Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
bioRxiv ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38766166

ABSTRACT

Tyrosine protein-kinase 2 (TYK2), a member of the Janus kinase family, mediates inflammatory signaling through multiple cytokines, including interferon-α (IFNα), interleukin (IL)-12, and IL-23. Missense mutations in TYK2 are associated with protection against type 1 diabetes (T1D), and inhibition of TYK2 shows promise in the management of other autoimmune conditions. Here, we evaluated the effects of specific TYK2 inhibitors (TYK2is) in pre-clinical models of T1D. First, human ß cells, cadaveric donor islets, and iPSC-derived islets were treated in vitro with IFNα in combination with a small molecule TYK2i (BMS-986165 or a related molecule BMS-986202). TYK2 inhibition prevented IFNα-induced ß cell HLA class I up-regulation, endoplasmic reticulum stress, and chemokine production. In co-culture studies, pre-treatment of ß cells with a TYK2i prevented IFNα-induced activation of T cells targeting an epitope of insulin. In vivo administration of BMS-986202 in two mouse models of T1D ( RIP-LCMV-GP mice and NOD mice) reduced systemic and tissue-localized inflammation, prevented ß cell death, and delayed T1D onset. Transcriptional phenotyping of pancreatic islets, pancreatic lymph nodes (PLN), and spleen during early disease pathogenesis highlighted a role for TYK2 inhibition in modulating signaling pathways associated with inflammation, translational control, stress signaling, secretory function, immunity, and diabetes. Additionally, TYK2i treatment changed the composition of innate and adaptive immune cell populations in the blood and disease target tissues, resulting in an immune phenotype with a diminished capacity for ß cell destruction. Overall, these findings indicate that TYK2i has beneficial effects in both the immune and endocrine compartments in models of T1D, thus supporting a path forward for testing TYK2 inhibitors in human T1D.

2.
bioRxiv ; 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38562689

ABSTRACT

We previously showed that miR-146a-5p is upregulated in pancreatic islets treated with pro-inflammatory cytokines. Others have reported that miR-146a-5p overexpression is associated with ß cell apoptosis and impaired insulin secretion. However, the molecular mechanisms mediating these effects remain elusive. To investigate the role of miR-146a-5p in ß cell function, we developed stable MIN6 cell lines to either overexpress or inhibit the expression of miR-146a-5p. Monoclonal cell populations were treated with pro-inflammatory cytokines (IL-1ß, IFNγ, and TNFα) to model T1D in vitro. We found that overexpression of miR-146a-5p increased cell death under conditions of inflammatory stress, whereas inhibition of miR-146a-5p reversed these effects. Additionally, inhibition of miR-146a-5p increased mitochondrial DNA copy number, respiration rate, and ATP production. Further, RNA sequencing data showed enrichment of pathways related to insulin secretion, apoptosis, and mitochondrial function when the expression levels of miR-146a-5p were altered. Finally, a temporal increase in miR-146a-5p expression levels and a decrease in mitochondria function markers was observed in islets derived from NOD mice. Collectively, these data suggest that miR-146a-5p may promote ß cell dysfunction and death during inflammatory stress by suppressing mitochondrial function.

3.
Genome Med ; 16(1): 43, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38515211

ABSTRACT

BACKGROUND: Limited understanding of the diversity of variants in the cystic fibrosis transmembrane conductance regulator (CFTR) gene across ancestries hampers efforts to advance molecular diagnosis of cystic fibrosis (CF). The consequences pose a risk of delayed diagnoses and subsequently worsened health outcomes for patients. Therefore, characterizing the spectrum of CFTR variants across ancestries is critical for revolutionizing molecular diagnoses of CF. METHODS: We analyzed 454,727 UK Biobank (UKBB) whole-exome sequences to characterize the diversity of CFTR variants across ancestries. Using the PanUKBB classification, the participants were assigned into six major groups: African (AFR), American/American Admixed (AMR), Central South Asia (CSA), East Asian (EAS), European (EUR), and Middle East (MID). We segregated ancestry-specific CFTR variants, including those that are CF-causing or clinically relevant. The ages of certain CF-causing variants were determined and analyzed for selective pressure effects, and curated phenotype analysis was performed for participants with clinically relevant CFTR genotypes. RESULTS: We detected over 4000 CFTR variants, including novel ancestry-specific variants, across six ancestries. Europeans had the most unique CFTR variants [n = 2212], while the American group had the least unique variants [n = 23]. F508del was the most prevalent CF-causing variant found in all ancestries, except in EAS, where V520F was the most prevalent. Common EAS variants such as 3600G > A, V456A, and V520, which appeared approximately 270, 215, and 338 generations ago, respectively, did not show evidence of selective pressure. Sixteen participants had two CF-causing variants, with two being diagnosed with CF. We found 154 participants harboring a CF-causing and varying clinical consequences (VCC) variant. Phenotype analysis performed for participants with multiple clinically relevant variants returned significant associations with CF and its pulmonary phenotypes [Bonferroni-adjusted p < 0.05]. CONCLUSIONS: We leveraged the UKBB database to comprehensively characterize the broad spectrum of CFTR variants across ancestries. The detection of over 4000 CFTR variants, including several ancestry-specific and uncharacterized CFTR variants, warrants the need for further characterization of their functional and clinical relevance. Overall, the presentation of classical CF phenotypes seen in non-CF diagnosed participants with more than one CF-causing variant indicates that they may benefit from current CFTR modulator therapies.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Humans , Biological Specimen Banks , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Exome , Mutation , UK Biobank
4.
Front Immunol ; 14: 1278184, 2023.
Article in English | MEDLINE | ID: mdl-37954612

