Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell Rep Med ; 4(11): 101261, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37918404

ABSTRACT

In preclinical models, α-difluoromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor, delays the onset of type 1 diabetes (T1D) by reducing ß cell stress. However, the mechanism of DFMO action and its human tolerability remain unclear. In this study, we show that mice with ß cell ODC deletion are protected against toxin-induced diabetes, suggesting a cell-autonomous role of ODC during ß cell stress. In a randomized controlled trial (ClinicalTrials.gov: NCT02384889) involving 41 recent-onset T1D subjects (3:1 drug:placebo) over a 3-month treatment period with a 3-month follow-up, DFMO (125-1,000 mg/m2) is shown to meet its primary outcome of safety and tolerability. DFMO dose-dependently reduces urinary putrescine levels and, at higher doses, preserves C-peptide area under the curve without apparent immunomodulation. Transcriptomics and proteomics of DFMO-treated human islets exposed to cytokine stress reveal alterations in mRNA translation, nascent protein transport, and protein secretion. These findings suggest that DFMO may preserve ß cell function in T1D through islet cell-autonomous effects.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Mice , Animals , Diabetes Mellitus, Type 1/drug therapy , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors/pharmacology , Eflornithine/pharmacology , Eflornithine/therapeutic use , Putrescine/metabolism
2.
J Diabetes Sci Technol ; 17(4): 976-987, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35343269

ABSTRACT

OBJECTIVES: Achieving optimal glycemic outcomes in young children with type 1 diabetes (T1D) is challenging. This study examined the durability of continuous glucose monitoring (CGM) coupled with a family behavioral intervention (FBI) to improve glycemia. STUDY DESIGN: This one-year study included an initial 26-week randomized controlled trial of CGM with FBI (CGM+FBI) and CGM alone (Standard-CGM) compared with blood glucose monitoring (BGM), followed by a 26-week extension phase wherein the BGM Group received the CGM+FBI (BGM-Crossover) and both original CGM groups continued this technology. RESULTS: Time in range (70-180 mg/dL) did not improve with CGM use (CGM+FBI: baseline 37%, 52 weeks 41%; Standard-CGM: baseline 41%, 52 weeks 44%; BGM-Crossover: 26 weeks 38%, 52 weeks 40%). All three groups sustained decreases in hypoglycemia (<70 mg/dL) with CGM use (CGM+FBI: baseline 3.4%, 52 weeks 2.0%; Standard-CGM: baseline 4.1%, 52 weeks 2.1%; BGM-Crossover: 26 weeks 4.5%, 52 weeks 1.7%, P-values <.001). Hemoglobin A1c was unchanged with CGM use (CGM+FBI: baseline 8.3%, 52 weeks 8.2%; Standard-CGM: baseline 8.2%, 52 weeks 8.0%; BGM-Crossover: 26 weeks 8.1%, 52 weeks 8.3%). Sensor use remained high (52-week study visit: CGM+FBI 91%, Standard-CGM 92%, BGM-Crossover 88%). CONCLUSION: Over 12 months young children with T1D using newer CGM technology sustained reductions in hypoglycemia and, in contrast to prior studies, persistently wore CGM. However, pervasive hyperglycemia remained unmitigated. This indicates an urgent need for further advances in diabetes technology, behavioral support, and diabetes management educational approaches to optimize glycemia in young children.


Subject(s)
Diabetes Mellitus, Type 1 , Hyperglycemia , Hypoglycemia , Humans , Child , Child, Preschool , Blood Glucose , Diabetes Mellitus, Type 1/drug therapy , Blood Glucose Self-Monitoring
4.
Pediatr Diabetes ; 22(5): 734-741, 2021 08.
Article in English | MEDLINE | ID: mdl-33934456

