Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 330
Filtrar
1.
medRxiv ; 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38883740

RESUMO

Outcomes for adult patients with a high-grade glioma continue to be dismal and new treatment paradigms are urgently needed. To optimize the opportunity for discovery, we performed a phase 0/1 dose-escalation clinical trial that investigated tumor pharmacokinetics, pharmacodynamics, and single nucleus transcriptomics following combined ribociclib (CDK4/6 inhibitor) and everolimus (mTOR inhibitor) treatment in recurrent high-grade glioma. Patients with a recurrent high-grade glioma (n = 24) harboring 1) CDKN2A / B deletion or CDK4 / 6 amplification, 2) PTEN loss or PIK3CA mutations, and 3) wild-type retinoblastoma protein (Rb) were enrolled. Patients received neoadjuvant ribociclib and everolimus treatment and no dose-limiting toxicities were observed. The median unbound ribociclib concentrations in Gadolinium non-enhancing tumor regions were 170 nM (range, 65 - 1770 nM) and 634 nM (range, 68 - 2345 nM) in patients receiving 5 days treatment at the daily dose of 400 and 600 mg, respectively. Unbound everolimus concentrations were below the limit of detection (< 0.1 nM) in both enhancing and non-enhancing tumor regions at all dose levels. We identified a significant decrease in MIB1 positive cells suggesting ribociclib-associated cell cycle inhibition. Single nuclei RNAseq (snRNA) based comparisons of 17 IDH-wild-type on-trial recurrences to 31 IDH-wild-type standard of care treated recurrences data demonstrated a significantly lower fraction of cycling and neural progenitor-like (NPC-like) malignant cell populations. We validated the CDK4/6 inhibitor-directed malignant cell state shifts using three patient-derived cell lines. The presented clinical trial highlights the value of integrating pharmacokinetics, pharmacodynamics, and single nucleus transcriptomics to assess treatment effects in phase 0/1 surgical tissues, including malignant cell state shifts. ClinicalTrials.gov identifier: NCT03834740 .

2.
bioRxiv ; 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38853909

RESUMO

BACKGROUND: MYBPC3 , encoding cardiac myosin binding protein-C (cMyBP-C), is the most mutated gene known to cause hypertrophic cardiomyopathy (HCM). However, since little is known about the underlying etiology, additional in vitro studies are crucial to defining the underlying molecular mechanisms. Accordingly, this study aimed to investigate the molecular mechanisms underlying the pathogenesis of HCM associated with a polymorphic variant (D389V) in MYBPC3 by using human-induced pluripotent stem cell (hiPSC)-derived cardiac organoids (hCOs). METHODS: The hiPSC-derived cardiomyocytes (hiPSC-CMs) and hCOs were generated from human subjects to define the molecular, cellular, and functional changes caused by the MYBPC3 D389V variant. This variant is associated with increased fractional shortening and is highly prevalent in South Asian descendants. Recombinant C0-C2, N'-region of cMyBP-C (wildtype and D389V), and myosin S2 proteins were also utilized to perform binding and motility assays in vitro . RESULTS: Confocal and electron microscopic analyses of hCOs generated from noncarriers (NC) and carriers of the MYBPC3 D389V variant revealed the presence of highly organized sarcomeres. Furthermore, functional experiments showed hypercontractility with increased contraction velocity, faster calcium cycling, and faster contractile kinetics in hCOs expressing MYBPC3 D389V than NC hCOs. Interestingly, significantly increased cMyBP-C phosphorylation in MYBPC3 D389V hCOs was observed, but without changes in total protein levels, in addition to higher oxidative stress and lower mitochondrial membrane potential (ΔΨm). Next, spatial mapping revealed the presence of endothelial cells, fibroblasts, macrophages, immune cells, and cardiomyocytes in the hCOs. The hypercontractile function was significantly improved after treatment with the myosin inhibitor mavacamten (CAMZYOS®) in MYBPC3 D389V hCOs. Lastly, various in vitro binding assays revealed a significant loss of affinity in the presence of MYBPC3 D389V with myosin S2 region as a likely mechanism for hypercontraction. CONCLUSIONS: Conceptually, we showed the feasibility of assessing the functional and molecular mechanisms of HCM using highly translatable hCOs through pragmatic experiments that led to determining the MYBPC3 D389V hypercontractile phenotype, which was rescued by administration of a myosin inhibitor. Novelty and Significance: What Is Known?: MYBPC3 mutations have been implicated in hypertrophic cardiomyopathy. D389V is a polymorphic variant of MYBPC3 predicted to be present in 53000 US South Asians owing to the founder effect. D389V carriers have shown evidence of hyperdynamic heart, and human-induced pluripotent stem cells (hiPSC)-derived cardiomyocytes with D389V show cellular hypertrophy and irregular calcium transients. The molecular mechanism by which the D389V variant develops pathological cardiac dysfunction remains to be conclusively determined.What New Information Does This Article Contribute ?: The authors leveraged a highly translational cardiac organoid model to explore the role of altered cardiac calcium handling and cardiac contractility as a common pathway leading to pathophysiological phenotypes in patients with early HCM. The MYBPC3 D389V -mediated pathological pathway is first studied here by comparing functional properties using three-dimensional cardiac organoids differentiated from hiPSC and determining the presence of hypercontraction. Our data demonstrate that faster sarcomere kinetics resulting from lower binding affinity between D389V-mutated cMyBP-C protein and myosin S2, as evidenced by in vitro studies, could cause hypercontractility which was rescued by administration of mavacamten (CAMZYOS®), a myosin inhibitor. In addition, hypercontractility causes secondary mitochondrial defects such as higher oxidative stress and lower mitochondrial membrane potential (ΔΨm), highlighting a possible early adaptive response to primary sarcomeric changes. Early treatment of MYBPC3 D389V carriers with mavacamten may prevent or reduce early HCM-related pathology. GRAPHICAL ABSTRACT: A graphical abstract is available for this article.

