Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38542391

ABSTRACT

(1) Mesenchymal stem cells (MSCs) are a valuable cell model to study the bone pathology of Osteogenesis Imperfecta (OI), a rare genetic collagen-related disorder characterized by bone fragility and skeletal dysplasia. We aimed to generate a novel OI induced mesenchymal stem cell (iMSC) model from induced pluripotent stem cells (iPSCs) derived from human dermal fibroblasts. For the first time, OI iMSCs generation was based on an intermediate neural crest cell (iNCC) stage. (2) Skin fibroblasts from healthy individuals and OI patients were reprogrammed into iPSCs and subsequently differentiated into iMSCs via iNCCs. (3) Successful generation of iPSCs from acquired fibroblasts was confirmed with changes in cell morphology, expression of iPSC markers SOX2, NANOG, and OCT4 and three germ-layer tests. Following differentiation into iNCCs, cells presented increased iNCC markers including P75NTR, TFAP2A, and HNK-1 and decreased iPSC markers, shown to reach the iNCC stage. Induction into iMSCs was confirmed by the presence of CD73, CD105, and CD90 markers, low expression of the hematopoietic, and reduced expression of the iNCC markers. iMSCs were trilineage differentiation-competent, confirmed using molecular analyses and staining for cell-type-specific osteoblast, adipocyte, and chondrocyte markers. (4) In the current study, we have developed a multipotent in vitro iMSC model of OI patients and healthy controls able to differentiate into osteoblast-like cells.


Subject(s)
Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Osteogenesis Imperfecta , Humans , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Cell Differentiation , Collagen/metabolism , Skin , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics
2.
Front Endocrinol (Lausanne) ; 14: 1145125, 2023.
Article in English | MEDLINE | ID: mdl-36967771

ABSTRACT

Introduction: Osteogenesis Imperfecta is a rare genetic connective tissue disorder, characterized by skeletal dysplasia and fragile bones. Currently only two mouse models have been reported for haploinsufficient (HI) mild Osteogenesis Imperfecta (OI); the Col1a1 +/Mov13 (Mov13) and the Col1a1 +/-365 mouse model. The Mov13 mice were created by random insertion of the Mouse Moloney leukemia virus in the first intron of the Col1a1 gene, preventing the initiation of transcription. Since the development of the Mov13 mice almost four decades ago and its basic phenotypic characterization in the 90s, there have not been many further studies. We aimed to extensively characterize the Mov13 mouse model in order to critically evaluate its possible use for preclinical studies of HI OI. Methods: Bone tissue from ten heterozygous Mov13 and ten wild-type littermates (WT) C57BL/6J mice (50% males per group) was analyzed at eight weeks of age with bone histomorphometry, micro computed tomography (microCT), 3-point bending, gene expression of different collagens, as well as serum markers of bone turnover. Results: The Mov13 mouse presented a lower bone strength and impaired material properties based on our results of 3-point bending and microCT analysis respectively. In contrast, no significant differences were found for all histomorphometric parameters. In addition, no significant differences in Col1a1 bone expression were present, but there was a significant lower P1NP concentration, a bone formation marker, measured in serum. Furthermore, bone tissue of Mov13 mice presented significantly higher expression of collagens (Col1a2, Col5a1 and Col5a2), and bone metabolism markers (Bglap, Fgf23, Smad7, Edn1 and Eln) compared to WT. Finally, we measured a significantly lower Col1a1 expression in heart and skin tissue and also determined a higher expression of other collagens in the heart tissue. Conclusion: Although we did not detect a significant reduction in Col1a1 expression in the bone tissue, a change in bone structure and reduction in bone strength was noted. Regrettably, the variability of the bone phenotype and the appearance of severe lymphoma in adult Mov13 mice, does not favor their use for the testing of new long-term drug studies. As such, a new HI OI type 1 mouse model is urgently needed.