ABSTRACT

BACKGROUND: Dasiglucagon, a next-generation, ready-to-use aqueous glucagon analog formulation, has been developed to treat severe hypoglycemia in individuals with diabetes. OBJECTIVE: The aim of this trial was to evaluate the safety and efficacy of dasiglucagon in pediatric individuals with type 1 diabetes (T1DM). Participants were children and adolescents (6-17 years) with T1DM. METHODS: In this randomized double-blind trial, 42 participants were randomly allocated (2:1:1) to a single subcutaneous (SC) injection of dasiglucagon (0.6 mg), placebo, or reconstituted glucagon (GlucaGen; dosed per label) during insulin-induced hypoglycemia. The primary endpoint was time to plasma glucose (PG) recovery (first PG increase ≥20 mg/dL after treatment initiation without rescue intravenous glucose). The primary comparison was dasiglucagon vs. placebo; glucagon acted as a reference. RESULTS: The median time (95% confidence interval) to PG recovery following SC injection was 10 min (8-12) for dasiglucagon vs. 30 min (20 to -) for placebo (P < .001); the median time for glucagon was 10 min (8-12), which did not include the time taken to reconstitute the lyophilized powder. PG recovery was achieved in all participants in the dasiglucagon and glucagon groups within 20 min of dosing compared to 2 out of 11 patients (18%) with placebo. The most frequent adverse events were nausea and vomiting, as expected with glucagon treatment. CONCLUSIONS: Consistent with adult phase 3 trials, dasiglucagon rapidly and effectively restored PG levels following insulin-induced hypoglycemia in children and adolescents with T1DM, with an overall safety profile similar to glucagon.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glucagon/analogs & derivatives , Hypoglycemia/drug therapy , Adolescent , Blood Glucose/drug effects , Blood Glucose/metabolism , Child , Diabetes Mellitus, Type 1/blood , Double-Blind Method , Female , Germany , Glucagon/administration & dosage , Humans , Hypoglycemia/chemically induced , Hypoglycemia/pathology , Injections, Subcutaneous , Insulin/therapeutic use , Male , Patient Acuity , Slovenia , Treatment Outcome , United States
5.
Diabetologia ; 64(8): 1822-1833, 2021 08.
Article in English | MEDLINE | ID: mdl-34003304

ABSTRACT

AIMS/HYPOTHESIS: The circadian clock influences both diabetes and immunity. Our goal in this study was to characterise more thoroughly the circadian patterns of immune cell populations and cytokines that are particularly relevant to the immune pathology of type 1 diabetes and thus fill in a current gap in our understanding of this disease. METHODS: Ten individuals with established type 1 diabetes (mean disease duration 11 years, age 18-40 years, six female) participated in a circadian sampling protocol, each providing six blood samples over a 24 h period. RESULTS: Daily ranges of population frequencies were sometimes large and possibly clinically significant. Several immune populations, such as dendritic cells, CD4 and CD8 T cells and their effector memory subpopulations, CD4 regulatory T cells, B cells and cytokine IL-6, exhibited statistically significant circadian rhythmicity. In a comparison with historical healthy control individuals, but using shipped samples, we observed that participants with type 1 diabetes had statistically significant phase shifts occurring in the time of peak occurrence of B cells (+4.8 h), CD4 and CD8 T cells (~ +5 h) and their naive and effector memory subsets (~ +3.3 to +4.5 h), and regulatory T cells (+4.1 h). An independent streptozotocin murine experiment confirmed the phase shifting of CD8 T cells and suggests that circadian dysrhythmia in type 1 diabetes might be an effect and not a cause of the disease. CONCLUSIONS/INTERPRETATION: Future efforts investigating this newly described aspect of type 1 diabetes in human participants are warranted. Peripheral immune populations should be measured near the same time of day in order to reduce circadian-related variation.


Subject(s)
Chronobiology Disorders/immunology , Circadian Rhythm/immunology , Diabetes Mellitus, Type 1/immunology , Immune System/physiology , Adolescent , Adult , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Circadian Clocks/genetics , Dendritic Cells/immunology , Female , Flow Cytometry , Humans , Interleukin-6/blood , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Young Adult
6.
Clin Diabetes ; 38(3): 266-272, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32699475

ABSTRACT

Physical activity is an important element of type 1 diabetes management, and hypoglycemia is a known risk. There are few data on strategies adolescents use to mitigate this risk. We surveyed 66 adolescents with type 1 diabetes who were 12-18 years of age about blood glucose monitoring, carbohydrate intake, and insulin management before, during, and after exercise. The adolescents completed the International Physical Activity Questionnaire-Short Form and the Children's Hypoglycemia Fear Survey. We found that adolescents with type 1 diabetes do not generally follow guidelines about glucose monitoring or about food and insulin adjustment around exercise. More targeted education and interventions are needed to improve adolescents' uptake of recommended behaviors and improve outcomes.