4.
bioRxiv ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38746225

RESUMO

During heart failure, gene and protein expression profiles undergo extensive compensatory and pathological remodeling. We previously observed that fast skeletal myosin binding protein-C (fMyBP-C) is upregulated in diseased mouse hearts. While fMyBP-C shares significant homology with its cardiac paralog, cardiac myosin binding protein-C (cMyBP-C), there are key differences that may affect cardiac function. However, it is unknown if the expression of fMyBP-C expression in the heart is a pathological or compensatory response. We aim to elucidate the cardiac consequence of either increased or knockout of fMyBP-C expression. To determine the sufficiency of fMyBP-C to cause cardiac dysfunction, we generated cardiac-specific fMyBP-C over-expression mice. These mice were further crossed into a cMyBP-C null model to assess the effect of fMyBP-C in the heart in the complete absence of cMyBP-C. Finally, fMyBP-C null mice underwent transverse aortic constriction (TAC) to define the requirement of fMyBP-C during heart failure development. We confirmed the upregulation of fMyBP-C in several models of cardiac disease, including the use of lineage tracing. Low levels of fMyBP-C caused mild cardiac remodeling and sarcomere dysfunction. Exclusive expression of fMyBP-C in a heart failure model further exacerbated cardiac pathology. Following 8 weeks of TAC, fMyBP-C null mice demonstrated greater protection against heart failure development. Mechanistically, this may be due to the differential regulation of the myosin super-relaxed state. These findings suggest that the elevated expression of fMyBP-C in diseased hearts is a pathological response. Targeted therapies to prevent upregulation of fMyBP-C may prove beneficial in the treatment of heart failure. Significance Statement: Recently, the sarcomere - the machinery that controls heart and muscle contraction - has emerged as a central target for development of cardiac therapeutics. However, there remains much to understand about how the sarcomere is modified in response to disease. We recently discovered that a protein normally expressed in skeletal muscle, is present in the heart in certain settings of heart disease. How this skeletal muscle protein affects the function of the heart remained unknown. Using genetically engineered mouse models to modulate expression of this skeletal muscle protein, we determined that expression of this skeletal muscle protein in the heart negatively affects cardiac performance. Importantly, deletion of this protein from the heart could improve heart function suggesting a possible therapeutic avenue.

