Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 103, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643194

RESUMEN

BACKGROUND: Colony stimulating factor 1 receptor (CSF1R) signaling is crucial for the maintenance and function of various myeloid subsets. CSF1R antagonism was previously shown to mitigate clinical severity in experimental autoimmune encephalomyelitis (EAE). The associated mechanisms are still not well delineated. METHODS: To assess the effect of CSF1R signaling, we employed the CSF1R antagonist PLX5622 formulated in chow (PLX5622 diet, PD) and its control chow (control diet, CD). We examined the effect of PD in steady state and EAE by analyzing cells isolated from peripheral immune organs and from the CNS via flow cytometry. We determined CNS infiltration sites and assessed the extent of demyelination using immunohistochemistry of cerebella and spinal cords. Transcripts of genes associated with neuroinflammation were also analyzed in these tissues. RESULTS: In addition to microglial depletion, PD treatment reduced dendritic cells and macrophages in peripheral immune organs, both during steady state and during EAE. Furthermore, CSF1R antagonism modulated numbers and relative frequencies of T effector cells both in the periphery and in the CNS during the early stages of the disease. Classical neurological symptoms were milder in PD compared to CD mice. Interestingly, a subset of PD mice developed atypical EAE symptoms. Unlike previous studies, we observed that the CNS of PD mice was infiltrated by increased numbers of peripheral immune cells compared to that of CD mice. Immunohistochemical analysis showed that CNS infiltrates in PD mice were mainly localized in the cerebellum while in CD mice infiltrates were primarily localized in the spinal cords during the onset of neurological deficits. Accordingly, during the same timepoint, cerebella of PD but not of CD mice had extensive demyelinating lesions, while spinal cords of CD but not of PD mice were heavily demyelinated. CONCLUSIONS: Our findings suggest that CSF1R activity modulates the cellular composition of immune cells both in the periphery and within the CNS, and affects lesion localization during the early EAE stages.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Ratones , Animales , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Compuestos Orgánicos/farmacología , Médula Espinal/patología , Microglía , Receptores del Factor Estimulante de Colonias , Proteínas Tirosina Quinasas Receptoras , Ratones Endogámicos C57BL
2.
Clin Exp Allergy ; 54(3): 169-184, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38423799

RESUMEN

Randomised controlled trials investigating the efficacy of oral tolerance induction to peanut have enabled detailed comparison of their clinical and immunological success. They have demonstrated that the regular consumption of peanut for at least 2 years by babies who are not allergic enables protection from developing peanut allergy. The LEAP study intervention tested the impact of regular peanut consumption for 4 years and demonstrated a sustained protection against the development of peanut allergy even after 12 months of peanut avoidance from 5 to 6 years of age. The PreventADALL trial introduced multiple allergens into babies' diets from early infancy and reduced the prevalence of food allergy at 3 years, especially by protecting against peanut allergy. Immunological studies from the LEAP cohort demonstrated that regular peanut consumption was associated with a prompt induction of peanut-specific IgG4 and reduced manufacture of peanut and Ara h 2-specific IgE. Even after stopping peanut consumption for 5 years, there continued to be a significant fall in peanut-specific Ara h 2 IgE in the consumption group from 5 to 6 years of age (p < .01). Children who developed peanut allergy by 5 years started to develop increasing sensitisation to linear sequential peanut epitopes from 2.5 years of age, suggesting that putative disease-modifying interventions should commence before 3 years. Data comparing clinical outcomes between children undergoing peanut immunotherapy from infancy suggest that younger children can consume higher portions of peanut without reaction on challenge whilst taking immunotherapy, have fewer side effects and are more likely to enjoy remission of PA. Peanut oral immunotherapy modulates T-cell populations in order to bring about hypo-responsiveness of allergy effector cells. Studies are now needed to characterise and compare different states of immunological tolerance. This will accelerate the design of interventions which can promote primary, secondary and tertiary levels of PA prevention across a range of age groups.


Asunto(s)
Hipersensibilidad a los Alimentos , Hipersensibilidad al Cacahuete , Niño , Lactante , Humanos , Preescolar , Hipersensibilidad al Cacahuete/prevención & control , Inmunoglobulina E , Epítopos , Arachis , Alérgenos , Antígenos de Plantas
3.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34620713

RESUMEN

In the skin, tissue injury results in fibrosis in the form of scars composed of dense extracellular matrix deposited by fibroblasts. The therapeutic goal of regenerative wound healing has remained elusive, in part because principles of fibroblast programming and adaptive response to injury remain incompletely understood. Here, we present a multimodal -omics platform for the comprehensive study of cell populations in complex tissue, which has allowed us to characterize the cells involved in wound healing across both time and space. We employ a stented wound model that recapitulates human tissue repair kinetics and multiple Rainbow transgenic lines to precisely track fibroblast fate during the physiologic response to skin injury. Through integrated analysis of single cell chromatin landscapes and gene expression states, coupled with spatial transcriptomic profiling, we are able to impute fibroblast epigenomes with temporospatial resolution. This has allowed us to reveal potential mechanisms controlling fibroblast fate during migration, proliferation, and differentiation following skin injury, and thereby reexamine the canonical phases of wound healing. These findings have broad implications for the study of tissue repair in complex organ systems.