ABSTRACT

Oral administration of antigen induces regulatory T cells (Treg) that can not only control local immune responses in the small intestine, but also traffic to the central immune system to deliver systemic suppression. Employing murine models of the inherited bleeding disorder hemophilia, we find that oral antigen administration induces three CD4+ Treg subsets, namely FoxP3+LAP-, FoxP3+LAP+, and FoxP3-LAP+. These T cells act in concert to suppress systemic antibody production induced by therapeutic protein administration. Whilst both FoxP3+LAP+ and FoxP3-LAP+ CD4+ T cells express membrane-bound TGF-ß (latency associated peptide, LAP), phenotypic, functional, and single cell transcriptomic analyses reveal distinct characteristics in the two subsets. As judged by an increase in IL-2Rα and TCR signaling, elevated expression of co-inhibitory receptor molecules and upregulation of the TGFß and IL-10 signaling pathways, FoxP3+LAP+ cells are an activated form of FoxP3+LAP- Treg. Whereas FoxP3-LAP+ cells express low levels of genes involved in TCR signaling or co-stimulation, engagement of the AP-1 complex members Jun/Fos and Atf3 is most prominent, consistent with potent IL-10 production. Single cell transcriptomic analysis further reveals that engagement of the Jun/Fos transcription factors is requisite for mediating TGFß expression. This can occur via an Il2ra dependent or independent process in FoxP3+LAP+ or FoxP3-LAP+ cells respectively. Surprisingly, both FoxP3+LAP+ and FoxP3-LAP+ cells potently suppress and induce FoxP3 expression in CD4+ conventional T cells. In this process, FoxP3-LAP+ cells may themselves convert to FoxP3+ Treg. We conclude that orally induced suppression is dependent on multiple regulatory cell types with complementary and interconnected roles.


Subject(s)
Interleukin-10 , T-Lymphocytes, Regulatory , Mice , Animals , Interleukin-10/metabolism , Transforming Growth Factor beta/metabolism , Forkhead Transcription Factors/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
5.
Diabetologia ; 66(11): 2042-2061, 2023 11.
Article in English | MEDLINE | ID: mdl-37537395

ABSTRACT

AIMS/HYPOTHESIS: Increased circulating levels of incompletely processed insulin (i.e. proinsulin) are observed clinically in type 1 and type 2 diabetes. Previous studies have suggested that Ca2+ signalling within beta cells regulates insulin processing and secretion; however, the mechanisms that link impaired Ca2+ signalling with defective insulin maturation remain incompletely understood. METHODS: We generated mice with beta cell-specific sarcoendoplasmic reticulum Ca2+ ATPase-2 (SERCA2) deletion (ßS2KO mice) and used an INS-1 cell line model of SERCA2 deficiency. Whole-body metabolic phenotyping, Ca2+ imaging, RNA-seq and protein processing assays were used to determine how loss of SERCA2 impacts beta cell function. To test key findings in human model systems, cadaveric islets were treated with diabetogenic stressors and prohormone convertase expression patterns were characterised. RESULTS: ßS2KO mice exhibited age-dependent glucose intolerance and increased plasma and pancreatic levels of proinsulin, while endoplasmic reticulum (ER) Ca2+ levels and glucose-stimulated Ca2+ synchronicity were reduced in ßS2KO islets. Islets isolated from ßS2KO mice and SERCA2-deficient INS-1 cells showed decreased expression of the active forms of the proinsulin processing enzymes PC1/3 and PC2. Additionally, immunofluorescence staining revealed mis-location and abnormal accumulation of proinsulin and proPC2 in the intermediate region between the ER and the Golgi (i.e. the ERGIC) and in the cis-Golgi in beta cells of ßS2KO mice. Treatment of islets from human donors without diabetes with high glucose and palmitate concentrations led to reduced expression of the active forms of the proinsulin processing enzymes, thus phenocopying the findings observed in ßS2KO islets and SERCA2-deficient INS-1 cells. Similar findings were observed in wild-type mouse islets treated with brefeldin A, a compound that perturbs ER-to-Golgi trafficking. CONCLUSIONS/INTERPRETATION: Taken together, these data highlight an important link between ER Ca2+ homeostasis and proinsulin processing in beta cells. Our findings suggest a model whereby chronic ER Ca2+ depletion due to SERCA2 deficiency impairs the spatial regulation of prohormone trafficking, processing and maturation within the secretory pathway. DATA AVAILABILITY: RNA-seq data have been deposited in the Gene Expression Omnibus (GEO; accession no.: GSE207498).