5.
J Proteome Res ; 23(4): 1285-1297, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38480473

RESUMO

C18ORF25 was recently shown to be phosphorylated at S67 by AMP-activated protein kinase (AMPK) in the skeletal muscle, following acute exercise in humans. Phosphorylation was shown to improve the ex vivo skeletal muscle contractile function in mice, but our understanding of the molecular mechanisms is incomplete. Here, we profiled the interactome of C18ORF25 in mouse myotubes using affinity purification coupled to mass spectrometry. This analysis included an investigation of AMPK-dependent and S67-dependent protein/protein interactions. Several nucleocytoplasmic and contractile-associated proteins were identified, which revealed a subset of GTPases that associate with C18ORF25 in an AMPK- and S67 phosphorylation-dependent manner. We confirmed that C18ORF25 is localized to the nucleus and the contractile apparatus in the skeletal muscle. Mice lacking C18Orf25 display defects in calcium handling specifically in fast-twitch muscle fibers. To investigate these mechanisms, we developed an integrated single fiber physiology and single fiber proteomic platform. The approach enabled a detailed assessment of various steps in the excitation-contraction pathway including SR calcium handling and force generation, followed by paired single fiber proteomic analysis. This enabled us to identify >700 protein/phenotype associations and 36 fiber-type specific differences, following loss of C18Orf25. Taken together, our data provide unique insights into the function of C18ORF25 and its role in skeletal muscle physiology.


Assuntos
Proteínas Quinases Ativadas por AMP , Fibras Musculares de Contração Lenta , Camundongos , Humanos , Animais , Fibras Musculares de Contração Lenta/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Proteômica/métodos , Cálcio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contração Rápida/metabolismo , Músculo Esquelético/metabolismo , Contração Muscular , Espectrometria de Massas
7.
Lancet Rheumatol ; 6(3): e168-e177, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301682

RESUMO

BACKGROUND: Mycophenolate mofetil is an immunosuppressant commonly used to treat systemic lupus erythematosus (SLE) and lupus nephritis. It is a known teratogen associated with significant toxicities, including an increased risk of infections and malignancies. Mycophenolate mofetil withdrawal is desirable once disease quiescence is reached, but the timing of when to do so and whether it provides a benefit has not been well-studied. We aimed to determine the effects of mycophenolate mofetil withdrawal on the risk of clinically significant disease reactivation in patients with quiescent SLE on long-term mycophenolate mofetil therapy. METHODS: This multicenter, open-label, randomised trial was conducted in 19 centres in the USA. Eligible patients were aged between 18 and 70 years old, met the American College of Rheumatology (ACR) 1997 SLE criteria, and had a clinical SLEDAI score of less than 4 at screening. Mycophenolate mofetil therapy was required to be stable or decreasing for 2 years or more if initiated for renal indications, or for 1 year or more for non-renal indications. Participants were randomly allocated in a 1:1 ratio to a withdrawal group, who tapered off mycophenolate mofetil over 12 weeks, or a maintenance group who maintained their baseline dose (1-3g per day) for 60 weeks. Adaptive random allocation ensured groups were balanced for study site, renal versus non-renal disease, and baseline mycophenolate mofetil dose (≥2 g per day vs <2 g per day). Clinically significant disease reactivation by week 60 following random allocation, requiring increased doses or new immunosuppressive therapy was the primary endpoint, in the modified intention-to-treat population (all randomly allocated participants who began study-provided mycophenolate mofetil). Non-inferiority was evaluated using an estimation-based approach. The trial was registered at ClinicalTrials.gov (NCT01946880) and is completed. FINDINGS: Between Nov 6, 2013, and April 27, 2018, 123 participants were screened, of whom 102 were randomly allocated to the maintenance group (n=50) or the withdrawal group (n=52). Of the 100 participants included in the modified intention-to-treat analysis (49 maintenance, 51 withdrawal), 84 (84%) were women, 16 (16%) were men, 40 (40%) were White, 41 (41%) were Black, and 76 (76%) had a history of lupus nephritis. The average age was 42 (SD 12·7). By week 60, nine (18%) of 51 participants in the withdrawal group had clinically significant disease reactivation, compared to five (10%) of 49 participants in the maintenance group. The risk of clinically significant disease reactivation was 11% (95% CI 5-24) in the maintenance group and 18% (10-32) in the withdrawal group. The estimated increase in the risk of clinically significant disease reactivation with mycophenolate mofetil withdrawal was 7% (one-sided upper 85% confidence limit 15%). Similar rates of adverse events were observed in the maintenance group (45 [90%] of 50 participants) and the withdrawal group (46 [88%] of 52 participants). Infections were more frequent in the mycophenolate mofetil maintenance group (32 [64%]) compared with the withdrawal group (24 [46%]). INTERPRETATIONS: Mycophenolate mofetil withdrawal is not significantly inferior to mycophenolate mofetil maintenance. Estimates for the rates of disease reactivation and increases in risk with withdrawal can assist clinicians in making informed decisions on withdrawing mycophenolate mofetil in patients with stable SLE. FUNDING: The National Institute of Allergy and Infectious Diseases and the National Institute of Arthritis and Musculoskeletal and Skin Diseases.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Masculino , Humanos , Feminino , Adulto , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Idoso , Ácido Micofenólico/efeitos adversos , Nefrite Lúpica/tratamento farmacológico , Resultado do Tratamento , Imunossupressores/efeitos adversos , Lúpus Eritematoso Sistêmico/tratamento farmacológico
8.
Prog Orthod ; 25(1): 1, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38168740

