had 30- 60% of the specific activity of Fe(III)-CBS, and still responded to allosteric activation by S-adenosyl-L-methio- nine. Treatment of S. cerevisiae with the chemical chaperone trimethylamine-N-oxide resulted in near complete restoration of function to human CBS produced in a heme-deficient strain. Taken together, these results suggest that porphyrin moiety of the heme plays a critical role in proper CBS folding and assembly, but that the metal ion is not essential for this function or for allosteric regulation by S-adenosyl-L-methionine. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
192. The Birt-Hogg-Dube and Tuberous Sclerosis Complex Homologs Have Opposing Roles in Amino Acid Homeostasis in Schizosaccharomyces pombe.
- Author
-
van Slegtenhorst, Marjon, Khabibullin, Damir, Hartman, Tiffiney R., Nicolas, Emmanuelle, Kruger, Warren D., and Henske, Elizabeth Petri
- Subjects
- *
TUBEROUS sclerosis , *TUMOR suppressor genes , *AMINO acid metabolism , *HOMEOSTASIS , *RENAL cell carcinoma , *LUNG diseases , *SCHIZOSACCHAROMYCES pombe - Abstract
Birt-Hogg-Dube (BHD) is a tumor suppressor gene disorder characterized by skin hamartomas, cystic lung disease, and renal cell carcinoma. The fact that hamartomas, lung cysts, and renal cell carcinoma can also occur in tuberous sclerosis complex (TSC) suggests that the BHD and TSC proteins may function within a common pathway. To evaluate this hypothesis, we deleted the BHD homolog in Schizosaccharomyces pombe. Expression profiling revealed that six permease and transporter genes, known to be down-regulated in Δtsc1 and Δtsc2, were up-regulated in Δbhd, and levels of specific intracellular amino acids known to be low in Δtsc1 and Δtsc2 were elevated in Δbhd. This "opposite" profile was unexpected, given the overlapping clinical phenotypes. The TSC1/2 proteins inhibit Rheb in mammals, and Tsc1/Tsc2 inhibit Rhb1 in S. pombe. Expression of a hypomorphic allele of rhb1+ dramatically increased permease expression levels in Δbhd but not in wild-type yeast. Loss of Bhd sensitized yeast to rapamycin-induced increases in permease expression levels, and rapamycin induced lethality in Δbhd yeast expressing the hypomorphic Rhb1 allele. In S. pombe, it is known that Rhb1 binds Tor2, and Tor2 inhibition leads to up-regulation of permeases including those that are regulated by Bhd. Our data, therefore, suggest that Bhd activates Tor2. If the mammalian BHD protein, folliculin, similarly activates mammalian target of rapamycin, it will be of great interest to determine how mammalian target of rapamycin inhibition in BHD patients and mammalian target of rapamycin activation in TSC patients lead to overlapping clinical phenotypes. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
193. Chemical chaperone rescue of mutant human cystathionine β-synthase
- Author
-
Singh, Laishram R., Chen, Xulin, Kozˇich, Viktor, and Kruger, Warren D.
- Subjects
- *
MOLECULAR chaperones , *LYASES , *METABOLISM , *SACCHAROMYCES - Abstract
Abstract: Missense mutations in the cystathionine beta-synthase (CBS) gene, such as I278T, are responsible for CBS deficiency, the most common inherited disorder in sulfur metabolism. Expression of human mutant CBS proteins in Saccharomyces cerevisiae reveals that most disease causing mutations severely inhibit enzyme activity and cannot support growth of yeast on cysteine-free media. Here, we show that the osmolyte chemical chaperones glycerol, trimethylamine-N-oxide, dimethylsulfoxide, proline or sorbitol, when added to yeast media, allows growth on cysteine-free media and causes increased enzyme activity from I278T and three other mutant CBS proteins. Rescuable mutants are ones that are predicted to cause a decrease in solvent accessible surface area. The increase in enzyme activity is associated with stabilization of the tetramer form of the enzyme. This effect is not specific to yeast, as addition of the chaperone glycerol resulted in increased I278T activity when the enzyme is produced either in Escherichia coli or in a coupled in vitro transcription/translation reaction. However, no stimulation of specific activity was observed when chaperones were added directly to purified I278T indicating that the presence of chemical chaperones is required during translation. We also found that by mixing different chaperones we could achieve rescue at significantly lower chaperone concentrations. Taken together, our data show that chemical chaperones present during the initial folding process can facilitate proper folding of several mutant CBS proteins and suggest it may be possible to treat some inborn errors of metabolism with agents that enhance proper protein folding. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
194. The methionine salvage pathway compound 4-methylthio-2-oxobutanate causes apoptosis independent of down-regulation of ornithine decarboxylase
- Author
-
Tang, Baiqing, Kadariya, Yuwaraj, Murphy, Maureen E., and Kruger, Warren D.
