Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Birth Defects Res ; 115(10): 959-966, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37218073

RESUMEN

The Society for Birth Defects Research and Prevention (BDRP) strives to understand and protect against potential hazards to developing embryos, fetuses, children, and adults by bringing together scientific knowledge from diverse fields. The theme of 62nd Annual Meeting of BDRP, "From Bench to Bedside and Back Again", represented the cutting-edge research areas of high relevance to public health and significance in the fields of birth defects research and surveillance. The multidisciplinary Research Needs Workshop (RNW) convened at the Annual Meeting continues to identify pressing knowledge gaps and encourage interdisciplinary research initiatives. The multidisciplinary RNW was first introduced at the 2018 annual meeting to provide an opportunity for annual meeting attendees to participate in breakout discussions on emerging topics in birth defects research and to foster collaboration between basic researchers, clinicians, epidemiologists, drug developers, industry partners, funding agencies, and regulators to discuss state-of-the-art methods and innovative projects. Initially, a list of workshop topics was compiled by the RNW planning committee and circulated among the members of BDRP to obtain the most popular topics for the Workshop discussions. Based on the pre-meeting survey results, the top three discussion topics selected were, A) Inclusion of pregnant and lactating women in clinical trials. When, why, and how? B) Building multidisciplinary teams across disciplines: What cross-training is needed? And C) Challenges in applications of Artificial Intelligence (AI) and machine learning for risk factor analysis in birth defects research. This report summarizes the key highlights of the RNW workshop and specific topic discussions.


Asunto(s)
Inteligencia Artificial , Investigación Interdisciplinaria , Embarazo , Niño , Femenino , Humanos , Lactancia , Estudios Interdisciplinarios , Sociedades
2.
Front Cardiovasc Med ; 9: 742850, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36386365

RESUMEN

Calcific aortic valve disease (CAVD) is an increasingly prevalent condition among the elderly population that is associated with significant morbidity and mortality. Insufficient understanding of the underlying disease mechanisms has hindered the development of pharmacologic therapies for CAVD. Recently, we described nitric oxide (NO) mediated S-nitrosylation as a novel mechanism for preventing the calcific process. We demonstrated that NO donor or an S-nitrosylating agent, S-nitrosoglutathione (GSNO), inhibits spontaneous calcification in porcine aortic valve interstitial cells (pAVICs) and this was supported by single-cell RNA sequencing (scRNAseq) that demonstrated NO donor and GSNO inhibited myofibroblast activation of pAVICs. Here, we investigated novel signaling pathways that are critical for the calcification of pAVICs that are altered by NO and GSNO by performing an in-depth analysis of the scRNA-seq dataset. Transcriptomic analysis revealed 1,247 differentially expressed genes in pAVICs after NO donor or GSNO treatment compared to untreated cells. Pathway-based analysis of the differentially expressed genes revealed an overrepresentation of the integrin signaling pathway, along with the Rho GTPase, Wnt, TGF-ß, and p53 signaling pathways. We demonstrate that ITGA8 and VCL, two of the identified genes from the integrin signaling pathway, which are known to regulate cell-extracellular matrix (ECM) communication and focal adhesion, were upregulated in both in vitro and in vivo calcific conditions. Reduced expression of these genes after treatment with NO donor suggests that NO inhibits calcification by targeting myofibroblast adhesion and ECM remodeling. In addition, withdrawal of NO donor after 3 days of exposure revealed that NO-mediated transcriptional and translational regulation is a transient event and requires continuous NO exposure to inhibit calcification. Overall, our data suggest that NO and S-nitrosylation regulate the integrin signaling pathway to maintain healthy cell-ECM interaction and prevent CAVD.