RESUMO

OBJECTIVE: To compare the effects on facial soft tissues produced by maxillary expansion generated by rapid maxillary expansion (RME) versus slow maxillary expansion (SME). MATERIALS AND METHODS: Patients in the mixed dentition were included with a transverse discrepancy between the two arches of at least 3 mm. A conventional RME screw was compared to a new expansion screw (Leaf expander) designed to produce SME. Both screws were incorporated in a fixed expander. The primary outcome was the difference of the facial tissue changes in the nasal area measured on facial 3D images captured immediately before application of the expander (T0) and after one year of retention, immediately after the expander removal (T1). Secondary outcomes were soft tissue changes of other facial regions (mouth, lips, and chin). Analysis of covariance was used for statistical analysis. RESULTS: Fourteen patients were allocated to the RME group, and 14 patients were allocated to the SME group. There were no dropouts. Nasal width change showed a difference between the two groups (1.3 mm greater in the RME group, 95% CI from 0.4 to 2.2, P = 0.005). Also, intercanthal width showed a difference between treatments (0.7 mm greater in the RME group, 95% CI from 0.0 to 1.3, P = 0.044). Nasal columella width, mouth width, nasal tip angle, upper lip angle, and lower lip angle did not show any statistically significant differences. The Y-axis (anterior-posterior) components of the nasal landmark showed a statistically significant difference between the two groups (0.5 mm of forward displacement greater in the RME group, 95% CI from 0.0 to 1.2, P = 0.040). Also, Z-axis (superior-inferior) components of the lower lip landmark was statistically significant (0.9 mm of downward displacement in favor of the RME group, 95% CI from 0.1 to 1.7, P = 0.027). All the other comparisons of the three-dimensional assessments were not statistically significant. CONCLUSIONS: RME produced significant facial soft tissue changes when compared to SME. RME induced greater increases in both nasal and intercanthal widths (1.3 mm and 0.7 mm, respectively). These findings, though statistically significant, probably are not clinically relevant. Trial registration ISRCTN, ISRCTN18263886. Registered 8 November 2016, https://www.isrctn.com/ISRCTN18263886?q=Franchi&filters=&sort=&offset=2&totalResults=2&page=1&pageSize=10.