- Subjects
- *
APOPTOSIS , *CELL death , *PHOSPHORYLATION , *ALIPHATIC compounds - Abstract
Abstract: 4-Methylthio-2-oxobutanoic acid (MTOB) is the final compound of the methionine salvage pathway that converts the polyamine byproduct methylthioadenosine to adenine and methionine. Here we find that MTOB inhibits growth of several human cell lines in a dose-dependent manner. Growth inhibition was specific for MTOB as we did not observe any inhibition with other chemically related compounds. MTOB treatment causes apoptosis and reduction of ornithine decarboxylase (ODC) activity but not ODC mRNA. To determine if MTOB exerts its effects primarily via ODC inhibition, we compared the effects of MTOB with the ODC-specific inhibitor difluoromethylornithine (DFMO). We found that MTOB was a more potent inducer of apoptosis than DFMO, lacked activation of caspase 3/7, and was able to induce apoptosis in cells lacking p53. Our results show that MTOB-induced growth inhibition and apoptosis is not simply secondary due to ODC inhibition and implies that MTOB activates apoptosis via other mechanisms. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
195. Growth inhibition and induction of apoptosis in mesothelioma cells by selenium and dependence on selenoprotein SEP15 genotype.
- Author
-
Apostolou, Sinoula, Klein, Julian O., Mitsuuchi, Yasuhiro, Shetler, Justin N., Poulikakos, Poulikos I., Jhanwar, Suresh C., Kruger, Warren D., and Testa, Joseph R.
- Subjects
- *
SELENIUM , *APOPTOSIS , *CELL death , *LUNG tumors , *PREVENTIVE medicine , *RESPIRATORY organs , *CELL culture , *CELL lines ,CHEST tumors - Abstract
Malignant mesotheliomas (MMs) are aggressive tumors derived from mesothelial cells lining the lungs, pericardium and peritoneum, and are often associated with occupational asbestos exposure. Suppression subtractive hybridization was used to identify genes differentially expressed in MM cells compared to normal mesothelial cells. A gene, SEP15, encoding a 15-kDa selenium-containing protein was isolated using this approach and was subsequently shown to be downregulated in ~60% of MM cell lines and tumor specimens. A SEP15 polymorphic variant, 1125A, resides in the SECIS recognition element in the 3'-UTR and may influence the efficiency of Sec incorporation into the protein during translation. Since previous studies have implicated a potential role of the trace element selenium as a chemopreventive agent in animal models and in several types of human cancer, we investigated the effect of selenium on MM cells and its dependence on SEP15 genotype. Selenium was shown to inhibit cell growth and induce apoptosis in a dose-dependent manner in MM cells but had minimal effect on normal mesothelial cells. However, MM cells with downregulated SEP15 or the 1125A variant were somewhat less responsive to the growth inhibitory and apoptotic effects of selenium than MM cells expressing wild-type protein. RNAi-based knockdown studies demonstrated that SEP15 inhibition makes sensitive MM cells more resistant to selenium. These data imply that selenium may be useful as a chemopreventive agent in individuals at high risk of MM due to asbestos exposure, although those with the 1125A polymorphism may be less responsive to the protective benefits of dietary selenium supplementation.Oncogene (2004) 23, 5032-5040. doi:10.1038/sj.onc.1207683 Published online 26 April 2004 [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
196. CRY1-CBS binding regulates circadian clock function and metabolism
- Author
-