3.
Commun Biol ; 5(1): 820, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35970860

RESUMEN

Congenital heart disease (CHD) is the most prevalent birth defect, often linked to genetic variations, environmental exposures, or combination of both. Epidemiological studies reveal that maternal pregestational diabetes is associated with ~5-fold higher risk of CHD in the offspring; however, the causal mechanisms affecting cardiac gene-regulatory-network (GRN) during early embryonic development remain poorly understood. In this study, we utilize an established murine model of pregestational diabetes to uncover the transcriptional responses in key cell-types of the developing heart exposed to maternal hyperglycemia (matHG). Here we show that matHG elicits diverse cellular responses in E9.5 and E11.5 embryonic hearts compared to non-diabetic hearts by single-cell RNA-sequencing. Through differential-gene-expression and cellular trajectory analyses, we identify perturbations in genes, predominantly affecting Isl1+ second heart field progenitors and Tnnt2+ cardiomyocytes with matHG. Using cell-fate mapping analysis in Isl1-lineage descendants, we demonstrate that matHG impairs cardiomyocyte differentiation and alters the expression of lineage-specifying cardiac genes. Finally, our work reveals matHG-mediated transcriptional changes in second heart field lineage that elevate CHD risk by perturbing Isl1-GRN during cardiomyocyte differentiation. Gene-environment interaction studies targeting the Isl1-GRN in cardiac progenitor cells will have a broader impact on understanding the mechanisms of matHG-induced risk of CHD associated with diabetic pregnancies.


Asunto(s)
Diabetes Gestacional , Cardiopatías Congénitas , Hiperglucemia , Animales , Modelos Animales de Enfermedad , Femenino , Cardiopatías Congénitas/genética , Humanos , Hiperglucemia/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Embarazo , Células Madre , Transcriptoma
4.
Birth Defects Res ; 114(16): 926-947, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35261209

RESUMEN

Congenital heart disease (CHD) represents a major class of birth defects worldwide and is associated with cardiac malformations that often require surgical intervention immediately after birth. Despite the intense efforts from multicentric genome/exome sequencing studies that have identified several genetic variants, the etiology of CHD remains diverse and often unknown. Genetically modified animal models with candidate gene deficiencies continue to provide novel molecular insights that are responsible for fetal cardiac development. However, the past decade has seen remarkable advances in the field of human induced pluripotent stem cell (hiPSC)-based disease modeling approaches to better understand the development of CHD and discover novel preventative therapies. The iPSCs are derived from reprogramming of differentiated somatic cells to an embryonic-like pluripotent state via overexpression of key transcription factors. In this review, we describe how differentiation of hiPSCs to specialized cardiac cellular identities facilitates our understanding of the development and pathogenesis of CHD subtypes. We summarize the molecular and functional characterization of hiPSC-derived differentiated cells in support of normal cardiogenesis, those that go awry in CHD and other heart diseases. We illustrate how stem cell-based disease modeling enables scientists to dissect the molecular mechanisms of cell-cell interactions underlying CHD. We highlight the current state of hiPSC-based studies that are in the verge of translating into clinical trials. We also address limitations including hiPSC-model reproducibility and scalability and differentiation methods leading to cellular heterogeneity. Last, we provide future perspective on exploiting the potential of hiPSC technology as a predictive model for patient-specific CHD, screening pharmaceuticals, and provide a source for cell-based personalized medicine. In combination with existing clinical and animal model studies, data obtained from hiPSCs will yield further understanding of oligogenic, gene-environment interaction, pathophysiology, and management for CHD and other genetic cardiac disorders.


Asunto(s)
Cardiopatías Congénitas , Células Madre Pluripotentes Inducidas , Animales , Cardiopatías Congénitas/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Organogénesis , Preparaciones Farmacéuticas , Reproducibilidad de los Resultados , Factores de Transcripción
5.
Genesis ; 59(11): e23449, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34498806