7.
J Clin Endocrinol Metab ; 104(10): 4356-4364, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31127824

ABSTRACT

CONTEXT: Use of continuous glucose monitoring (CGM) is increasing for insulin-requiring patients with diabetes. Although data on glycemic profiles of healthy, nondiabetic individuals exist for older sensors, assessment of glycemic metrics with new-generation CGM devices is lacking. OBJECTIVE: To establish reference sensor glucose ranges in healthy, nondiabetic individuals across different age groups using a current generation CGM sensor. DESIGN: Multicenter, prospective study. SETTING: Twelve centers within the T1D Exchange Clinic Network. PATIENTS OR PARTICIPANTS: Nonpregnant, healthy, nondiabetic children and adults (age ≥6 years) with nonobese body mass index. INTERVENTION: Each participant wore a blinded Dexcom G6 CGM, with once-daily calibration, for up to 10 days. MAIN OUTCOME MEASURES: CGM metrics of mean glucose, hyperglycemia, hypoglycemia, and glycemic variability. RESULTS: A total of 153 participants (age 7 to 80 years) were included in the analyses. Mean average glucose was 98 to 99 mg/dL (5.4 to 5.5 mmol/L) for all age groups except those over 60 years, in whom mean average glucose was 104 mg/dL (5.8 mmol/L). The median time between 70 to 140 mg/dL (3.9 to 7.8 mmol/L) was 96% (interquartile range, 93 to 98). Mean within-individual coefficient of variation was 17 ± 3%. Median time spent with glucose levels >140 mg/dL was 2.1% (30 min/d), and median time spent with glucose levels <70 mg/dL (3.9 mmol/L) was 1.1% (15 min/d). CONCLUSION: By assessing across age groups in a healthy, nondiabetic population, normative sensor glucose data have been derived and will be useful as a benchmark for future research studies.


Subject(s)
Blood Glucose Self-Monitoring/methods , Blood Glucose/analysis , Hyperglycemia/diagnosis , Hypoglycemia/diagnosis , Monitoring, Physiologic/methods , Adolescent , Adult , Age Factors , Aged , Benchmarking , Child , Circadian Rhythm , Female , Healthy Volunteers , Humans , Male , Middle Aged , Prospective Studies , Reference Values , Sex Factors , Young Adult
8.
Contemp Clin Trials ; 82: 60-65, 2019 07.
Article in English | MEDLINE | ID: mdl-31129370

ABSTRACT

Parents of young children with type 1 diabetes (T1D) experience unique, developmental challenges in managing their child's T1D, resulting in psychosocial distress. Only a small portion of young children reach glucose goals and adherence to diabetes devices that help improve T1D management have historically been low in this population. The purpose of this study is to test four interventions that couple developmentally tailored behavioral supports with education to optimize use of diabetes devices, improve glucose control, and reduce psychosocial distress for parents of young children with T1D. The study team designed four behavioral interventions, two aimed at improving glucose control and two aimed at optimizing use of diabetes devices. The goal of this paper is to describe the behavioral interventions developed for this study, including the results of a pilot test, and describe the methods and analysis plan to test this intervention strategy with ninety participants in a large-scale, randomized trial using a sequential multiple assignment randomization trial (SMART) design. A SMART design will permit a clinically relevant evaluation of the intervention strategy, as it allows multiple randomizations based on individualized assessments throughout the study instead of a fixed intervention dose seen in most traditional randomized controlled trials.


Subject(s)
Blood Glucose Self-Monitoring/methods , Blood Glucose/analysis , Diabetes Mellitus, Type 1/diagnosis , Adult , Blood Glucose Self-Monitoring/psychology , Child, Preschool , Diabetes Mellitus, Type 1/therapy , Humans , Infant , Parents/education , Parents/psychology , Patient Compliance/psychology , Patient Education as Topic/methods , Randomized Controlled Trials as Topic , Surveys and Questionnaires
9.
Pediatr Diabetes ; 19(1): 114-120, 2018 02.
Article in English | MEDLINE | ID: mdl-28429581