Aalim M. Weljie, Lauren J. Francey, Sibel Cal-Kayitmazbatir, Seth D. Rhoades, Aziz Sancar, Christopher P. Selby, Eylem Kulkoyluoglu-Cotul, John B. Hogenesch, Warren D. Kruger, Ibrahim Halil Kavakli, Jacqueline Growe, Hasimcan Oner, Ron C. Anafi, Hande Asimgil, Dania Malik, Kayıtmazbatır, Sibel Çal, Çotul, Eylem Külköyluoğlu, Öner, Haşimcan, Asımgil, Hande, Kavaklı, İbrahim Halil (ORCID 0000-0001-6624-3505 & YÖK ID 40319), Growe, Jacqueline, Selby, Christopher P., Rhoades, Seth D., Malik, Dania, Francey, Lauren J., Sancar, Aziz, Kruger, Warren D., Hogenesch, John B., Weljie, Aalim, Anafi, Ron C., Graduate School of Sciences and Engineering, College of Engineering, Department of Molecular Biology and Genetics, Department of Chemical and Biological Engineering, and Department of Materials Science and Engineering
- Subjects
0301 basic medicine ,Male ,Circadian clock ,CLOCK Proteins ,Transsulfuration ,Endogeny ,Biochemistry ,E-Box Elements ,Mice ,0302 clinical medicine ,Cryptochrome ,Circadian rhythm ,Cystathionine beta-synthase ,Metabolism ,Transcriptional regulation ,Mice, Knockout ,0303 health sciences ,biology ,Chemistry ,ARNTL Transcription Factors ,Period Circadian Proteins ,Circadian Rhythm ,Cell biology ,030220 oncology & carcinogenesis ,Metabolome ,Female ,Metabolic Networks and Pathways ,Cryptochrome-1 ,Protein Binding ,Signal Transduction ,inorganic chemicals ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system ,animal structures ,Period (gene) ,Cystathionine beta-Synthase ,Repressor ,Article ,03 medical and health sciences ,Circadian Clocks ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,030304 developmental biology ,Sequence Homology, Amino Acid ,fungi ,Wild type ,nutritional and metabolic diseases ,Cell Biology ,Cystathionine beta synthase ,Cryptochromes ,030104 developmental biology ,HEK293 Cells ,Mutation ,biology.protein ,sense organs ,Protein Processing, Post-Translational ,Sequence Alignment ,030217 neurology & neurosurgery - Abstract
Circadian disruption influences metabolic health. Metabolism modulates circadian function. However, the mechanisms coupling circadian rhythms and metabolism remain poorly understood. Here, we report that cystathionine beta-synthase (CBS), a central enzyme in one-carbon metabolism, functionally interacts with the core circadian protein cryptochrome 1 (CRY1). In cells, CBS augments CRY1-mediated repression of the CLOCK/BMAL1 complex and shortens circadian period. Notably, we find that mutant CBS-I278T protein, the most common cause of homocystinuria, does not bind CRY1 or regulate its repressor activity. Transgenic Cbs(Zn/Zn) mice, while maintaining circadian locomotor activity period, exhibit reduced circadian power and increased expression of E-BOX outputs. CBS function is reciprocally influenced by CRY1 binding. CRY1 modulates enzymatic activity of the CBS. Liver extracts from Cry1(-/-) mice show reduced CBS activity that normalizes after the addition of exogenous wild-type (WT) CRY1. Metabolomic analysis of WT, Cbs(Zn/Zn), Cry1(-/-), and Cry2(-/-) samples highlights the metabolic importance of endogenous CRY1. We observed temporal variation in one-carbon and transsulfuration pathways attributable to CRY1-induced CBS activation. CBS-CRY1 binding provides a post-translational switch to modulate cellular circadian physiology and metabolic control., İstanbul Development Agency; Defense Advanced Research Projects Agency; National Institute of Neurological Disorders and Stroke; Scientific and Technological Research Council of Turkey (TÜBİTAK) BIDEB 2214-A Scholarship
- Published
- 2020
- Full Text
- View/download PDF
197. Impact of primary sequence changes on the self-association properties of mammalian cystathionine beta-synthase enzymes.