Assuntos
Face , Técnica de Expansão Palatina , Humanos , Face/diagnóstico por imagem , Lábio , Fotogrametria , Dentição Mista , Maxila
9.
Orthod Craniofac Res ; 27(3): 429-438, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38146808

RESUMO

OBJECTIVE: To assess the short- and long-term dentoskeletal effects of early Class III treatment with rapid maxillary expansion and facemask (RME/FM) followed by fixed appliances. MATERIALS AND METHODS: A total of 44 patients (27 females, 17 males) treated consecutively with RME/FM were included from the archives of 3 centres. Three lateral cephalograms were available: T0 (before the start of RME/FM therapy, mean age 8.1 ± 1.8 years), T1 (immediately after RME/FM, mean age 9.8 ± 1.6 years), and T2 (long-term observation, mean age 19.5 ± 1.6 years). A control group of 17 untreated Class III subjects (12 females and 5 males) also was selected. Between-group statistical comparisons were performed with ANCOVA. RESULTS: No statistically significant differences for any of the cephalometric variables were found at T0. In the short term, the treated group showed significant improvements in ANB (+2.9°), Wits appraisal (+2.7 mm), SNA (+1.8°) and SNB (-1.1°). A significant closure of CoGoMe angle (-1.3°) associated with smaller increments along Co-Gn (-2.4 mm) also was found together with a significant increase in intermaxillary divergence (+1.3°). In the long-term, significant improvements in ANB (+2.6°), Wits appraisal (+2.7 mm) and SNB (-1.7°) were recorded together with a significant closure of the CoGoMe angle (-2.9°). No significant long-term changes in vertical skeletal relationships were found. CONCLUSIONS: RME/FM therapy was effective in improving Class III dentoskeletal relationships in the short term. These changes remained stable in the long-term due mainly to favourable mandibular changes.


Assuntos
Cefalometria , Aparelhos de Tração Extrabucal , Má Oclusão Classe III de Angle , Aparelhos Ortodônticos Fixos , Técnica de Expansão Palatina , Criança , Feminino , Humanos , Masculino , Adulto Jovem , Má Oclusão Classe III de Angle/terapia , Mandíbula , Maxila , Técnica de Expansão Palatina/instrumentação , Estudos Prospectivos , Estudos Retrospectivos , Resultado do Tratamento
10.
bioRxiv ; 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38076858

RESUMO

Skeletal muscle is the largest organ in the body, responsible for gross movement and metabolic regulation. Recently, variants in the MYBPC1 gene have been implicated in a variety of developmental muscle diseases, such as distal arthrogryposis. How MYBPC1 variants cause disease is not well understood. Here, through a collection of novel gene-edited mouse models, we define a critical role for slow myosin binding protein-C (sMyBP-C), encoded by MYBPC1, across muscle development, growth, and maintenance during prenatal, perinatal, postnatal and adult stages. Specifically, Mybpc1 knockout mice exhibited early postnatal lethality and impaired skeletal muscle formation and structure, skeletal deformity, and respiratory failure. Moreover, a conditional knockout of Mybpc1 in perinatal, postnatal and adult stages demonstrates impaired postnatal muscle growth and function secondary to disrupted actomyosin interaction and sarcomere structural integrity. These findings confirm the essential role of sMyBP-C in skeletal muscle and reveal specific functions in both prenatal embryonic musculoskeletal development and postnatal muscle growth and function.

11.
JCI Insight ; 8(22)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37991020

RESUMO

Histone deacetylase (HDAC) inhibitors have garnered considerable interest for the treatment of adult and pediatric malignant brain tumors. However, owing to their broad-spectrum nature and inability to effectively penetrate the blood-brain barrier, HDAC inhibitors have failed to provide substantial clinical benefit to patients with glioblastoma (GBM) to date. Moreover, global inhibition of HDACs results in widespread toxicity, highlighting the need for selective isoform targeting. Although no isoform-specific HDAC inhibitors are currently available, the second-generation hydroxamic acid-based HDAC inhibitor quisinostat possesses subnanomolar specificity for class I HDAC isoforms, particularly HDAC1 and HDAC2. It has been shown that HDAC1 is the essential HDAC in GBM. This study analyzed the neuropharmacokinetic, pharmacodynamic, and radiation-sensitizing properties of quisinostat in preclinical models of GBM. It was found that quisinostat is a well-tolerated and brain-penetrant molecule that extended survival when administered in combination with radiation in vivo. The pharmacokinetic-pharmacodynamic-efficacy relationship was established by correlating free drug concentrations and evidence of target modulation in the brain with survival benefit. Together, these data provide a strong rationale for clinical development of quisinostat as a radiosensitizer for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Criança , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/uso terapêutico , Histona Desacetilases/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Isoformas de Proteínas/metabolismo , Encéfalo/metabolismo
12.
J Clin Med ; 12(21)2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37959395