RESUMEN

Congenital heart disease (CHD) is the leading cause of birth defect-related death in infants and is a global pediatric health concern. While the genetic causes of CHD have become increasingly recognized with advances in genome sequencing technologies, the etiology for the majority of cases of CHD is unknown. The maternal environment during embryogenesis has a profound impact on cardiac development, and numerous environmental factors are associated with an elevated risk of CHD. Maternal diabetes mellitus (matDM) is associated with up to a fivefold increased risk of having an infant with CHD. The rising prevalence of diabetes mellitus has led to a growing interest in the use of experimental diabetic models to elucidate mechanisms underlying this associated risk for CHD. The purpose of this review is to provide a comprehensive summary of rodent models that are being used to investigate alterations in cardiac developmental pathways when exposed to a maternal diabetic setting and to summarize the key findings from these models. The majority of studies in the field have utilized the chemically induced model of matDM, but recent advances have also been made using diet based and genetic models. Each model provides an opportunity to investigate unique aspects of matDM and is invaluable for a comprehensive understanding of the molecular and cellular mechanisms underlying matDM-associated CHD.


Asunto(s)
Diabetes Gestacional/metabolismo , Cardiopatías Congénitas/etiología , Corazón/embriología , Hiperglucemia/metabolismo , Embarazo en Diabéticas/metabolismo , Animales , Diabetes Gestacional/genética , Femenino , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/genética , Embarazo , Embarazo en Diabéticas/genética
6.
Sci Adv ; 7(6)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33547080

RESUMEN

Calcific aortic valve disease (CAVD) is an increasingly prevalent condition, and endothelial dysfunction is implicated in its etiology. We previously identified nitric oxide (NO) as a calcification inhibitor by its activation of NOTCH1, which is genetically linked to human CAVD. Here, we show NO rescues calcification by an S-nitrosylation-mediated mechanism in porcine aortic valve interstitial cells and single-cell RNA-seq demonstrated NO regulates the NOTCH pathway. An unbiased proteomic approach to identify S-nitrosylated proteins in valve cells found enrichment of the ubiquitin-proteasome pathway and implicated S-nitrosylation of USP9X (ubiquitin specific peptidase 9, X-linked) in NOTCH regulation during calcification. Furthermore, S-nitrosylated USP9X was shown to deubiquitinate and stabilize MIB1 for NOTCH1 activation. Consistent with this, genetic deletion of Usp9x in mice demonstrated CAVD and human calcified aortic valves displayed reduced S-nitrosylation of USP9X. These results demonstrate a previously unidentified mechanism by which S-nitrosylation-dependent regulation of a ubiquitin-associated pathway prevents CAVD.

8.
Birth Defects Res ; 110(20): 1504-1516, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576094

RESUMEN

Congenital heart disease (CHD) is the most common type of birth defect and is both a significant pediatric and adult health problem, in light of a growing population of survivors. The etiology of CHD has been considered to be multifactorial with genetic and environmental factors playing important roles. The combination of advances in cardiac developmental biology, which have resulted in the elucidation of molecular pathways regulating normal cardiac morphogenesis, and genome sequencing technology have allowed the discovery of numerous genetic contributors of CHD ranging from chromosomal abnormalities to single gene variants. Conversely, mechanistic details of the contribution of environmental factors to CHD remain unknown. Maternal diabetes mellitus (matDM) is a well-established and increasingly prevalent environmental risk factor for CHD, but the underlying etiologic mechanisms by which pregestational matDM increases the vulnerability of embryos to cardiac malformations remains largely elusive. Here, we will briefly discuss the multifactorial etiology of CHD with a focus on the epidemiologic link between matDM and CHD. We will describe the animal models used to study the underlying mechanisms between matDM and CHD and review the numerous cellular and molecular pathways affected by maternal hyperglycemia in the developing heart. Last, we discuss how this increased understanding may open the door for the development of novel prevention strategies to reduce the incidence of CHD in this high-risk population.