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Humans , Animals , Proinsulin/genetics , Proinsulin/metabolism , Insulin-Secreting Cells/metabolism , Diabetes Mellitus, Type 2/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Insulin/metabolism , Glucose/metabolism , Islets of Langerhans/metabolism
6.
Physiol Meas ; 44(10)2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37652033

ABSTRACT

Objective. To examine whether heart rate interval based rapid alert (HIRA) score derived from a combination model of heart rate variability (HRV) and modified early warning score (MEWS) is a surrogate for the detection of acute respiratory failure (ARF) in critically ill sepsis patients.Approach. Retrospective HRV analysis of sepsis patients admitted to Emory healthcare intensive care unit (ICU) was performed between sepsis-related ARF and sepsis controls without ARF. HRV measures such as time domain, frequency domain, and nonlinear measures were analyzed up to 24 h after patient admission, 1 h before the onset of ARF, and a random event time in the sepsis controls. Statistical significance was computed by the Wilcoxon Rank Sum test. Machine learning algorithms such as eXtreme Gradient Boosting and logistic regression were developed to validate the HIRA score model. The performance of HIRA and early warning score models were evaluated using the area under the receiver operating characteristic (AUROC).Main Results. A total of 89 (ICU) patients with sepsis were included in this retrospective cohort study, of whom 31 (34%) developed sepsis-related ARF and 58 (65%) were sepsis controls without ARF. Time-domain HRV for Electrocardiogram (ECG) Beat-to-Beat RR intervals strongly distinguished ARF patients from controls. HRV measures for nonlinear and frequency domains were significantly altered (p< 0.05) among ARF compared to controls. The HIRA score AUC: 0.93; 95% confidence interval (CI): 0.88-0.98) showed a higher predictive ability to detect ARF when compared to MEWS (AUC: 0.71; 95% CI: 0.50-0.90).Significance. HRV was significantly impaired across patients who developed ARF when compared to controls. The HIRA score uses non-invasively derived HRV and may be used to inform diagnostic and therapeutic decisions regarding the severity of sepsis and earlier identification of the need for mechanical ventilation.


Subject(s)
Respiratory Insufficiency , Sepsis , Humans , Retrospective Studies , Heart Rate/physiology , Sepsis/complications , Sepsis/diagnosis , Intensive Care Units , ROC Curve , Respiratory Insufficiency/complications , Respiratory Insufficiency/diagnosis , Transcription Factors , Cell Cycle Proteins , Histone Chaperones
7.
bioRxiv ; 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37398133

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that play a crucial role in modulating gene expression and are enriched in cell-derived extracellular vesicles (EVs). We investigated whether miRNAs from human islets and islet-derived EVs could provide insight into ß cell stress pathways activated during type 1 diabetes (T1D) evolution, therefore serving as potential disease biomarkers. We treated human islets from 10 cadaveric donors with IL-1ß and IFN-γ to model T1D ex vivo. MicroRNAs were isolated from islets and islet-derived EVs, and small RNA sequencing was performed. We found 20 and 14 differentially expressed (DE) miRNAs in cytokine- versus control-treated islets and EVs, respectively. Interestingly, the miRNAs found in EVs were mostly different from those found in islets. Only two miRNAs, miR-155-5p and miR-146a-5p, were upregulated in both islets and EVs, suggesting selective sorting of miRNAs into EVs. We used machine learning algorithms to rank DE EV-associated miRNAs, and developed custom label-free Localized Surface Plasmon Resonance-based biosensors to measure top ranked EVs in human plasma. Results from this analysis revealed that miR-155, miR-146, miR-30c, and miR-802 were upregulated and miR-124-3p was downregulated in plasma-derived EVs from children with recent-onset T1D. In addition, miR-146 and miR-30c were upregulated in plasma-derived EVs of autoantibody positive (AAb+) children compared to matched non-diabetic controls, while miR-124 was downregulated in both T1D and AAb+ groups. Furthermore, single-molecule fluorescence in situ hybridization confirmed increased expression of the most highly upregulated islet miRNA, miR-155, in pancreatic sections from organ donors with AAb+ and T1D.

8.
Diabetes ; 72(10): 1433-1445, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37478155

ABSTRACT

Altered endoplasmic reticulum (ER) Ca2+ signaling has been linked with ß-cell dysfunction and diabetes development. Store-operated Ca2+ entry replenishes ER Ca2+ through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor, stromal interaction molecule 1 (STIM1). For characterization of the in vivo impact of STIM1 loss, mice with ß-cell-specific STIM1 deletion (STIM1Δß mice) were generated and challenged with high-fat diet. Interestingly, ß-cell dysfunction was observed in female, but not male, mice. Female STIM1Δß mice displayed reductions in ß-cell mass, a concomitant increase in α-cell mass, and reduced expression of markers of ß-cell maturity, including MafA and UCN3. Consistent with these findings, STIM1 expression was inversely correlated with HbA1c levels in islets from female, but not male, human organ donors. Mechanistic assays demonstrated that the sexually dimorphic phenotype observed in STIM1Δß mice was due, in part, to loss of signaling through the noncanonical 17-ß estradiol receptor (GPER1), as GPER1 knockdown and inhibition led to a similar loss of expression of ß-cell maturity genes in INS-1 cells. Together, these data suggest that STIM1 orchestrates pancreatic ß-cell function and identity through GPER1-mediated estradiol signaling. ARTICLE HIGHLIGHTS: Store-operated Ca2+ entry replenishes endoplasmic reticulum (ER) Ca2+ through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor, stromal interaction molecule 1 (STIM1). ß-Cell-specific deletion of STIM1 results in a sexually dimorphic phenotype, with ß-cell dysfunction and loss of identity in female but not male mice. Expression of the noncanonical 17-ß estradiol receptor (GPER1) is decreased in islets of female STIM1Δß mice, and modulation of GPER1 levels leads to alterations in expression of ß-cell maturity genes in INS-1 cells.