RESUMO

BACKGROUND: to determine the role of treatment timing in the long-term effects produced by rapid maxillary expansion and facemask therapy (RME/FM) in Class III patients. METHODS: This study compared two sample groups treated with RME/FM followed by fixed appliances: the early prepubertal group (EPG) (17 patients; mean age before treatment (T0), 5.8 ± 0.7 years; range, 4.3-6.9 years) and the late prepubertal group (LPG) (17 patients; mean age at T0, 10.1 ± 0.8 years; range, 9.0-11.1 years). Lateral cephalograms for the two groups were examined before treatment (T0) and at a long-term observation (T1) (EPG, 19.8 ± 1.0 years; LPG, 21.0 ± 2.1 years). Independent sample t-tests were performed to compare the two groups at T0 and T1. RESULTS: No statistically significant differences were found for any of the cephalometric variables at T0, except for the total mandibular length, overjet, and inclination of the maxillary incisors to the palatal plane, which were greater in the LPG. At T1, no statistically significant differences were detected for any of the cephalometric variables. CONCLUSIONS: There were no significant long-term differences when treating Class III patients with RME/FM, either during an early prepubertal phase (≤7 years of age) or during a late prepubertal phase (≥9 years of age).

13.
Artigo em Inglês | MEDLINE | ID: mdl-37962617

RESUMO

PURPOSE: Staphylococcus aureus is the most common and impactful multi-drug resistant pathogen implicated in (periprosthetic) joint infections (PJI) and fracture-related infections (FRI). Therefore, the present proof-of-principle study was aimed at the rapid detection of S. aureus in synovial fluids and biofilms on extracted osteosynthesis materials through bacteria-targeted fluorescence imaging with the 'smart-activatable' DNA-based AttoPolyT probe. This fluorogenic oligonucleotide probe yields large fluorescence increases upon cleavage by micrococcal nuclease, an enzyme secreted by S. aureus. METHODS: Synovial fluids from patients with suspected PJI and extracted osteosynthesis materials from trauma patients with suspected FRI were inspected for S. aureus nuclease activity with the AttoPolyT probe. Biofilms on osteosynthesis materials were imaged with the AttoPolyT probe and a vancomycin-IRDye800CW conjugate (vanco-800CW) specific for Gram-positive bacteria. RESULTS: 38 synovial fluid samples were collected and analyzed. Significantly higher fluorescence levels were measured for S. aureus-positive samples compared to, respectively, other Gram-positive bacterial pathogens (p < 0.0001), Gram-negative bacterial pathogens (p = 0.0038) and non-infected samples (p = 0.0030), allowing a diagnosis of S. aureus-associated PJI within 2 h. Importantly, S. aureus-associated biofilms on extracted osteosynthesis materials from patients with FRI were accurately imaged with the AttoPolyT probe, allowing their correct distinction from biofilms formed by other Gram-positive bacteria detected with vanco-800CW within 15 min. CONCLUSION: The present study highlights the potential clinical value of the AttoPolyT probe for fast and accurate detection of S. aureus infection in synovial fluids and biofilms on extracted osteosynthesis materials.

14.
Epilepsy Res ; 198: 107256, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38000152

RESUMO

The lack of preventive and disease modifying therapies for temporal lobe epilepsy (TLE) is a major unmet medical need. Search for such therapies utilize mouse models and require detection of seizures in electroencephalography (EEG) recordings. The labor-intensive nature of reviewing EEGs spanning many weeks underscores the need for a method of automated detection. Here we report a simple automated method of detecting seizures in long term EEG recordings from electrodes implanted in the hippocampus in animal models of TLE. We utilize a 2-pronged approach that relies on the increase in power within the gamma band range (20-50hz) during the seizure followed by suppression of activity following the seizure (post-ictal suppression [PIS]). We demonstrate the utility of this method for detecting seizures in hippocampal and amygdala EEG recordings from multiple models of TLE.