Asunto(s)
Corazón Fetal/efectos de los fármacos , Cardiopatías Congénitas/etiología , Hiperglucemia/complicaciones , Animales , Cardiomegalia/etiología , Diabetes Mellitus/fisiopatología , Diabetes Gestacional/fisiopatología , Modelos Animales de Enfermedad , Femenino , Corazón Fetal/embriología , Cardiopatías Congénitas/genética , Humanos , Incidencia , Ratones , Madres , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Prevalencia , Factores de Riesgo
9.
JCI Insight ; 2(20)2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29046480

RESUMEN

Birth defects are the leading cause of infant mortality, and they are caused by a combination of genetic and environmental factors. Environmental risk factors may contribute to birth defects in genetically susceptible infants by altering critical molecular pathways during embryogenesis, but experimental evidence for gene-environment interactions is limited. Fetal hyperglycemia associated with maternal diabetes results in a 5-fold increased risk of congenital heart disease (CHD), but the molecular basis for this correlation is unknown. Here, we show that the effects of maternal hyperglycemia on cardiac development are sensitized by haploinsufficiency of Notch1, a key transcriptional regulator known to cause CHD. Using ATAC-seq, we found that hyperglycemia decreased chromatin accessibility at the endothelial NO synthase (Nos3) locus, resulting in reduced NO synthesis. Transcription of Jarid2, a regulator of histone methyltransferase complexes, was increased in response to reduced NO, and this upregulation directly resulted in inhibition of Notch1 expression to levels below a threshold necessary for normal heart development. We extended these findings using a Drosophila maternal diabetic model that revealed the evolutionary conservation of this interaction and the Jarid2-mediated mechanism. These findings identify a gene-environment interaction between maternal hyperglycemia and Notch signaling and support a model in which environmental factors cause birth defects in genetically susceptible infants.


Asunto(s)
Complicaciones de la Diabetes , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Interacción Gen-Ambiente , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Embarazo en Diabéticas/metabolismo , Animales , Enfermedades Cardiovasculares/etiología , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/metabolismo , Femenino , Feto , Predisposición Genética a la Enfermedad , Corazón/embriología , Corazón/crecimiento & desarrollo , Histona Metiltransferasas/metabolismo , Humanos , Hiperglucemia/complicaciones , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III , Embarazo , Receptor Notch1/metabolismo , Factores de Transcripción/metabolismo
10.
Circ Cardiovasc Genet ; 9(4): 320-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27418595

RESUMEN

BACKGROUND: Congenital heart disease (CHD) is the most common type of birth defect with family- and population-based studies supporting a strong genetic cause for CHD. The goal of this study was to determine whether a whole exome sequencing (WES) approach could identify pathogenic-segregating variants in multiplex CHD families. METHODS AND RESULTS: WES was performed on 9 kindreds with familial CHD, 4 with atrial septal defects, 2 with patent ductus arteriosus, 2 with tetralogy of Fallot, and 1 with pulmonary valve dysplasia. Rare variants (<1% minor allele frequency) that segregated with disease were identified by WES, and variants in 69 CHD candidate genes were further analyzed. These selected variants were subjected to in silico analysis to predict pathogenicity and resulted in the discovery of likely pathogenic mutations in 3 of 9 (33%) families. A GATA4 mutation in the transactivation domain, p.G115W, was identified in familial atrial septal defects and demonstrated decreased transactivation ability in vitro. A p.I263V mutation in TLL1 was identified in an atrial septal defects kindred and is predicted to affect the enzymatic functionality of TLL1. A disease-segregating splice donor site mutation in MYH11 (c.4599+1delG) was identified in familial patent ductus arteriosus and found to disrupt normal splicing of MYH11 mRNA in the affected individual. CONCLUSIONS: Our findings demonstrate the clinical utility of WES to identify causative mutations in familial CHD and demonstrate the successful use of a CHD candidate gene list to allow for a more streamlined approach enabling rapid prioritization and identification of likely pathogenic variants from large WES data sets. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov; Unique Identifier: NCT0112048.