Subject(s)
Osteogenesis Imperfecta , Male , Mice , Animals , Female , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , X-Ray Microtomography , Mice, Inbred C57BL , Collagen/genetics , Phenotype
3.
Int J Mol Sci ; 24(3)2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36768622

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a catastrophic, ultra-rare disease of heterotopic ossification caused by genetic defects in the ACVR1 gene. The mutant ACVR1 receptor, when triggered by an inflammatory process, leads to heterotopic ossification of the muscles and ligaments. Activin A has been discovered as the main osteogenic ligand of the FOP ACVR1 receptor. However, the source of Activin A itself and the trigger of its production in FOP individuals have remained elusive. We used primary dermal fibroblasts from five FOP patients to investigate Activin A production and how this is influenced by inflammatory cytokines in FOP. FOP fibroblasts showed elevated Activin A production compared to healthy controls, both in standard culture and osteogenic transdifferentiation conditions. We discovered TGFß1 to be an FOP-specific stimulant of Activin A, shown by the upregulation of the INHBA gene and protein expression. Activin A and TGFß1 were both induced by BMP4 in FOP and control fibroblasts. Treatment with TNFα and IL6 produced negligible levels of Activin A and TGFß1 in both cell groups. We present for the first time TGFß1 as a triggering factor of Activin A production in FOP. As TGFß1 can promote the induction of the main driver of FOP, TGFß1 could also be considered a possible therapeutic target in FOP treatment.


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Humans , Myositis Ossificans/genetics , Myositis Ossificans/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Ossification, Heterotopic/genetics , Fibroblasts/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Mutation
4.
Sci Rep ; 12(1): 14686, 2022 08 29.
Article in English | MEDLINE | ID: mdl-36038602

ABSTRACT

Inherited bone disorders account for about 10% of documented Mendelian disorders and are associated with high financial burden. Their study requires osteoblasts which play a critical role in regulating the development and maintenance of bone tissue. However, bone tissue is not always available from patients. We developed a highly efficient platelet lysate-based approach to directly transdifferentiate skin-derived human fibroblasts to osteoblast-like cells. We extensively characterized our in vitro model by examining the expression of osteoblast-specific markers during the transdifferentiation process both at the mRNA and protein level. The transdifferentiated osteoblast-like cells showed significantly increased expression of a panel of osteogenic markers. Mineral deposition and ALP activity were also shown, confirming their osteogenic properties. RNA-seq analysis allowed the global study of changes in the transcriptome of the transdifferentiated cells. The transdifferentiated cells clustered separately from the primary fibroblasts with regard to the significantly upregulated genes indicating a distinct transcriptome profile; transdifferentiated osteoblasts also showed significant enrichment in gene expression related to skeletal development and bone mineralization. Our presented in vitro model may potentially contribute to the prospect of studying osteoblast-dependent disorders in patient-derived cells.


Subject(s)
Cell Transdifferentiation , Osteoblasts , Calcification, Physiologic/genetics , Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Fibroblasts , Humans , Osteoblasts/metabolism , Osteogenesis/genetics
5.
Genes (Basel) ; 13(3)2022 02 24.
Article in English | MEDLINE | ID: mdl-35327962