ABSTRACT

BACKGROUND: Fear of hypoglycemia is common in parents of young children with type 1 diabetes (T1D), but little is known about the specific fears that parents most often experience. Hypoglycemia fear has been associated with poorer glycemic control in older children, though not yet studied in a large cohort of very young children. MATERIALS AND METHODS: Parents of 549 children <7 years (mean 5.2 ± 1.2 years [19% <3 years]) with a mean diabetes duration of 2.4 ± 1.0 years (range 1-6 years) and mean HbA1c 8.2% ± 1.1% (66 ± 12 mmol/mol) registered in the T1D Exchange completed the worry scale of the Hypoglycemia Fear Survey modified for parents (HFS-P). RESULTS: Mean parental fear of hypoglycemia worry score was 36.1 ± 23.1 (possible range 0-100), with most frequent worries related to the child having a low while asleep and the child not recognizing a low. The mean worry score was not associated with the child's age, glycemic control, or recent severe hypoglycemic event. Parental worries about lows while sleeping were significantly higher in pump users than non-users (61% vs. 45%; P < .001), and tended to be higher in CGM users than non-users (62% vs 51%; P = .02). CONCLUSIONS: The greatest worries of parents of young children with T1D were related to hypoglycemia during sleep and other times/circumstances during which it would be difficult to detect hypoglycemia. Using advanced diabetes technologies may be an effort to temper fears about hypoglycemia during sleep, though the directionality of this relationship is undetermined. Additional studies can clarify this association and leverage use of diabetes technologies to improve glycemic control.


Subject(s)
Diabetes Mellitus, Type 1 , Fear , Hypoglycemia/chemically induced , Parents/psychology , Registries , Adult , Child , Child, Preschool , Circadian Rhythm , Female , Humans , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Male
10.
J Diabetes Sci Technol ; 12(2): 349-355, 2018 03.
Article in English | MEDLINE | ID: mdl-28895431

ABSTRACT

The purpose of this study is to examine timing of meal insulin and further determine whether an association exists between timing of meal insulin and missed meal insulin doses. The cohort included 4768 T1D Exchange clinic registry participants <26 years with type 1 diabetes ≥1 year. Chi-square tests, t-tests, and regression were used to assess the relationship between participant characteristics and timing of meal insulin and missed meal doses, respectively. Timing of meal insulin and association with missed meal doses was analyzed using logistic regression. In all, 21% reported administering insulin several minutes before, 44% immediately before, 10% during, and 24% after meal. Participants who gave insulin prior to a meal had significantly lower HbA1c than those who gave insulin during or after meal (8.4% ± 1.5% vs 8.8% ± 1.6%, adjusted P < .001), but no significant association was observed regarding DKA events. Those who reported missing ≥1 insulin dose per week had higher HbA1c (9.8% ± 1.9% vs 8.3% ± 1.3%, adjusted P < .001) and were more likely to experience at least one DKA event (9% vs 5%, adjusted P = .001) compared with those who rarely missed a meal insulin dose. Participants who reported administering insulin during or after a meal were more likely to report missing ≥1 meal insulin dose per week compared with those who administered insulin before a meal (28% vs 14%, adjusted P < .001). Premeal insulin was associated with lower HbA1c and fewer missed meal insulin doses. Providers may use this information to discuss the benefits of premeal insulin on glycemic control and adherence to therapy.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Medication Adherence/statistics & numerical data , Adolescent , Adult , Blood Glucose , Female , Humans , Male , Meals , Young Adult
11.
Ann Intern Med ; 167(2): 95-102, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28605777