Asunto(s)
Exoma , Cardiopatías Congénitas/genética , Mutación , Adolescente , Células Cultivadas , Niño , Preescolar , Simulación por Computador , Análisis Mutacional de ADN/métodos , Bases de Datos Genéticas , Conducto Arterioso Permeable/diagnóstico , Conducto Arterioso Permeable/genética , Femenino , Factor de Transcripción GATA4/genética , Frecuencia de los Genes , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/terapia , Defectos del Tabique Interatrial/diagnóstico , Defectos del Tabique Interatrial/genética , Herencia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Modelos Genéticos , Tasa de Mutación , Cadenas Pesadas de Miosina/genética , Linaje , Fenotipo , Factores de Riesgo , Tetralogía de Fallot/diagnóstico , Tetralogía de Fallot/genética , Metaloproteinasas Similares a Tolloid/genética
11.
Hum Mol Genet ; 23(19): 5025-35, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24858909

RESUMEN

Mutations in GATA4 and TBX5 are associated with congenital heart defects in humans. Interaction between GATA4 and TBX5 is important for normal cardiac septation, but the underlying molecular mechanisms are not well understood. Here, we show that Gata4 and Tbx5 are co-expressed in the embryonic atria and ventricle, but after E15.5, ventricular expression of Tbx5 decreases. Co-localization and co-immunoprecipitation studies demonstrate an interaction of Gata4 and Tbx5 in the developing atria and ventricles, but the ventricular interaction declines after E14.5. Gata4(+/-);Tbx5(+/-) mouse embryos display decreased atrial and ventricular myocardial thickness at E11.5, prior to cardiac septation. To determine the cell lineage in which the interaction was functionally significant in vivo, mice heterozygous for Gata4 in the myocardium or endocardium and heterozygous for Tbx5 (Gata4(MyoDel/wt);Tbx5(+/-) and Gata4(EndoDel/wt);Tbx5(+/-), respectively) were generated. Gata4(MyoDel/wt);Tbx5(+/-) mice displayed embryonic lethality, thin myocardium with reduced cell proliferation, and atrioventricular septation defects similar to Gata4;Tbx5 compound heterozygotes while Gata4(EndoDel/wt);Tbx5(+/-) embryos were normal. Cdk4 and Cdk2, cyclin-dependent kinases required for myocardial development and septation were reduced in Gata4(+/-);Tbx5(+/-) hearts. Cdk4 is a known direct target of Gata4 and the regulation of Cdk2 in the developing heart has not been studied. Chromatin immunoprecipitation and transactivation studies demonstrate that Gata4 and Tbx5 directly regulate Cdk4 while only Tbx5 activates Cdk2 expression. These findings highlight the mechanisms by which disruption of the Gata4 and Tbx5 interaction in the myocardium contributes to cardiac septation defects in humans.


Asunto(s)
Factor de Transcripción GATA4/genética , Defectos de los Tabiques Cardíacos/genética , Defectos de los Tabiques Cardíacos/patología , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/genética , Animales , Linaje de la Célula/genética , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Epistasis Genética , Factor de Transcripción GATA4/deficiencia , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Defectos de los Tabiques Cardíacos/embriología , Heterocigoto , Ratones , Ratones Noqueados , Morfogénesis/genética , Miocardio/metabolismo , Miocardio/patología , Organogénesis/genética , Proteínas de Dominio T Box/deficiencia
13.
Artículo en Inglés | MEDLINE | ID: mdl-24334381

RESUMEN

Biochemical networks normally operate in the neighborhood of one of its multiple steady states. It may reach from one steady state to other within a finite time span. In this paper, a closed-loop control scheme is proposed to steer states of the glycolysis and glycogenolysis (GG) pathway from one of its steady states to other. The GG pathway is modeled in the synergism and saturation system formalism, known as S-system. This S-system model is linearized into the controllable Brunovsky canonical form using a feedback linearization technique. For closed-loop control, the linear-quadratic regulator (LQR) and the linear-quadratic gaussian (LQG) regulator are invoked to design a controller for tracking prespecified steady states. In the feedback linearization technique, a global diffeomorphism function is proposed that facilitates in achieving the regulation requirement. The robustness of the regulated GG pathway is studied considering input perturbation and with measurement noise.