Assuntos
Epilepsia do Lobo Temporal , Animais , Camundongos , Epilepsia do Lobo Temporal/diagnóstico , Convulsões/diagnóstico , Eletroencefalografia/métodos , Hipocampo , Tonsila do Cerebelo , Modelos Animais de Doenças
16.
AJO DO Clin Companion ; 3(2): 93-109, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37636594

RESUMO

Treatment effects occurring during Class II malocclusion treatment with the clear aligner mandibular advancement protocol were evaluated in two growing patients: one male (12 years, 3 months) and one female (11 years, 9 months). Both patients presented with full cusp Class II molar and canine relationships. Intraoral scans and cone-beam computed tomography were acquired before treatment and after mandibular advancement. Three-dimensional skeletal and dental long-axis changes were quantified, in which the dental long axis was determined by registering the dental crowns obtained from intraoral scans to the root canals in cone-beam computed tomography scans obtained at the same time points. Class II correction was achieved by a combination of mandibular skeletal and dental changes. A similar direction of skeletal and dental changes was observed in both patients, with downward and forward displacement of the mandible resulting from the growth of the mandibular condyle and ramus. Dental changes in both patients included mesialization of the mandibular posterior teeth with flaring of mandibular anterior teeth. In these two patients, clear aligner mandibular advancement was an effective treatment modality for Class II malocclusion correction with skeletal and dental effects and facial profile improvement.

17.
Am J Orthod Dentofacial Orthop ; 164(6): 824-836, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37598337

RESUMO

INTRODUCTION: This study aimed to quantify the outcomes of adolescent patients with Class II malocclusion treated with the Carriere Motion 3D Appliance (CMA) combined with full fixed appliances. METHODS: Cone-beam computed tomography scans of 22 patients were available before orthodontic treatment (T1), at removal of the CMA (T2), and posttreatment (T3). The average age of the patients was 13.5 ± 1.6 years at T1, 14.1 ± 0.2 years at T2, and 15.6 ± 0.5 years at T3. The 3-dimensional image analysis procedures were performed using ITK-SNAP (version 3.6.0; www.itksnap.org, Hatfield, Pa) and SlicerCMF (version 4.11.0; http://www.slicer.org, Cambridge, Mass); skeletal and dentoalveolar changes relative to cranial base, maxillary, and mandibular regional superimpositions were evaluated. RESULTS: Changes were analyzed with 1 sample t tests using the mean differences during the CMA phase (T1 to T2) and total treatment time (T1 to T3). Significant skeletal changes included a slight reduction of ANB from T1 to T3, mandibular growth (Co-Gn increment of 1.2 mm and 3.3 mm from T1 to T2 and T1 to T3, respectively), inferior displacement of point A, and anterior and inferior displacement of point B. The mandibular plane did not change significantly during treatment. During the CMA treatment, posterior tipping and distal rotation of the maxillary molars, tip back and inferior displacement of the maxillary canines, significant mesial rotation, and superior displacement of the mandibular molars were observed. These movements rebounded during the full fixed appliance phase except for the molar and canine vertical displacements. Clinically significant dental changes during treatment included a reduction in overjet and overbite, Class II correction of the molar and canine relationship, and proclination of the mandibular incisors. CONCLUSIONS: The CMA is an effective treatment modality for Class II correction in growing patients because of a combination of mesial movement of the mandibular molar, distal rotation of the maxillary molar, and anterior displacement of the mandible.