ABSTRACT

Background: Debate exists as to whether the higher hemoglobin A1c (HbA1c) levels observed in black persons than in white persons are due to worse glycemic control or racial differences in the glycation of hemoglobin. Objective: To determine whether a racial difference exists in the relationship of mean glucose and HbA1c. Design: Prospective, 12-week observational study. Setting: 10 diabetes centers in the United States. Participants: 104 black persons and 104 white persons aged 8 years or older who had had type 1 diabetes for at least 2 years and had an HbA1c level of 6.0% to 12.0%. Measurements: Mean glucose concentration, measured by using continuous glucose monitoring and compared by race with HbA1c, glycated albumin, and fructosamine values. Results: The mean HbA1c level was 9.1% in black persons and 8.3% in white persons. For a given HbA1c level, the mean glucose concentration was significantly lower in black persons than in white persons (P = 0.013), which was reflected in mean HbA1c values in black persons being 0.4 percentage points (95% CI, 0.2 to 0.6 percentage points) higher than those in white persons for a given mean glucose concentration. In contrast, no significant racial differences were found in the relationship of glycated albumin and fructosamine levels with the mean glucose concentration (P > 0.20 for both comparisons). Limitation: There were too few participants with HbA1c levels less than 6.5% to generalize the results to such individuals. Conclusion: On average, HbA1c levels overestimate the mean glucose concentration in black persons compared with white persons, possibly owing to racial differences in the glycation of hemoglobin. However, because race only partially explains the observed HbA1c differences between black persons and white persons, future research should focus on identifying and modifying barriers impeding improved glycemic control in black persons with diabetes. Primary Funding Source: Helmsley Charitable Trust.


Subject(s)
Black People , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/ethnology , Glycated Hemoglobin/metabolism , White People , Adolescent , Adult , Blood Glucose Self-Monitoring , Child , Female , Fructosamine/blood , Glycation End Products, Advanced , Humans , Male , Middle Aged , Prospective Studies , Serum Albumin/metabolism , United States , Young Adult , Glycated Serum Albumin
13.
J Am Soc Nephrol ; 25(11): 2435-43, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24876116

ABSTRACT

Several genes, mainly involved in podocyte cytoskeleton regulation, have been implicated in familial forms of primary FSGS. We identified a homozygous missense mutation (p.P209L) in the TTC21B gene in seven families with FSGS. Mutations in this ciliary gene were previously reported to cause nephronophthisis, a chronic tubulointerstitial nephropathy. Notably, tubular basement membrane thickening reminiscent of that observed in nephronophthisis was present in patients with FSGS and the p.P209L mutation. We demonstrated that the TTC21B gene product IFT139, an intraflagellar transport-A component, mainly localizes at the base of the primary cilium in developing podocytes from human fetal tissue and in undifferentiated cultured podocytes. In contrast, in nonciliated adult podocytes and differentiated cultured cells, IFT139 relocalized along the extended microtubule network. We further showed that knockdown of IFT139 in podocytes leads to primary cilia defects, abnormal cell migration, and cytoskeleton alterations, which can be partially rescued by p.P209L overexpression, indicating its hypomorphic effect. Our results demonstrate the involvement of a ciliary gene in a glomerular disorder and point to a critical function of IFT139 in podocytes. Altogether, these data suggest that this homozygous TTC21B p.P209L mutation leads to a novel hereditary kidney disorder with both glomerular and tubulointerstitial damages.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cilia/physiology , Glomerulosclerosis, Focal Segmental/genetics , Microtubule-Associated Proteins/genetics , Podocytes/physiology , Adolescent , Adult , Animals , Cell Line, Transformed , Child , Cilia/pathology , Family Health , Female , Glomerulosclerosis, Focal Segmental/pathology , Haplotypes , Homozygote , Humans , Male , Mice , Mutation, Missense , Pedigree , Phenotype , Podocytes/pathology , Stress Fibers/pathology , Stress Fibers/physiology , Young Adult
14.
Nat Genet ; 46(3): 299-304, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509478

ABSTRACT

Monogenic disorders result from defects in a single gene. According to Mendel's laws, these disorders are inherited in either a recessive or dominant fashion. Autosomal-recessive disorders require a disease-causing variant on both alleles, and according to our current understanding, their pathogenicities are not influenced by each other. Here we present an autosomal-recessive disorder, nephrotic syndrome type 2 (MIM 600995), in which the pathogenicity of an NPHS2 allele encoding p.Arg229Gln depends on the trans-associated mutation. We show that, contrary to expectations, this allele leads to a disease phenotype only when it is associated specifically with certain 3' NPHS2 mutations because of an altered heterodimerization and mislocalization of the encoded p.Arg229Gln podocin. The disease-associated 3' mutations exert a dominant-negative effect on p.Arg229Gln podocin but behave as recessive alleles when associated with wild-type podocin. Therefore, the transmission rates for couples carrying the disease-associated mutations and p.Arg229Gln may be substantially different from those expected in autosomal-recessive disorders.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Nephrotic Syndrome/congenital , Adult , Amino Acid Substitution , Cell Membrane/metabolism , Child , Cohort Studies , Exons , Female , Gene Frequency , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/pathology , Protein Multimerization , Protein Structure, Quaternary
15.
J Clin Invest ; 123(12): 5179-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24270420