ABSTRACT

Osteogenesis imperfecta (OI) is a syndromic disorder of bone fragility with high variation in its clinical presentation. Equally variable is molecular aetiology; recessive forms are caused by approximately 20 different genes, many of which are directly implicated in collagen type I biosynthesis. Biallelic variants in prolyl 3-hydroxylase 1 (P3H1) are known to cause severe OI by affecting the competence of the prolyl 3-hydroxylation­cartilage associated protein­peptidyl-prolyl cis-trans isomerase B (P3H1-CRTAP-CyPB) complex, which acts on the Pro986 residue of collagen type I α 1 (COL1A1) and Pro707 collagen type I α 2 (COL1A2) chains. The investigation of an OI cohort of 146 patients in Vietnam identified 14 families with P3H1 variants. The c.1170+5G>C variant was found to be very prevalent (12/14) and accounted for 10.3% of the Vietnamese OI cohort. New P3H1 variants were also identified in this population. Interestingly, the c.1170+5G>C variants were found in families with the severe clinical Sillence types 2 and 3 but also the milder types 1 and 4. This is the first time that OI type 1 is reported in patients with P3H1 variants expanding the clinical spectrum. Patients with a homozygous c.1170+5G>C variant shared severe progressively deforming OI type 3: bowed long bones, deformities of ribcage, long phalanges and hands, bluish sclera, brachycephaly, and early intrauterine fractures. Although it remains unclear if the c.1170+5G>C variant constitutes a founder mutation in the Vietnamese population, its prevalence makes it valuable for the molecular diagnosis of OI in patients of the Kinh ethnicity. Our study provides insight into the clinical and genetic variation of P3H1-related OI in the Vietnamese population.


Subject(s)
Membrane Glycoproteins/genetics , Osteogenesis Imperfecta , Prolyl Hydroxylases/genetics , Proteoglycans/genetics , Asian People , Biological Variation, Population , Collagen Type I/genetics , Extracellular Matrix Proteins/genetics , Humans , Molecular Chaperones/genetics , Mutation , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Vietnam/epidemiology
6.
Nutr Neurosci ; 25(6): 1219-1230, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33236691

ABSTRACT

Objective: Vanishing white matter (VWM) is a genetic brain white matter disorder caused by mutations in eIF2B. eIF2B is central in the integrated stress response (ISR), during which its activity is inhibited by various cellular stresses. VWM is a chronic progressive disease with episodes of rapid neurological deterioration provoked by stresses. VWM patients and VWM mouse models show ISR deregulation in brain, correlating with chronic disease development. ISR inhibition ameliorates the chronic disease in VWM mice. The subacute deteriorations have not been modeled yet. We hypothesized that ISR activation could worsen disease progression in mice and model the episodic neurological deterioration.Method: We chose to activate the ISR by subjecting wild-type (wt) and VWM mice to an isocaloric low protein diet. This model would allow us to investigate the contribution of ISR activation in subacute decline in VWM.Results: We found that the low protein diet did not significantly affect amino acid levels nor ISR levels in wt and VWM mouse brain. Our study serendipitously led to the discovery of increased levels of glycine, asparagine and Fgf21 mRNA in VWM mouse brain irrespective of the dietary protein content. Strikingly, the ISR was not activated by the low protein diet in the liver of VWM in contrast to wt mice, due to a modest ISR deregulation in this organ.Discussion: A model for subacute neurological deterioration in VWM was not established. Possibly, ISR deregulation in VWM results in reduced ISR responsiveness.


Subject(s)
Leukoencephalopathies , White Matter , Animals , Brain/metabolism , Chronic Disease , Diet, Protein-Restricted , Disease Models, Animal , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Liver/metabolism , Mice , White Matter/metabolism
7.
Hum Genet ; 140(8): 1121-1141, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34169326

