5 research outputs found

    Delineating the role of FANCA in glucose-stimulated insulin secretion in β cells through its protein interactome

    Get PDF
    Hyperinsulinemia affects 72% of Fanconi anemia (FA) patients and an additional 25% experience lowered glucose tolerance or frank diabetes. The underlying molecular mechanisms contributing to the dysfunction of FA pancreas β cells is unknown. Therefore, we sought to evaluate the functional role of FANCA, the most commonly mutated gene in FA, in glucosestimulated insulin secretion (GSIS). This study reveals that FANCA or FANCB knockdown impairs GSIS in human pancreas β cell line EndoC-βH3. To identify potential pathways by which FANCA might regulate GSIS, we employed a proteomics approach to identify FANCA protein interactions in EndoC-βH3 differentially regulated in response to elevated glucose levels. Glucose-dependent changes in the FANCA interaction network were observed, including increased association with other FA family proteins, suggesting an activation of the DNA damage response in response to elevated glucose levels. Reactive oxygen species increase in response to glucose stimulation and are necessary for GSIS in EndoC-βH3 cells. Glucose-induced activation of the DNA damage response was also observed as an increase in the DNA damage foci marker γ-H2AX and dependent upon the presence of reactive oxygen species. These results illuminate the role of FANCA in GSIS and its protein interactions regulated by glucose stimulation that may explain the prevalence of β cell-specific endocrinopathies in FA patients

    Delineating the role of FANCA in glucose-stimulated insulin secretion in β cells through its protein interactome

    Get PDF
    Hyperinsulinemia affects 72% of Fanconi anemia (FA) patients and an additional 25% experience lowered glucose tolerance or frank diabetes. The underlying molecular mechanisms contributing to the dysfunction of FA pancreas β cells is unknown. Therefore, we sought to evaluate the functional role of FANCA, the most commonly mutated gene in FA, in glucosestimulated insulin secretion (GSIS). This study reveals that FANCA or FANCB knockdown impairs GSIS in human pancreas β cell line EndoC-βH3. To identify potential pathways by which FANCA might regulate GSIS, we employed a proteomics approach to identify FANCA protein interactions in EndoC-βH3 differentially regulated in response to elevated glucose levels. Glucose-dependent changes in the FANCA interaction network were observed, including increased association with other FA family proteins, suggesting an activation of the DNA damage response in response to elevated glucose levels. Reactive oxygen species increase in response to glucose stimulation and are necessary for GSIS in EndoC-βH3 cells. Glucose-induced activation of the DNA damage response was also observed as an increase in the DNA damage foci marker γ-H2AX and dependent upon the presence of reactive oxygen species. These results illuminate the role of FANCA in GSIS and its protein interactions regulated by glucose stimulation that may explain the prevalence of β cell-specific endocrinopathies in FA patients

    The Impact of Type 1 Diabetes on Neural Dynamics

    No full text
    Type 1 diabetes (T1D) has been shown to affect the structure and function of the brain. The current study sought to uncover the neural dynamics underlying cognitive processing in adults with T1D using non-invasive neuroimaging. Adults with T1D and without major complications and a demographically-matched control group underwent magnetoencephalography (MEG) while performing tasks tapping attention, working memory and motor functioning. MEG data from each task was examined using a beamformer source imaging approach and probed statistically for group differences. Our results for the flanker attention task indicated that neural activity in the anterior cingulate, paracentral lobule, and parietal function was altered, such that participants with T1D had stronger flanker interference responses in parietal and weaker responses in anterior cingulate regions (p \u3c .001). Further, the overall strength of the anterior cingulate and paracentral lobule responses significantly correlated with disease duration, r = -.46, p = .006, and r = -.42, p = .013, respectively. Group differences in parietal-occipital responses were found throughout encoding and maintenance phases of the working memory task, where participants with T1D had stronger parietal activity in encoding and weakened parietal-occipital activity in maintenance (ps \u3c .01). Activity in several regions correlated with duration and A1C in participants with T1D (ps \u3c .01). Motor responses were also altered in participants with T1D, where specific frequency responses differentially predicted behavioral outcomes. These findings demonstrate significant alterations in neurophysiology underlying major cognitive processes, likely affecting outcomes in later life

    Delineating the role of FANCA in glucose-stimulated insulin secretion in β cells through its protein interactome.

    No full text
    Hyperinsulinemia affects 72% of Fanconi anemia (FA) patients and an additional 25% experience lowered glucose tolerance or frank diabetes. The underlying molecular mechanisms contributing to the dysfunction of FA pancreas β cells is unknown. Therefore, we sought to evaluate the functional role of FANCA, the most commonly mutated gene in FA, in glucose-stimulated insulin secretion (GSIS). This study reveals that FANCA or FANCB knockdown impairs GSIS in human pancreas β cell line EndoC-βH3. To identify potential pathways by which FANCA might regulate GSIS, we employed a proteomics approach to identify FANCA protein interactions in EndoC-βH3 differentially regulated in response to elevated glucose levels. Glucose-dependent changes in the FANCA interaction network were observed, including increased association with other FA family proteins, suggesting an activation of the DNA damage response in response to elevated glucose levels. Reactive oxygen species increase in response to glucose stimulation and are necessary for GSIS in EndoC-βH3 cells. Glucose-induced activation of the DNA damage response was also observed as an increase in the DNA damage foci marker γ-H2AX and dependent upon the presence of reactive oxygen species. These results illuminate the role of FANCA in GSIS and its protein interactions regulated by glucose stimulation that may explain the prevalence of β cell-specific endocrinopathies in FA patients
    corecore