ABSTRACT

Identification of single-gene causes of steroid-resistant nephrotic syndrome (SRNS) has furthered the understanding of the pathogenesis of this disease. Here, using a combination of homozygosity mapping and whole human exome resequencing, we identified mutations in the aarF domain containing kinase 4 (ADCK4) gene in 15 individuals with SRNS from 8 unrelated families. ADCK4 was highly similar to ADCK3, which has been shown to participate in coenzyme Q10 (CoQ10) biosynthesis. Mutations in ADCK4 resulted in reduced CoQ10 levels and reduced mitochondrial respiratory enzyme activity in cells isolated from individuals with SRNS and transformed lymphoblasts. Knockdown of adck4 in zebrafish and Drosophila recapitulated nephrotic syndrome-associated phenotypes. Furthermore, ADCK4 was expressed in glomerular podocytes and partially localized to podocyte mitochondria and foot processes in rat kidneys and cultured human podocytes. In human podocytes, ADCK4 interacted with members of the CoQ10 biosynthesis pathway, including COQ6, which has been linked with SRNS and COQ7. Knockdown of ADCK4 in podocytes resulted in decreased migration, which was reversed by CoQ10 addition. Interestingly, a patient with SRNS with a homozygous ADCK4 frameshift mutation had partial remission following CoQ10 treatment. These data indicate that individuals with SRNS with mutations in ADCK4 or other genes that participate in CoQ10 biosynthesis may be treatable with CoQ10.


Subject(s)
Nephrotic Syndrome/genetics , Protein Kinases/physiology , Ubiquinone/analogs & derivatives , Adolescent , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use , Amino Acid Sequence , Animals , Cells, Cultured , Child , Consanguinity , Conserved Sequence , DNA Mutational Analysis , Disease Models, Animal , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drug Resistance , Exome/genetics , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Mitochondria/physiology , Molecular Sequence Data , Mutation , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/ultrastructure , Protein Kinases/deficiency , Protein Kinases/genetics , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Ubiquinone/antagonists & inhibitors , Ubiquinone/biosynthesis , Ubiquinone/metabolism , Ubiquinone/therapeutic use , Young Adult , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
16.
J Am Soc Nephrol ; 24(8): 1216-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23687361

ABSTRACT

LMX1B encodes a homeodomain-containing transcription factor that is essential during development. Mutations in LMX1B cause nail-patella syndrome, characterized by dysplasia of the patellae, nails, and elbows and FSGS with specific ultrastructural lesions of the glomerular basement membrane (GBM). By linkage analysis and exome sequencing, we unexpectedly identified an LMX1B mutation segregating with disease in a pedigree of five patients with autosomal dominant FSGS but without either extrarenal features or ultrastructural abnormalities of the GBM suggestive of nail-patella-like renal disease. Subsequently, we screened 73 additional unrelated families with FSGS and found mutations involving the same amino acid (R246) in 2 families. An LMX1B in silico homology model suggested that the mutated residue plays an important role in strengthening the interaction between the LMX1B homeodomain and DNA; both identified mutations would be expected to diminish such interactions. In summary, these results suggest that isolated FSGS could result from mutations in genes that are also involved in syndromic forms of FSGS. This highlights the need to include these genes in all diagnostic approaches to FSGS that involve next-generation sequencing.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , LIM-Homeodomain Proteins/genetics , Nail-Patella Syndrome/genetics , Transcription Factors/genetics , Adolescent , Adult , Child , Female , Genes, Dominant , Humans , Male , Middle Aged , Mutation , Pedigree , Sequence Analysis, DNA , Young Adult
17.
Diabetes Care ; 36(7): 2035-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23340893