ABSTRACT

Osteogenesis Imperfecta (OI) comprises a heterogeneous group of patients who share bone fragility and deformities as the main characteristics, albeit with different degrees of severity. Phenotypic variation also exists in other connective tissue aspects of the disease, complicating disease classification and disease course prediction. Although collagen type I defects are long established as the primary cause of the bone pathology, we are still far from comprehending the complete mechanism. In the last years, the advent of next generation sequencing has triggered the discovery of many new genetic causes for OI, helping to draw its molecular landscape. It has become clear that, in addition to collagen type I genes, OI can be caused by multiple proteins connected to different parts of collagen biosynthesis. The production of collagen entails a complex process, starting from the production of the collagen Iα1 and collagen Iα2 chains in the endoplasmic reticulum, during and after which procollagen is subjected to a plethora of posttranslational modifications by chaperones. After reaching the Golgi organelle, procollagen is destined to the extracellular matrix where it forms collagen fibrils. Recently discovered mutations in components of the retrograde transport of chaperones highlight its emerging role as critical contributor of OI development. This review offers an overview of collagen regulation in the context of recent gene discoveries, emphasizing the significance of transport disruptions in the OI mechanism. We aim to motivate exploration of skeletal fragility in OI from the perspective of these pathways to identify regulatory points which can hint to therapeutic targets.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/biosynthesis , Osteoblasts/metabolism , Osteogenesis Imperfecta/metabolism , Procollagen/biosynthesis , Protein Processing, Post-Translational , Bone and Bones/pathology , Collagen Type I/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , High-Throughput Nucleotide Sequencing , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Osteoblasts/pathology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Procollagen/genetics , Protein Biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Transport , Severity of Illness Index
8.
Mol Genet Genomic Med ; 9(3): e1593, 2021 03.
Article in English | MEDLINE | ID: mdl-33432707

ABSTRACT

BACKGROUND: Vanishing white matter (VWM) is a leukodystrophy, caused by recessive mutations in eukaryotic initiation factor 2B (eIF2B)-subunit genes (EIF2B1-EIF2B5); 80% are missense mutations. Clinical severity is highly variable, with a strong, unexplained genotype-phenotype correlation. MATERIALS AND METHODS: With information from a recent natural history study, we severity-graded 97 missense mutations. Using in silico modeling, we created a new human eIF2B model structure, onto which we mapped the missense mutations. Mutated residues were assessed for location in subunits, eIF2B complex, and functional domains, and for information on biochemical activity. RESULTS: Over 50% of mutations have (ultra-)severe phenotypic effects. About 60% affect the ε-subunit, containing the catalytic domain, mostly with (ultra-)severe effects. About 55% affect subunit cores, with variable clinical severity. About 36% affect subunit interfaces, mostly with severe effects. Very few mutations occur on the external eIf2B surface, perhaps because they have minor functional effects and are tolerated. One external surface mutation affects eIF2B-substrate interaction and is associated with ultra-severe phenotype. CONCLUSION: Mutations that lead to (ultra-)severe disease mostly affect amino acids with pivotal roles in complex formation and function of eIF2B. Therapies for VWM are emerging and reliable mutation-based phenotype prediction is required for propensity score matching for trials and in the future for individualized therapy decisions.


Subject(s)
Eukaryotic Initiation Factor-2B/genetics , Leukoencephalopathies/genetics , Mutation, Missense , Phenotype , Humans , Molecular Dynamics Simulation , Protein Domains
9.
Ann Clin Transl Neurol ; 6(8): 1407-1422, 2019 08.
Article in English | MEDLINE | ID: mdl-31402619

ABSTRACT

OBJECTIVE: Vanishing white matter (VWM) is a fatal, stress-sensitive leukodystrophy that mainly affects children and is currently without treatment. VWM is caused by recessive mutations in eukaryotic initiation factor 2B (eIF2B) that is crucial for initiation of mRNA translation and its regulation during the integrated stress response (ISR). Mutations reduce eIF2B activity. VWM pathomechanisms remain unclear. In contrast with the housekeeping function of eIF2B, astrocytes are selectively affected in VWM. One study objective was to test our hypothesis that in the brain translation of specific mRNAs is altered by eIF2B mutations, impacting primarily astrocytes. The second objective was to investigate whether modulation of eIF2B activity could ameliorate this altered translation and improve the disease. METHODS: Mice with biallelic missense mutations in eIF2B that recapitulate human VWM were used to screen for mRNAs with altered translation in brain using polysomal profiling. Findings were verified in brain tissue from VWM patients using qPCR and immunohistochemistry. The compound ISRIB (for "ISR inhibitor") was administered to VWM mice to increase eIF2B activity. Its effect on translation, neuropathology, and clinical signs was assessed. RESULTS: In brains of VWM compared to wild-type mice we observed the most prominent changes in translation concerning ISR mRNAs; their expression levels correlated with disease severity. We substantiated these findings in VWM patients' brains. ISRIB normalized expression of mRNA markers, ameliorated brain white matter pathology and improved motor skills in VWM mice. INTERPRETATION: The present findings show that ISR deregulation is central in VWM pathomechanisms and a viable target for therapy.