- Author
-
Lee HO, Gupta K, Wang L, Dunbrack RL, Majtan T, and Kruger WD
- Subjects
- Humans, Animals, Mice, Homocystinuria genetics, Homocystinuria enzymology, Homocystinuria metabolism, Protein Multimerization, Mutation, Protein Structure, Quaternary, Amino Acid Sequence, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Recombinant Proteins genetics, Cystathionine beta-Synthase chemistry, Cystathionine beta-Synthase genetics, Cystathionine beta-Synthase metabolism
- Abstract
Cystathionine beta-synthase (CBS) is an evolutionarily conserved enzyme that plays a key role in mammalian sulfur amino acid biochemistry, mutations in which are the cause of classical homocystinuria (HCU), an inborn error of metabolism. Although there is agreement in the literature that CBS is a homomultimer, its precise structure is a source of confusion. Here, we performed a series of experiments examining the quaternary structure of various wild-type and mutant CBS enzymes using a combination of native gel electrophoresis, in situ activity assays, analytical ultracentrifugation, and gel filtration. Our data show that recombinantly expressed and purified full-length wild-type human CBS enzyme (hCBS) and HCU-causing variants (p.P422L, p.I435T, and p.R125Q CBS) form high molecular weight assemblies that are consistent with the properties expected of a filament. The filament is enzymatically active, and its size is sensitive to protein concentration. This behavior contrasts sharply with hCBS enzymes containing small deletions within the Bateman domain, which form stable tetramers and octamers regardless of concentration. Examination of liver lysates from humans and mice confirms the existence of enzymatically active high molecular weight aggregates in vivo, but also shows that these aggregates are specific to human CBS and do not occur in mice. Molecular modeling using AlphaFold2 suggests that these experimentally observed differences may be explained by subtle differences in the interaction mediated by the Bateman domains. Our results show that small differences in amino acid sequence can cause large differences in the size and shape of CBS multimers., (© 2024 The Protein Society.)
- Published
- 2024
- Full Text
- View/download PDF
198. Deciphering pathophysiological mechanisms underlying cystathionine beta-synthase-deficient homocystinuria using targeted metabolomics, liver proteomics, sphingolipidomics and analysis of mitochondrial function.
- Author
-
Majtan T, Olsen T, Sokolova J, Krijt J, Křížková M, Ida T, Ditrói T, Hansikova H, Vit O, Petrak J, Kuchař L, Kruger WD, Nagy P, Akaike T, and Kožich V
- Subjects
- Animals, Mice, Mitochondria metabolism, Lipidomics methods, Proteome metabolism, Homocystinuria metabolism, Homocystinuria genetics, Proteomics methods, Cystathionine beta-Synthase metabolism, Cystathionine beta-Synthase deficiency, Cystathionine beta-Synthase genetics, Liver metabolism, Disease Models, Animal, Metabolomics methods, Mice, Transgenic, Sphingolipids metabolism
- Abstract
Background: Cystathionine β-synthase (CBS)-deficient homocystinuria (HCU) is an inherited disorder of sulfur amino acid metabolism with varying severity and organ complications, and a limited knowledge about underlying pathophysiological processes. Here we aimed at getting an in-depth insight into disease mechanisms using a transgenic mouse model of HCU (I278T)., Methods: We assessed metabolic, proteomic and sphingolipidomic changes, and mitochondrial function in tissues and body fluids of I278T mice and WT controls. Furthermore, we evaluated the efficacy of methionine-restricted diet (MRD) in I278T mice., Results: In WT mice, we observed a distinct tissue/body fluid compartmentalization of metabolites with up to six-orders of magnitude differences in concentrations among various organs. The I278T mice exhibited the anticipated metabolic imbalance with signs of an increased production of hydrogen sulfide and disturbed persulfidation of free aminothiols. HCU resulted in a significant dysregulation of liver proteome affecting biological oxidations, conjugation