Asunto(s)
Biología Computacional/métodos , Simulación por Computador , Glucogenólisis/fisiología , Glucólisis/fisiología , Modelos Biológicos , Retroalimentación , Modelos Lineales
14.
Infect Genet Evol ; 18: 247-56, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747831

RESUMEN

Development of a vaccine against Plasmodium falciparum infection is an urgent priority particularly because of widespread resistance to most traditionally used drugs. Multiple evidences point to apical membrane antigen-1(AMA-1) as a prime vaccine candidate directed against P. falciparum asexual blood-stages. To gain understanding of the genetic and demographic forces shaping the parasite sequence diversity in Kolkata, a part of Pfama-1 gene covering domain-I was sequenced from 100 blood samples of malaria patients. Statistical and phylogenetic analyses of the sequences were performed using DnaSP and MEGA. Very high haplotype diversity was detected both at nucleotide (0.998±0.002) and amino-acid (0.996±0.001) levels. An abundance of low frequency polymorphisms (Tajima's D=-1.190, Fu & Li's D(∗) and F(∗)=-3.068 and -2.722), unimodal mismatch distribution and a star-like median-joining network of ama-1 haplotypes indicated a recent population expansion among Kolkata parasites. The high minimum number of recombination events (Rm=26) and a significantly high dN/dS of 3.705 (P<0.0001) in Kolkata suggested recombination and positive selection as major forces in the generation and maintenance of ama-1 allelic diversity. To evaluate the impact of observed non-synonymous substitutions in the context of AMA-1 functionality, PatchDock and FireDock protein-protein interaction solutions were mapped between PfAMA-1-PfRON2 and PfAMA-1-host IgNAR. Alterations in the desolvation and global energies of PfAMA-1-PfRON2 interaction complexes at the hotspot contact residues were observed together with redistribution of surface electrostatic potentials at the variant alleles with respect to referent Pf3D7 sequence. Finally, a comparison of P. falciparum subpopulations in five Indian regional isolates retrieved from GenBank revealed a significant level of genetic differentiation (FST=0.084-0.129) with respect to Kolkata sequences. Collectively, our results indicated a very high allelic and haplotype diversity, a high recombination rate and a signature of natural selection favoring accumulation of non-synonymous substitutions that facilitated PfAMA-1-PfRON2 interaction and hence parasite growth in Kolkata clinical isolates.


Asunto(s)
Antígenos de Protozoos/genética , Proteínas de la Membrana/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Antígenos de Protozoos/química , Antígenos de Protozoos/metabolismo , Secuencia de Bases , Biología Computacional , ADN Protozoario , Evolución Molecular , Genética de Población , Haplotipos , Humanos , India , Malaria Falciparum/parasitología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Selección Genética , Alineación de Secuencia
15.
IEEE Trans Nanobioscience ; 12(2): 128-34, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23694697

RESUMEN

A novel analysis and synthesis framework is devised for synergism and saturation system, commonly known as S-system, for improving the robustness of the TCA cycle. In order to minimize the perturbation sensitivity, a measure of robustness of the network, a new design framework is proposed. The design constraints are formulated in computationally attractive convex optimization framework. The proposed multi-objective optimization problem, framed as Linear Matrix Inequality (LMI), makes a trade-off between the robustness and the control effort of the synthesized TCA cycle.


Asunto(s)
Ciclo del Ácido Cítrico , Dictyostelium/metabolismo , Modelos Biológicos , Simulación por Computador , Biología de Sistemas
16.
IEEE Trans Biomed Eng ; 60(2): 554-61, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204270

RESUMEN

A robust synthesis technique is devised for synergism and saturation systems, commonly known as S-systems, for controlling the steady states of the glycolysis-glycogenolysis pathway. The development of the robust biochemical network is essential owing to the fragile response to the perturbation of intrinsic and extrinsic parameters of the nominal S-system. The synthesis problem is formulated in a computationally attractive convex optimization framework. The linear matrix inequalities are framed to aim at the minimization of steady-state error, improvement of robustness, and utilization of minimum control input to the biochemical network.