Subject(s)
Calcium Channels , Membrane Proteins , Animals , Mice , Female , Humans , Membrane Proteins/metabolism , Calcium Channels/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Calcium/metabolism , Receptors, Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Calcium Signaling , GTP-Binding Proteins/metabolism
9.
Int J Periodontics Restorative Dent ; 43(6): 725-733, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37347615

ABSTRACT

Gingival recession is an apical shift of the gingival margin with exposure of the root surface to the oral cavity, which creates an esthetic problem. The present study was attempted to compare vestibular incision subperiosteal tunnel access (VISTA) with and without advanced platelet-rich fibrin (A-PRF) in the treatment of Miller Class I gingival recessions. A total of 24 patients were assigned randomly to either the test group (VISTA with A-PRF) or the control group (VISTA alone). Clinical parameters like recession depth, recession width, clinical attachment loss, width of keratinized gingiva, gingival thickness, and probing depth were recorded at baseline and at 3 and 6 months postoperatively. Intergroup comparison of mean root coverage (RC) in mm, %RC, change in width of keratinized gingiva and clinical attachment gain revealed no statistically significant difference (P > .05). Change in gingival thickness showed statistically significant improvement in test group. Within the limitations of this study, both treatment options (VISTA with A-PRF and VISTA alone) have resulted in predictable and comparable RC, with increased gingival thickness in the test group.


Subject(s)
Gingival Recession , Platelet-Rich Fibrin , Surgical Wound , Humans , Gingival Recession/surgery , Treatment Outcome , Tooth Root/surgery , Surgical Flaps/surgery , Gingiva/surgery
10.
J Breath Res ; 17(3)2023 06 01.
Article in English | MEDLINE | ID: mdl-37220742

ABSTRACT

Volatile organic compounds (VOCs) originating from human metabolic activities can be detected in, for example, breath, urine, feces, and blood. Thus, attention has been given to identifying VOCs from the above matrices. Studies identifying and measuring human blood VOCs are limited to those focusing on monitoring specific pollutants, or blood storage and/or decomposition. However, a comprehensive characterization of VOCs in human blood collected for routine diagnostic testing is lacking. In this pilot study, 72 blood-derived plasma samples were obtained from apparently healthy adult participants. VOCs were extracted from plasma using solid-phase microextraction and analyzed using comprehensive two-dimensional gas chromatography tandem time-of-flight mass spectrometry. Chromatographic data were aligned, and putative compound identities were assigned via spectral library comparison. All statistical analysis, including contaminant removal, data normalization, and transformation were performed usingR. We identified 401 features which we called the pan volatilome of human plasma. Of the 401 features, 34 were present in all the samples with less than 15% variance (core molecules), 210 were present in ⩾10% but <100% of the samples (accessory molecules), and 157 were present in less than 10% of the samples (rare molecules). The core molecules, consisting of aliphatic, aromatic, and carbonyl compounds were validated using 25 additional samples. The validation accuracy was 99.9%. Of the 34 core molecules, 2 molecules (octan-2-one and 4-methyl heptane) have been identified from the plasma samples for the first time. Overall, our pilot study establishes the methodology of profiling VOCs in human plasma and will serve as a resource for blood-derived VOCs that can complement future biomarker studies using different matrices with more heterogeneous cohorts.


Subject(s)
Volatile Organic Compounds , Adult , Humans , Gas Chromatography-Mass Spectrometry/methods , Volatile Organic Compounds/analysis , Pilot Projects , Breath Tests , Biomarkers
11.
Pharmacol Res Perspect ; 11(1): e01036, 2023 02.
Article in English | MEDLINE | ID: mdl-36537346