ABSTRACT

OBJECTIVE: To assess the proportion of youth with type 1 diabetes under the care of pediatric endocrinologists in the United States meeting targets for HbA1c, blood pressure (BP), BMI, and lipids. RESEARCH DESIGN AND METHODS: Data were evaluated for 13,316 participants in the T1D Exchange clinic registry younger than 20 years old with type 1 diabetes for ≥1 year. RESULTS: American Diabetes Association HbA1c targets of <8.5% for those younger than 6 years, <8.0% for those 6 to younger than 13 years old, and <7.5% for those 13 to younger than 20 years old were met by 64, 43, and 21% of participants, respectively. The majority met targets for BP and lipids, and two-thirds met the BMI goal of <85th percentile. CONCLUSIONS: Most children with type 1 diabetes have HbA1c values above target levels. Achieving American Diabetes Association goals remains a significant challenge for the majority of youth in the T1D Exchange registry.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Adolescent , Adult , Body Mass Index , Child , Child, Preschool , Female , Glycated Hemoglobin/metabolism , Humans , Infant , Logistic Models , Male , Young Adult
18.
PLoS One ; 7(1): e29056, 2012.
Article in English | MEDLINE | ID: mdl-22253703

ABSTRACT

BACKGROUND: The cystathionine ß-synthase (CBS) gene, located on human chromosome 21q22.3, is a good candidate for playing a role in the Down Syndrome (DS) cognitive profile: it is overexpressed in the brain of individuals with DS, and it encodes a key enzyme of sulfur-containing amino acid (SAA) metabolism, a pathway important for several brain physiological processes. METHODOLOGY/PRINCIPAL FINDINGS: Here, we have studied the neural consequences of CBS overexpression in a transgenic mouse line (60.4P102D1) expressing the human CBS gene under the control of its endogenous regulatory regions. These mice displayed a ∼2-fold increase in total CBS proteins in different brain areas and a ∼1.3-fold increase in CBS activity in the cerebellum and the hippocampus. No major disturbance of SAA metabolism was observed, and the transgenic mice showed normal behavior in the rotarod and passive avoidance tests. However, we found that hippocampal synaptic plasticity is facilitated in the 60.4P102D1 line. CONCLUSION/SIGNIFICANCE: We demonstrate that CBS overexpression has functional consequences on hippocampal neuronal networks. These results shed new light on the function of the CBS gene, and raise the interesting possibility that CBS overexpression might have an advantageous effect on some cognitive functions in DS.


Subject(s)
Brain/physiology , Cystathionine beta-Synthase/metabolism , Amino Acids, Sulfur/metabolism , Animals , Behavior, Animal/physiology , Blotting, Western , Gene Dosage , Humans , Long-Term Potentiation/physiology , Metabolic Networks and Pathways , Metabolome , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Synapses/metabolism , Synaptic Transmission/physiology , Transgenes/genetics
19.
Exp Cell Res ; 317(20): 2800-13, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-21993218

ABSTRACT

Lamins A and C are nuclear intermediate filament proteins expressed in most differentiated somatic cells. Previous data suggested that prelamin A, the lamin A precursor, accumulates in some lipodystrophy syndromes caused by mutations in the lamin A/C gene, and binds and inactivates the sterol regulatory element binding protein 1 (SREBP1). Here we show that, in vitro, the tail regions of prelamin A, lamin A and lamin C bind a polypeptide of SREBP1. Such interactions also occur in HeLa cells, since expression of lamin tail regions impedes nucleolar accumulation of the SREBP1 polypeptide fused to a nucleolar localization signal sequence. In addition, the tail regions of A-type lamin variants that occur in Dunnigan-type familial partial lipodystrophy of (R482W) and Hutchison Gilford progeria syndrome (∆607-656) bind to the SREBP1 polypeptide in vitro, and the corresponding FLAG-tagged full-length lamin variants co-immunoprecipitate the SREBP1 polypeptide in cells. Overexpression of wild-type A-type lamins and variants favors SREBP1 polypeptide localization at the intranuclear periphery, suggesting its sequestration. Our data support the hypothesis that variation of A-type lamin protein level and spatial organization, in particular due to disease-linked mutations, influences the sequestration of SREBP1 at the nuclear envelope and thus contributes to the regulation of SREBP1 function.


Subject(s)
Lamin Type A/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , HeLa Cells , Humans , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Lipodystrophy, Familial Partial/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Progeria/genetics , Progeria/metabolism , Protein Binding , Protein Precursors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...