of compounds, and metabolism of amino acids, vitamins, cofactors and lipids. Liver sphingolipidomics indicated upregulation of the pro-proliferative sphingosine-1-phosphate signaling pathway. Liver mitochondrial function of HCU mice did not seem to be impaired compared to controls. MRD in I278T mice improved metabolic balance in all tissues and substantially reduced dysregulation of liver proteome., Conclusion: The study highlights distinct tissue compartmentalization of sulfur-related metabolites in normal mice, extensive metabolome, proteome and sphingolipidome disruptions in I278T mice, and the efficacy of MRD to alleviate some of the HCU-related biochemical abnormalities., Competing Interests: Declaration of competing interest The authors declare no conflict of interests., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
199. Recent therapeutic approaches to cystathionine beta-synthase-deficient homocystinuria.
- Author
-
Majtan T, Kožich V, and Kruger WD
- Subjects
- Humans, Cystathionine beta-Synthase genetics, Cystathionine beta-Synthase metabolism, Quality of Life, Mutation, Missense, Homocystinuria drug therapy, Homocystinuria genetics, Homocystinuria metabolism, Thromboembolism
- Abstract
Cystathionine beta-synthase (CBS)-deficient homocystinuria (HCU) is the most common inborn error of sulfur amino acid metabolism. The pyridoxine non-responsive form of the disease manifests itself by massively increasing plasma and tissue concentrations of homocysteine, a toxic intermediate of methionine metabolism that is thought to be the major cause of clinical complications including skeletal deformities, connective tissue defects, thromboembolism and cognitive impairment. The current standard of care involves significant dietary interventions that, despite being effective, often adversely affect quality of life of HCU patients, leading to poor adherence to therapy and inadequate biochemical control with clinical complications. In recent years, the unmet need for better therapeutic options has resulted in development of novel enzyme and gene therapies and exploration of pharmacological approaches to rescue CBS folding defects caused by missense pathogenic mutations. Here, we review scientific evidence and current state of affairs in development of recent approaches to treat HCU., (© 2022 The British Pharmacological Society.)
- Published
- 2023
- Full Text
- View/download PDF
200. How to fix a broken protein: restoring function to mutant human cystathionine β-synthase.
- Author
-
Kruger WD
- Subjects
- Humans, Molecular Chaperones genetics, Mutation, Phenotype, Cystathionine beta-Synthase genetics, Cystathionine beta-Synthase metabolism, Homocystinuria drug therapy, Homocystinuria therapy
- Abstract
Inborn errors of metabolism (IEM) comprise a large class of recessive genetic diseases involving disorders of cellular metabolism that tend to be caused by missense mutations in which a single incorrect amino acid is substituted in the polypeptide chain. Cystathionine beta-synthase (CBS) deficiency is an example of an IEM that causes large elevations of blood total homocysteine levels, resulting in phenotypes in several tissues. Current treatment strategies involve dietary restriction and vitamin therapy, but these are only partially effective and do not work in all patients. Over 85% of the described mutations in CBS-deficient patients are missense mutations in which the mutant protein fails to fold into an active conformation. The ability of CBS to achieve an active conformation is affected by a variety of intracellular protein networks including the chaperone system and the ubiquitin/proteasome system, collectively referred to as the proteostasis network. Proteostasis modulators are drugs that perturb various aspects of these networks. In this article, we will review the evidence that modulation of the intracellular protein folding environment can be used as a potential therapeutic strategy to treat CBS deficiency and discuss the pros and cons of such a strategy., (© 2021. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.