ABSTRACT

ABBV-47D11 is a neutralizing monoclonal antibody that targets a mutationally conserved hydrophobic pocket distal to the ACE2 binding site of SARS-CoV-2. This first-in-human safety, pharmacokinetics, and antiviral pharmacodynamic assessment in patients with COVID-19 provide an initial evaluation of this antibody that may allow further development. This multicenter, randomized, double-blind, and placebo-controlled single ascending dose study of ABBV-47D11 (180, 600, or 2400 mg) as an intravenous infusion, was in hospitalized and non-hospitalized (confined) adults with mild to moderate COVID-19. Primary outcomes were grade 3 or higher study drug-related adverse events and infusion-related reactions. Secondary outcomes were pharmacokinetic parameters and concentration-time profiles to Day 29, immunogenicity (anti-drug antibodies), and antiviral activity (change in RT-PCR viral load) from baseline to Days 15 and 29. ABBV-47D11 single doses up to 2400 mg were safe and tolerated and no safety signals were identified. The pharmacokinetics of ABBV-47D11 were linear and showed dose-proportional increases in serum concentrations with ascending doses. The exploratory anti-SARS-CoV-2 activity revealed a reduction of viral load at and above the 600 mg dose of ABBV-47D11 regardless of patient demographics and baseline characteristics, however; because of the high inter-individual variability and small sample size a statistical significance was not reached. There is potential for anti-SARS-CoV-2 activity with ABBV-47D11 doses of 600 mg or higher, which could be evaluated in future clinical trials designed and powered to assess viral load reductions and clinical benefit.


Subject(s)
COVID-19 , Adult , Humans , SARS-CoV-2 , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents , Antibodies, Neutralizing
12.
EBioMedicine ; 87: 104379, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36463755

ABSTRACT

BACKGROUND: Stress responses within the ß cell have been linked with both increased ß cell death and accelerated immune activation in type 1 diabetes (T1D). At present, information on the timing and scope of these responses as well as disease-related changes in islet ß cell protein expression during T1D development is lacking. METHODS: Data independent acquisition-mass spectrometry was performed on islets collected longitudinally from NOD mice and NOD-SCID mice rendered diabetic through T cell adoptive transfer. FINDINGS: In islets collected from female NOD mice at 10, 12, and 14 weeks of age, we found a time-restricted upregulation of proteins involved in stress mitigation and maintenance of ß cell function, followed by loss of expression of protective proteins that heralded diabetes onset. EIF2 signalling and the unfolded protein response, mTOR signalling, mitochondrial function, and oxidative phosphorylation were commonly modulated pathways in both NOD mice and NOD-SCID mice rendered acutely diabetic by T cell adoptive transfer. Protein disulphide isomerase A1 (PDIA1) was upregulated in NOD islets and pancreatic sections from human organ donors with autoantibody positivity or T1D. Moreover, PDIA1 plasma levels were increased in pre-diabetic NOD mice and in the serum of children with recent-onset T1D compared to non-diabetic controls. INTERPRETATION: We identified a core set of modulated pathways across distinct mouse models of T1D and identified PDIA1 as a potential human biomarker of ß cell stress in T1D. FUNDING: NIH (R01DK093954, DK127308, U01DK127786, UC4DK104166, R01DK060581, R01GM118470, and 5T32DK101001-09). VA Merit Award I01BX001733. JDRF (2-SRA-2019-834-S-B, 2-SRA-2018-493-A-B, 3-PDF-20016-199-A-N, 5-CDA-2022-1176-A-N, and 3-PDF-2017-385-A-N).


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Animals , Child , Female , Humans , Mice , Biomarkers/metabolism , Islets of Langerhans/metabolism , Mice, Inbred NOD , Mice, SCID , Protein Disulfide-Isomerases/metabolism , Proteomics , Insulin-Secreting Cells
13.
PLoS One ; 17(7): e0269775, 2022.
Article in English | MEDLINE | ID: mdl-35867653

ABSTRACT

Predictions of xenobiotic hepatic clearance in humans using in vitro-to-in vivo extrapolation methods are frequently inaccurate and problematic. Multiple strategies are being pursued to disentangle responsible mechanisms. The objective of this work is to evaluate the feasibility of using insights gained from independent virtual experiments on two model systems to begin unraveling responsible mechanisms. The virtual culture is a software analog of hepatocytes in vitro, and the virtual human maps to hepatocytes within a liver within an idealized model human. Mobile objects (virtual compounds) map to amounts of xenobiotics. Earlier versions of the two systems achieved quantitative validation targets for intrinsic clearance (virtual culture) and hepatic clearance (virtual human). The major difference between the two systems is the spatial organization of the virtual hepatocytes. For each pair of experiments (virtual culture, virtual human), hepatocytes are configured the same. Probabilistic rules govern virtual compound movements and interactions with other objects. We focus on highly permeable virtual compounds and fix their extracellular unbound fraction at one of seven values (0.05-1.0). Hepatocytes contain objects that can bind and remove compounds, analogous to metabolism. We require that, for a subset of compound properties, per-hepatocyte compound exposure and removal rates during culture experiments directly predict corresponding measures made during virtual human experiments. That requirement serves as a cross-system validation target; we identify compound properties that enable achieving it. We then change compound properties, ceteris paribus, and provide model mechanism-based explanations for when and why measures made during culture experiments under- (or over-) predict corresponding measures made during virtual human experiments. The results show that, from the perspective of compound removal, the organization of hepatocytes within virtual livers is more efficient than within cultures, and the greater the efficiency difference, the larger the underprediction. That relationship is noteworthy because most in vitro-to-in vivo extrapolation methods abstract away the structural organization of hepatocytes within a liver. More work is needed on multiple fronts, including the study of an expanded variety of virtual compound properties. Nevertheless, the results support the feasibility of the approach and plan.