Asunto(s)
Glucogenólisis , Glucólisis , Modelos Biológicos , Simulación por Computador , Cinética , Programas Informáticos , Biología de Sistemas/métodos
17.
PLoS One ; 7(10): e46441, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071570

RESUMEN

Genetic variations in toll-like receptors and cytokine genes of the innate immune pathways have been implicated in controlling parasite growth and the pathogenesis of Plasmodium falciparum mediated malaria. We previously published genetic association of TLR4 non-synonymous and TNF-α promoter polymorphisms with P.falciparum blood infection level and here we extend the study considerably by (i) investigating genetic dependence of parasite-load on interleukin-12B polymorphisms, (ii) reconstructing gene-gene interactions among candidate TLRs and cytokine loci, (iii) exploring genetic and functional impact of epistatic models and (iv) providing mechanistic insights into functionality of disease-associated regulatory polymorphisms. Our data revealed that carriage of AA (P = 0.0001) and AC (P = 0.01) genotypes of IL12B 3'UTR polymorphism was associated with a significant increase of mean log-parasitemia relative to rare homozygous genotype CC. Presence of IL12B+1188 polymorphism in five of six multifactor models reinforced its strong genetic impact on malaria phenotype. Elevation of genetic risk in two-component models compared to the corresponding single locus and reduction of IL12B (2.2 fold) and lymphotoxin-α (1.7 fold) expressions in patients'peripheral-blood-mononuclear-cells under TLR4Thr399Ile risk genotype background substantiated the role of Multifactor Dimensionality Reduction derived models. Marked reduction of promoter activity of TNF-α risk haplotype (C-C-G-G) compared to wild-type haplotype (T-C-G-G) with (84%) and without (78%) LPS stimulation and the loss of binding of transcription factors detected in-silico supported a causal role of TNF-1031. Significantly lower expression of IL12B+1188 AA (5 fold) and AC (9 fold) genotypes compared to CC and under-representation (P = 0.0048) of allele A in transcripts of patients' PBMCs suggested an Allele-Expression-Imbalance. Allele (A+1188C) dependent differential stability (2 fold) of IL12B-transcripts upon actinomycin-D treatment and observed structural modulation (P = 0.013) of RNA-ensemble were the plausible explanations for AEI. In conclusion, our data provides functional support to the hypothesis that de-regulated receptor-cytokine axis of innate immune pathway influences blood infection level in P. falciparum malaria.


Asunto(s)
Epistasis Genética , Inmunidad Innata/genética , Malaria Falciparum/genética , Polimorfismo Genético , Regiones no Traducidas 3' , Alelos , Línea Celular , Haplotipos , Humanos , Subunidad p40 de la Interleucina-12/genética , Malaria Falciparum/sangre , Malaria Falciparum/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/genética
18.
Infect Genet Evol ; 10(5): 686-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20307689