Subject(s)
Acetamides/pharmacology , Cyclohexylamines/pharmacology , Eukaryotic Initiation Factor-2B/genetics , Leukoencephalopathies/drug therapy , Leukoencephalopathies/pathology , Activating Transcription Factor 4/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Cycle Proteins/metabolism , Cerebellum/drug effects , Corpus Callosum/drug effects , Eukaryotic Initiation Factor-2B/metabolism , Humans , Leukoencephalopathies/genetics , Mice , Mutation , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , White Matter/pathology
10.
Sci Rep ; 8(1): 3773, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491431

ABSTRACT

Vanishing white matter (VWM) is a genetic childhood white matter disorder, characterized by chronic as well as episodic, stress provoked, neurological deterioration. Treatment is unavailable and patients often die within a few years after onset. VWM is caused by recessive mutations in the eukaryotic initiation factor 2B (eIF2B). eIF2B regulates protein synthesis rates in every cell of the body. In normal cells, various types of cellular stress inhibit eIF2B activity and induce the integrated stress response (ISR). We have developed a VWM mouse model homozygous for the pathogenic Arg191His mutation in eIF2Bε (2b5 ho ), representative of the human disease. Neuropathological examination of VWM patient and mouse brain tissue suggests that astrocytes are primarily affected. We hypothesized that VWM astrocytes are selectively hypersensitive to ISR induction, resulting in a heightened response. We cultured astrocytes from wildtype and VWM mice and investigated the ISR in assays that measure transcriptional induction of stress genes, protein synthesis rates and cell viability. We investigated the effects of short- and long-term stress as well as stress recovery. We detected congruent results amongst the various assays and did not detect a hyperactive ISR in VWM mouse astrocytes.


Subject(s)
Astrocytes/pathology , Brain/pathology , Disease Models, Animal , Eukaryotic Initiation Factor-2B/physiology , Leukoencephalopathies/pathology , Stress, Physiological , Unfolded Protein Response , Animals , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Mice , Mice, Knockout , Mutation
11.
Front Cell Neurosci ; 11: 411, 2017.
Article in English | MEDLINE | ID: mdl-29375313

ABSTRACT

Vanishing white matter (VWM) is a leukodystrophy with predominantly early-childhood onset. Affected children display various neurological signs, including ataxia and spasticity, and die early. VWM patients have bi-allelic mutations in any of the five genes encoding the subunits of the eukaryotic translation factor 2B (eIF2B). eIF2B regulates protein synthesis rates under basal and cellular stress conditions. The underlying molecular mechanism of how mutations in eIF2B result in VWM is unknown. Previous studies suggest that brain white matter astrocytes are primarily affected in VWM. We hypothesized that the translation rate of certain astrocytic mRNAs is affected by the mutations, resulting in astrocytic dysfunction. Here we subjected primary astrocyte cultures of wild type (wt) and VWM (2b5ho ) mice to pulsed labeling proteomics based on stable isotope labeling with amino acids in cell culture (SILAC) with an L-azidohomoalanine (AHA) pulse to select newly synthesized proteins. AHA was incorporated into newly synthesized proteins in wt and 2b5ho astrocytes with similar efficiency, without affecting cell viability. We quantified proteins synthesized in astrocytes of wt and 2b5ho mice. This proteomic profiling identified a total of 80 proteins that were regulated by the eIF2B mutation. We confirmed increased expression of PROS1 in 2b5ho astrocytes and brain. A DAVID enrichment analysis showed that approximately 50% of the eIF2B-regulated proteins used the secretory pathway. A small-scale metabolic screen further highlighted a significant change in the metabolite 6-phospho-gluconate, indicative of an altered flux through the pentose phosphate pathway (PPP). Some of the proteins migrating through the secretory pathway undergo oxidative folding reactions in the endoplasmic reticulum (ER), which produces reactive oxygen species (ROS). The PPP produces NADPH to remove ROS. The proteomic and metabolomics data together suggest a deregulation of ER function in 2b5ho mouse astrocytes.