Subject(s)
Hepatocytes , Liver , Hepatocytes/metabolism , Humans , Kinetics , Liver/metabolism , Metabolic Clearance Rate , Models, Biological
14.
Liver Int ; 42(6): 1278-1286, 2022 06.
Article in English | MEDLINE | ID: mdl-35220658

ABSTRACT

BACKGROUND AND AIMS: This study aimed to determine durability of sustained virologic response (SVR) in hepatitis C virus-infected participants treated with glecaprevir- and/or pibrentasvir-containing regimens. METHODS: M13-576, a rollover study, evaluated the durability of SVR in a follow-up period of approximately 3 years after hepatitis C virus genotype 1-6-infected participants received a glecaprevir- and/or pibrentasvir-containing regimen in previous phase 2/3 clinical trials. The primary efficacy endpoint was the percentage of participants maintaining SVR and the percentage of participants experiencing relapse or reinfections. Resistance-associated substitutions and safety outcomes related to liver progression were also assessed. RESULTS: Of 384 participants enroled, 377 participants were included in the as-observed population and 287 participants completed the study. In prior studies, 99.7% (376/377) of participants achieved SVR12; of those, an observed 99.5% (374/376) and 100% (286/286) completing the study, maintained SVR. After non-responder imputation of missing data, 286/376 participants (76%) maintained SVR. The participant previously not achieving SVR was a treatment-experienced male with compensated cirrhosis who had NS3 and NS5A substitutions at enrolment, which remained detectable for 12 months. Of the two participants not maintaining SVR, one was re-infected and one experienced late relapse at post-treatment week 60. Five participants (all with a fibrosis stage ≥F3) had hepatocellular carcinoma. No events were deemed related to glecaprevir/pibrentasvir. CONCLUSIONS: Glecaprevir/pibrentasvir demonstrated long-term durability of efficacy after SVR12 was achieved. Hepatic-related decompensation events were not seen. Owing to low incidence of virologic failure, conclusions were not drawn on persistence of resistance-associated substitutions.


Subject(s)
Hepatitis C, Chronic , Aminoisobutyric Acids , Antiviral Agents/therapeutic use , Benzimidazoles , Cyclopropanes , Follow-Up Studies , Genotype , Hepacivirus/genetics , Hepatitis C, Chronic/complications , Humans , Lactams, Macrocyclic , Leucine/analogs & derivatives , Male , Neoplasm Recurrence, Local , Proline/analogs & derivatives , Proline/therapeutic use , Pyrrolidines , Quinoxalines/therapeutic use , Sulfonamides , Sustained Virologic Response
15.
Cytometry B Clin Cytom ; 100(1): 19-32, 2021 01.
Article in English | MEDLINE | ID: mdl-34542933

ABSTRACT

With the morbidity and mortality associated with the COVID-19 pandemic that we are witnessing this year, the risks posed by emerging viral diseases to global health are all too obvious. This pandemic highlights the importance of antiviral drug discovery, which targets emerging viral pathogens, as well as existing pathogenic viruses that undergo continuous evolution. Drug discovery and development is a long and resource intensive process; however, the use of biomarkers can accelerate clinical development of antivirals by providing information regarding diagnosis of specific viral infections, status of infection, potential safety parameters, and antiviral responses. In clinical practice, many of the biomarkers initially utilized to support clinical development are also used for patient care. While viral load is a standard and essential biomarker used to detect the desired viral suppression induced by an antiviral agent, it has become apparent that additional biomarkers, whether related to the virus, the host or as a consequence of the drug's mechanistic effects, are also important for monitoring clinical outcomes associated with an antiviral therapy. This review summarizes the biomarkers used in the clinical development (as well as in clinical practice, where appropriate) of antiviral therapies for hepatitis C virus, hepatitis B virus, human immunodeficiency virus, and severe acute respiratory syndrome coronavirus 2.


Subject(s)
Antiviral Agents/therapeutic use , Biomarkers/analysis , Virus Diseases/drug therapy , Animals , Antiviral Agents/pharmacology , COVID-19/virology , Clinical Trials as Topic , Humans , SARS-CoV-2/physiology , COVID-19 Drug Treatment
16.
Ann Hepatol ; 20: 100257, 2021.
Article in English | MEDLINE | ID: mdl-32949786