RESUMEN

Dysregulated innate immune responses due to inappropriate signaling by Toll-like receptors (TLRs) and aberrant production of pro-inflammatory cytokines are implicated in the immunopathology and disease outcome in Plasmodium falciparum malaria. This study investigates the relationship between polymorphic variability of candidate genes including TLR-2, -4, -9, tumor necrosis factor-alpha and lymphotoxin-alpha and blood infection level in Indian mild malaria patients. Genotyping was carried out by PCR-RFLP and sequencing. Association of parasite load with genotypes was examined using model based and model free approaches. Allele and haplotype based risk assessment for disease severity was performed by stratifying the patients into high and low parasitemic groups on the basis of a threshold value derived by employing a two-component mixture model and expectation-maximization algorithm. The mean parasitemia was significantly increased for variant homozygous genotype (C/C) at TNF-alpha promoter -1031 and major homozygous genotypes encoding Asp/Asp and Thr/Thr at codons 299 and 399, respectively, on TLR4 polypeptide. Individuals harboring combined genotype C/C-Asp/Asp-Thr/Thr on TNF-alpha and TLR4 presented the highest parasite load. The frequencies of variant allele C in TNF-1031 (OR=1.91 with 95% CI=1.24-2.94) and TNF-alpha promoter haplotypes C-C-G-G (OR=1.99 with 95% CI=1.21-3.27) and C-C-G-A (OR=2.96 with 95% CI=1.19-7.37) pertaining to loci TNF-1031/-857/-308/-238 were significantly elevated in the high parasitemic group. On the contrary, the frequencies of variant allele encoding Ile at 399 (OR=0.55 with 95% CI=0.32-0.94) and haplotype corresponding to Gly-Ile (299-399) (OR=0.51 with 95% CI=0.28-0.9) in TLR4 were higher in low parasitemic group. In silico analysis indicate differential binding of transcription factors to TNF-alpha promoter haplotypes and alteration in the surface charge distribution of the TLR4 variant proteins. Our results support a genetic role of TLR4 and TNF-alpha in controlling the blood infection level in mild malaria.


Asunto(s)
Malaria Falciparum , Parasitemia , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Polimorfismo de Nucleótido Simple , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/genética , Adolescente , Adulto , Alelos , Animales , Femenino , Genotipo , Haplotipos , Interacciones Huésped-Parásitos , Humanos , Linfotoxina-alfa/sangre , Malaria Falciparum/sangre , Malaria Falciparum/genética , Malaria Falciparum/inmunología , Masculino , Persona de Mediana Edad , Modelos Moleculares , Conformación Proteica , Factores de Riesgo , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/química , Receptor Toll-Like 9/genética , Adulto Joven
19.
Microbiology (Reading) ; 144 ( Pt 8): 2131-2140, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9720034

RESUMEN

It was found that Mycobacterium smegmatis is unable to utilize galactose as the sole carbon source because the sugar alone cannot induce galactokinase. However, galactokinase was induced by glutamate alone, and was further stimulated by galactose. Rifampicin completely inhibited the glutamate-mediated expression of galK in both the absence and presence of galactose. Extracellular cAMP stimulated the expression of the enzyme only in the presence of glutamate plus galactose. The galK gene from M. smegmatis, including its upstream promoter region, was cloned in a plasmid in Escherichia coli. The expression of kinase from these clones in E. coli was dependent on cAMP and its receptor protein (CRP). The expression of UDP-galactose 4-epimerase was constitutive. This and other evidence suggests that the galK gene is not linked to galT and galE in the mycobacterial genome. In a glutamate-independent galactose-utilizing mutant (gin-1 mutant) of M. smegmatis, galK was expressed in the absence of both galactose and glutamate, while in the presence of galactose this expression was increased twofold in the absence of glutamate and fourfold in its presence. Extracellularly added cAMP reduced the expression of the enzyme in the presence of galactose plus glutamate nearly to the basal level. It is proposed that in M. smegmatis the galK gene is expressed from two different promoters; the expression from one promoter is dependent on glutamate but not on galactose and cAMP, while that from the other requires all three components. The role of galactose is possibly to derepress the latter promoter.


Asunto(s)
AMP Cíclico/fisiología , Galactoquinasa/biosíntesis , Ácido Glutámico/fisiología , Mycobacterium/enzimología , Transporte Biológico Activo , AMP Cíclico/farmacología , Proteína Receptora de AMP Cíclico/fisiología , Sinergismo Farmacológico , Galactoquinasa/genética , Galactosa/metabolismo , Galactosa/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Mycobacterium/genética , Transcripción Genética , UDPglucosa 4-Epimerasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...