12.
J Clin Invest ; 126(4): 1512-24, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26974157

ABSTRACT

Vanishing white matter (VWM) is a fatal leukodystrophy that is caused by mutations in genes encoding subunits of eukaryotic translation initiation factor 2B (eIF2B). Disease onset and severity are codetermined by genotype. White matter astrocytes and oligodendrocytes are almost exclusively affected; however, the mechanisms of VWM development remain unclear. Here, we used VWM mouse models, patients' tissue, and cell cultures to investigate whether astrocytes or oligodendrocytes are the primary affected cell type. We generated 2 mouse models with mutations (Eif2b5Arg191His/Arg191His and Eif2b4Arg484Trp/Arg484Trp) that cause severe VWM in humans and then crossed these strains to develop mice with various mutation combinations. Phenotypic severity was highly variable and dependent on genotype, reproducing the clinical spectrum of human VWM. In all mutant strains, impaired maturation of white matter astrocytes preceded onset and paralleled disease severity and progression. Bergmann glia and retinal Müller cells, nonforebrain astrocytes that have not been associated with VWM, were also affected, and involvement of these cells was confirmed in VWM patients. In coculture, VWM astrocytes secreted factors that inhibited oligodendrocyte maturation, whereas WT astrocytes allowed normal maturation of VWM oligodendrocytes. These studies demonstrate that astrocytes are central in VWM pathomechanisms and constitute potential therapeutic targets. Importantly, astrocytes should also be considered in the pathophysiology of other white matter disorders.


Subject(s)
Astrocytes/metabolism , Leukoencephalopathies/metabolism , White Matter/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Leukoencephalopathies/physiopathology , Mice , Mice, Mutant Strains , Oligodendroglia/metabolism , Oligodendroglia/pathology , White Matter/pathology , White Matter/physiopathology
13.
FASEB J ; 25(2): 632-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21030693

ABSTRACT

The highly variable extracellular loops in G protein-coupled receptors (GPCRs) have been implicated in receptor activation, the mechanism of which is poorly understood. In a random mutagenesis screen on the human adenosine A(2B) receptor (A(2B)R) using the MMY24 Saccharomyces cerevisiae strain as a read-out system, we found that two residues in the first extracellular loop, a phenylalanine and an aspartic acid at positions 71 and 74, respectively, are involved in receptor activation. We subsequently performed further site-directed and site-saturation mutagenesis. These experiments revealed that the introduction of mutations at either of the identified positions results in a wide variety of receptor activation profiles, with changes in agonist potency, constitutive activity, and intrinsic activity. Radioligand binding studies showed that the changes in activation were not due to changes in receptor expression. We interpret these data in the light of the recently revealed structure of the adenosine A(2A)R, the closest homologue of the A(2B)R. The two residues are suggested to be vital in maintaining the tertiary structure of a ß sheet in the extracellular domain of the A(2B)R. We hypothesize that deterioration of structure in the extracellular domains of GPCRs compromises overall receptor structure with profound consequences for receptor activation and constitutive activity.


Subject(s)
Gene Expression Regulation/physiology , Receptor, Adenosine A2B/chemistry , Receptor, Adenosine A2B/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/chemistry , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Amino Acid Sequence , Aminopyridines/pharmacology , Humans , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Protein Conformation , Receptor, Adenosine A2B/genetics , Saccharomyces cerevisiae/metabolism , Triazines/pharmacology , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...