ABSTRACT

INTRODUCTION AND OBJECTIVES: Glecaprevir/pibrentasvir is a highly effective and well tolerated treatment for hepatitis C infection. Brazilian patients were not included in the original development studies for glecaprevir/pibrentasvir. This study aimed to assess safety and efficacy of glecaprevir/pibrentasvir in treatment-naïve Brazilian adults without cirrhosis or with compensated cirrhosis. PATIENTS AND METHODS: EXPEDITION-3 was a Phase 3, open-label, multicenter study in treatment-naïve Brazilian adults with hepatitis C infection genotype 1-6. Patients without cirrhosis (F2 or F3) or with compensated cirrhosis (F4) received 8 or 12 weeks of glecaprevir/pibrentasvir, respectively. The primary efficacy endpoint was the rate of sustained virologic response at post-treatment Week 12. Secondary endpoints were on-treatment virologic failure and relapse rates. Baseline polymorphisms were assessed in NS3 and NS5A. Adverse events and laboratory abnormalities were monitored. RESULTS: 100 patients were enrolled, 75 received 8 weeks of treatment and 25 received 12 weeks; all patients completed treatment. Overall sustained virologic response at post-treatment Week 12 rate was high (98.0%; 98/100; 95% confidence interval: 93.0-99.4) and remained high regardless of baseline viral or host factors, including demographics, hepatitis C virus RNA levels, polymorphisms in NS3 and/or NS5A, genotype, and relevant comorbidities. 55% of patients reported ≥1 adverse event, the most common being headache (18.0%). Four patients reported serious adverse events; none were considered drug related or led to study drug discontinuation. No hepatic decompensations were observed. CONCLUSIONS: Glecaprevir/pibrentasvir was effective and well tolerated in treatment-naïve Brazilian patients with hepatitis C infection without cirrhosis and with compensated cirrhosis. TRIAL REGISTRATION: ClinicalTrials.gov NCT03219216.


Subject(s)
Benzimidazoles/therapeutic use , Hepacivirus , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Pyrrolidines/therapeutic use , Quinoxalines/therapeutic use , Sulfonamides/therapeutic use , Adult , Aged , Drug Administration Schedule , Drug Combinations , Female , Genotype , Humans , Male , Middle Aged , Sustained Virologic Response , Treatment Outcome
17.
J Indian Soc Periodontol ; 24(5): 481-485, 2020.
Article in English | MEDLINE | ID: mdl-33144779

ABSTRACT

The alveolar cleft is a bone-related developmental defect in the alveolar process of the maxillae, which is termed as cleft alveolus. The deformity occurs in 75% of the cleft palate and lip patients. Reconstructive surgery can provide both functional and esthetic benefits to such individuals. Conflicting opinions exist on the management of alveolar cleft, and these affect the treatment planning. We present the case of a 19-year-old female patient with a complaint of mobile teeth in the left frontal region of the upper jaw. On clinical examination, unilateral cleft alveolus was observed between the left lateral incisor and the canine region. A multidisciplinary approach was adopted, orthodontic treatment was started, and periodontal regenerative surgery was planned. This report also discusses the substitution of autogenous bone grafts with other materials such as allogenic grafts (demineralized freeze-dried bone allograft), platelet-rich plasma, platelet-rich fibrin membranes, and amnion membranes, which could serve as a new line of treatment for the condition.

18.
J Indian Soc Periodontol ; 24(4): 322-328, 2020.
Article in English | MEDLINE | ID: mdl-32831504

ABSTRACT

BACKGROUND: Several components of gingival crevicular fluid (GCF) reflect the course and predictability of periodontal disease and provide a pointer toward disease status. Potential biomarkers deoxypyridinoline (DPD), a metallophosphoesterase would correctly determine the presence of osteoclast-mediated bone turnover activity and seems to hold great promise as a predictive marker to determine bone destruction and active phases in the disease progression. AIM: The aim of the current study is proposed to investigate the biologic plausibility for the levels of DPD as biomarker in chronic periodontitis patients. MATERIALS AND METHODS: The present cross-sectional study comprised 15 periodontally healthy and 15 chronic periodontitis patients who were age and genders matched, recruited from the outpatient department of Periodontics. GCF and blood samples for DPD estimation were collected from all the patients and analyzed using enzyme-linked immunosorbent assay kit. The clinical parameters such as clinical attachment loss (CAL), probing pocket depth (PPD), modified gingival index, bleeding index , and plaque index were recorded. RESULTS: GCF DPD levels were significantly higher in chronic periodontitis patients when compared to periodontally healthy group. There were no significant correlations found among GCF and serum DPD levels with increasing age, gender, disease severity, and increase in PPD and CAL in both the groups. CONCLUSION: Within the limitations of this study, increased GCF DPD levels in chronic periodontitis can gauge ongoing periodontal destruction.

19.
Diabetes ; 69(11): 2364-2376, 2020 11.
Article in English | MEDLINE | ID: mdl-32820009

ABSTRACT

The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic ß-cell has not been tested. We used an informatics-based approach to develop a transcriptional signature of ß-cell GA stress using existing RNA sequencing and microarray data sets generated using human islets from donors with diabetes and islets where type 1 (T1D) and type 2 (T2D) diabetes had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated. In parallel, we generated an RNA-sequencing data set from human islets treated with brefeldin A (BFA), a known GA stress inducer. Overlapping the T1D and T2D groups with the BFA data set, we identified 120 and 204 differentially expressed genes, respectively. In both the T1D and T2D models, pathway analyses revealed that the top pathways were associated with GA integrity, organization, and trafficking. Quantitative RT-PCR was used to validate a common signature of GA stress that included ATF3, ARF4, CREB3, and COG6 Taken together, these data indicate that GA-associated genes are dysregulated in diabetes and identify putative markers of ß-cell GA stress.