Assuntos
Má Oclusão Classe II de Angle , Aparelhos Ortodônticos Funcionais , Sobremordida , Adolescente , Humanos , Criança , Cefalometria/métodos , Má Oclusão Classe II de Angle/diagnóstico por imagem , Má Oclusão Classe II de Angle/terapia , Sobremordida/terapia , Mandíbula/diagnóstico por imagem , Maxila , Desenho de Aparelho Ortodôntico
18.
medRxiv ; 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37502867

RESUMO

Teplizumab has been approved for the delay of the onset of type 1 diabetes and may modulate new onset disease. We found that patients who were EBV positive at baseline had a more robust response to drug in two clinical trials and therefore postulated that latent virus has general effects in modifying immune responses. We compared the phenotypes, transcriptomes, and development of peripheral blood cells before and after teplizumab treatment. Higher number of Tregs and partially exhausted CD8 + T cells were found in EBV seropositive individuals at the baseline in the TN10 trial and AbATE trial. Single cell transcriptomics and functional assays identified downregulation of the T cell receptor and other signaling pathways before treatment. Impairments in function of adaptive immune cells were enhanced by teplizumab treatment in EBV seropositive individuals. Our data indicate that EBV can impair signaling pathways generally in immune cells, that broadly redirect cell differentiation.

19.
Orthod Craniofac Res ; 26(2): 151-162, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35737876

RESUMO

OBJECTIVE: To compare the transverse dental and skeletal changes in patients treated with bone-anchored palatal expander (bone-borne, BB) compared to patients treated with tooth and bone-anchored palatal expanders (tooth-bone-borne, TBB) using cone-beam computer tomography (CBCT) and 3D image analysis. METHODS: The sample comprised 30 patients with transverse maxillary discrepancy treated with two different types of appliances: bone-borne (Group BB) and tooth-bone-borne (Group TBB) expanders. CBCT scans were acquired before (T1) and after completion of maxillary expansion (T2); the interval was 5.4 ± 3.4 and 6.2 ± 2.1 months between the T1 and the T2 scans of Group TBB (tooth-bone-borne) and Group BB (bone-borne), respectively. Transverse, anteroposterior and vertical linear and angular three-dimensional dentoskeletal changes were assessed after cranial base superimposition. RESULTS: Both groups displayed marked transverse skeletal expansion with a greater ratio of skeletal to dental changes. Greater changes at the nasal cavity, zygoma and orbital levels were found in Group BB. A relatively parallel sutural opening in an anterior-posterior direction was observed in Group TBB; however, the Group BB presented a somewhat triangular (V-shaped) opening of the suture that was wider anteriorly. Small downward-forward displacements were observed in both groups. Asymmetric expansion occurred in approximately 50% of the patients in both groups. CONCLUSION: Greater skeletal vs dental expansion ratio and expansion of the circummaxillary regions were found in Group BB, the group in which a bone-borne expander was used. Both groups presented skeletal and dental changes, with a similar amount of posterior palate expansion. Asymmetric expansion was observed in both groups.


Assuntos
Técnica de Expansão Palatina , Dente , Humanos , Adulto Jovem , Tomografia Computadorizada de Feixe Cônico/métodos , Maxila/diagnóstico por imagem , Palato
20.
Cell Metab ; 34(10): 1561-1577.e9, 2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-35882232

RESUMO

Exercise induces signaling networks to improve muscle function and confer health benefits. To identify divergent and common signaling networks during and after different exercise modalities, we performed a phosphoproteomic analysis of human skeletal muscle from a cross-over intervention of endurance, sprint, and resistance exercise. This identified 5,486 phosphosites regulated during or after at least one type of exercise modality and only 420 core phosphosites common to all exercise. One of these core phosphosites was S67 on the uncharacterized protein C18ORF25, which we validated as an AMPK substrate. Mice lacking C18ORF25 have reduced skeletal muscle fiber size, exercise capacity, and muscle contractile function, and this was associated with reduced phosphorylation of contractile and Ca2+ handling proteins. Expression of C18ORF25 S66/67D phospho-mimetic reversed the decreased muscle force production. This work defines the divergent and canonical exercise phosphoproteome across different modalities and identifies C18ORF25 as a regulator of exercise signaling and muscle function.


Assuntos
Proteínas Quinases Ativadas por AMP , Proteínas Adaptadoras de Transdução de Sinal , Exercício Físico , Músculo Esquelético , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Humanos , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Fosforilação , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...