Subject(s)
Computer Simulation , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation/physiology , Golgi Apparatus/physiology , Humans , Islets of Langerhans/metabolism , Models, Biological , Protein Array Analysis , Stress, Physiological
20.
Lancet Gastroenterol Hepatol ; 5(9): 839-849, 2020 09.
Article in English | MEDLINE | ID: mdl-32682494

ABSTRACT

BACKGROUND: Glecaprevir-pibrentasvir results in high rates of sustained virological response in patients with chronic hepatitis C virus (HCV) genotype 1-6 infection. Data for glecaprevir-pibrentasvir in non-Japanese Asian patients have been minimal. The aim of these studies was to assess the efficacy and safety of glecaprevir-pibrentasvir in treatment-naive and treatment-experienced Asian patients with chronic HCV genotype 1-6 infection without cirrhosis (VOYAGE-1) and with compensated cirrhosis (VOYAGE-2). METHODS: We did two phase 3 studies in treatment-naive and treatment-experienced patients with chronic HCV genotype 1-6 infection. VOYAGE-1 was a randomised, double-blind, placebo-controlled study that recruited patients without cirrhosis at 47 sites across China, South Korea, and Singapore. Randomisation was 2:1 with a fixed block size of three and stratified by geographical region and HCV genotype. Investigators, study site personnel, the study sponsor, and patients were masked to treatment allocation. VOYAGE-2 was a single-arm, open-label study that recruited patients with compensated cirrhosis at 34 sites across China and South Korea. Glecaprevir (300 mg) and pibrentasvir (120 mg) or placebo (VOYAGE-1, 2:1 ratio), administered as three tablets daily, was given for 8 weeks in patients without cirrhosis and for 12 weeks in those with cirrhosis (and for 16 weeks in treatment-experienced patients with genotype 3). The primary efficacy endpoint was the proportion of patients with a sustained virological response, defined as HCV RNA below the lower limit of quantification 12 weeks after the last dose of glecaprevir-pibrentasvir. We analysed efficacy and safety in all patients who received at least one dose of the study drug. These trials are registered with ClinicalTrials.gov, NCT03222583 (VOYAGE-1) and NCT03235349 (VOYAGE-2); both trials have been completed. This Article reports the results of the primary analysis for each study, undertaken when all patients who received glecaprevir-pibrentasvir (during the double-blind period in VOYAGE-1) had been followed up for 12 weeks following their last dose of study drug. Data from the double-blind period for placebo patients in VOYAGE-1 are also summarised. FINDINGS: Between Oct 4, 2017, and April 20, 2018, 546 patients with chronic HCV without cirrhosis were randomly assigned to treatment (363 to glecaprevir-pibrentasvir, 183 to placebo) in VOYAGE-1. One patient withdrew consent and did not receive treatment with glecaprevir-pibrentasvir. 352 of 362 patients who received glecaprevir-pibrentasvir achieved SVR12 (97·2% [95% CI 95·5-98·9]). Of 160 patients with compensated cirrhosis who were enrolled in VOYAGE-2 between Sept 29, 2017, and June 14, 2018, 159 of 160 achieved SVR12 (99·4%, 95% CI 98·2-100·0). 20 patients with HCV genotype 3b across both trials received glecaprevir-pibrentasvir; six of these patients were among the 11 patients who did not achieve SVR12. Upper respiratory tract infection was the most common adverse event (35 [10%] of 362 receiving glecaprevir-pibrentasvir and 18 [10%] of 183 receiving placebo in VOYAGE-1; 19 [12%] of 160 in VOYAGE-2). For patients receiving glecaprevir-pibrentasvir, serious adverse events occurred in three (<1%) of 362 patients in VOYAGE-1 and five (3%) of 160 patients in VOYAGE-2. Grade 3-4 adverse events in patients receiving glecaprevir-pibrentasvir occurred in five (1%) of 362 patients in VOYAGE-1 and six (4%) of 160 patients in VOYAGE-2; each type of event was experienced by at most one patient within a study. One patient with cirrhosis discontinued study drug because of an adverse event. INTERPRETATION: Glecaprevir-pibrentasvir showed high efficacy and an acceptable safety profile in these studies although responses were less common in the few patients with HCV genotype 3b. The results support the use of glecaprevir-pibrentasvir in these Asian populations. FUNDING: AbbVie.


Subject(s)
Benzimidazoles/therapeutic use , Hepatitis C, Chronic/drug therapy , Pyrrolidines/therapeutic use , Quinoxalines/therapeutic use , Sulfonamides/therapeutic use , Adult , Aged , Aminoisobutyric Acids , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Asia/epidemiology , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Case-Control Studies , Cyclopropanes , Double-Blind Method , Drug Combinations , Drug Therapy, Combination , Female , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/epidemiology , Humans , Lactams, Macrocyclic , Leucine/analogs & derivatives , Liver Cirrhosis/drug therapy , Liver Cirrhosis/epidemiology , Liver Cirrhosis/etiology , Male , Middle Aged , Placebos/administration & dosage , Prevalence , Proline/analogs & derivatives , Pyrrolidines/administration & dosage , Pyrrolidines/adverse effects , Quinoxalines/administration & dosage , Quinoxalines/adverse effects , Respiratory Tract Infections/chemically induced , Respiratory Tract Infections/epidemiology , Safety , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sustained Virologic Response , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...