Asunto(s)
Cicatriz/patología , Fibroblastos/metabolismo , Fibrosis/patología , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Matriz Extracelular/metabolismo , Femenino , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Piel/metabolismo
4.
J Autoimmun ; 140: 103088, 2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37549449

RESUMEN

Mechanistic studies of autoimmune disorders have identified circulating T follicular helper (cTfh) cells as drivers of autoimmunity. However, the quantification of cTfh cells is not yet used in clinical practice due to the lack of age-stratified normal ranges and the unknown sensitivity and specificity of this test for autoimmunity. We enrolled 238 healthy participants and 130 patients with common and rare disorders of autoimmunity or autoinflammation. Patients with infections, active malignancy, or any history of transplantation were excluded. In 238 healthy controls, median cTfh percentages (range 4.8%-6.2%) were comparable among age groups, sexes, races, and ethnicities, apart from a significantly lower percentages in children less than 1 year of age (median 2.1%, CI: 0.4%-6.8, p < 0.0001). Among 130 patients with over 40 immune regulatory disorders, a cTfh percentage exceeding 12% had 88% sensitivity and 94% specificity for differentiating disorders with adaptive immune cell dysregulation from those with predominantly innate cell defects. This threshold had a sensitivity of 86% and specificity of 100% for active autoimmunity and normalized with effective treatment. cTfh percentages exceeding 12% distinguish autoimmunity from autoinflammation, thereby differentiating two endotypes of immune dysregulation with overlapping symptoms and different therapies.

5.
J Allergy Clin Immunol ; 149(2): 569-578, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34958811

RESUMEN

Our understanding of risk factors and interventions influencing outcomes from coronavirus disease 2019 (COVID-19) has continued to evolve, revealing advances emerging from hypotheses formed at the start of the pandemic. Epidemiologic studies have shown that asthma control, rather than a diagnosis of asthma, is a determinant of COVID-19 severity. Clinical outcomes in patients with primary immunodeficiencies, even in those with impaired cellular immunity, are variable. IL-6 has emerged as a reliable biomarker of COVID-19 severity, and large clinical trials have shown the potential for improving outcomes through inhibition of IL-6 signaling in some patients. Studies of genetic risk factors for severe COVID-19 have also revealed the importance of interferon homeostasis in the defense against severe acute respiratory syndrome coronavirus 2. Because COVID-19 vaccines constitute the primary tool for ending this pandemic, strategies have been developed to address potential allergic and immune-mediated reactions. Here, we discuss advances in our understanding of COVID-19 risk factors and outcomes within the context of allergic and immunologic mechanisms.


Asunto(s)
Antivirales/uso terapéutico , Asma/terapia , Productos Biológicos/uso terapéutico , COVID-19/terapia , Síndromes de Inmunodeficiencia/terapia , SARS-CoV-2/efectos de los fármacos , Anticuerpos Monoclonales/uso terapéutico , Asma/inmunología , Asma/mortalidad , Asma/virología , Azetidinas/uso terapéutico , Biomarcadores/metabolismo , COVID-19/inmunología , COVID-19/mortalidad , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/mortalidad , Síndromes de Inmunodeficiencia/virología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/inmunología , Pronóstico , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Factores de Riesgo , SARS-CoV-2/patogenicidad , Sulfonamidas/uso terapéutico , Análisis de Supervivencia , Resultado del Tratamiento
6.
Proc Natl Acad Sci U S A ; 116(17): 8155-8160, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30971489

RESUMEN

Protein synthesis is central to maintaining cellular homeostasis and its study is critical to understanding the function and dysfunction of eukaryotic systems. Here we report L-2-tellurienylalanine (TePhe) as a noncanonical amino acid for direct measurement of protein synthesis. TePhe is synthetically accessible, nontoxic, stable under biological conditions, and the tellurium atom allows its direct detection with mass cytometry, without postexperiment labeling. TePhe labeling is competitive with phenylalanine but not other large and aromatic amino acids, demonstrating its molecular specificity as a phenylalanine mimic; labeling is also abrogated in vitro and in vivo by the protein synthesis inhibitor cycloheximide, validating TePhe as a translation reporter. In vivo, imaging mass cytometry with TePhe visualizes translation dynamics in the mouse gut, brain, and tumor. The strong performance of TePhe as a probe for protein synthesis, coupled with the operational simplicity of its use, suggests TePhe could become a broadly applied molecule for measuring translation in vitro and in vivo.


Asunto(s)
Citometría de Flujo/métodos , Citometría de Imagen/métodos , Fenilalanina/química , Biosíntesis de Proteínas/fisiología , Telurio/química , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Cicloheximida/farmacología , Células HCT116 , Humanos , Yeyuno/diagnóstico por imagen , Yeyuno/metabolismo , Células Jurkat , Ratones , Neoplasias Experimentales , Fenilalanina/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Telurio/metabolismo
7.
J Allergy Clin Immunol ; 148(3): 732-738.e1, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224783

RESUMEN

BACKGROUND: Multisystem inflammatory syndrome in children (MIS-C) is a pediatric complication of severe acute respiratory syndrome coronavirus 2 infection that is characterized by multiorgan inflammation and frequently by cardiovascular dysfunction. It occurs predominantly in otherwise healthy children. We previously reported haploinsufficiency of suppressor of cytokine signaling 1 (SOCS1), a negative regulator of type I and II interferons, as a genetic risk factor for MIS-C. OBJECTIVES: We aimed to identify additional genetic mechanisms underlying susceptibility to severe acute respiratory syndrome coronavirus 2-associated MIS-C. METHODS: In a single-center, prospective cohort study, whole exome sequencing was performed on patients with MIS-C. The impact of candidate variants was tested by using patients' PBMCs obtained at least 7 months after recovery. RESULTS: We enrolled 18 patients with MIS-C (median age = 8 years; interquartile range = 5-12.25 years), of whom 89% had no conditions other than obesity. In 2 boys with no significant infection history, we identified and validated hemizygous deleterious defects in XIAP, encoding X-linked inhibitor of apoptosis, and CYBB, encoding cytochrome b-245, beta subunit. Including the previously reported SOCS1 haploinsufficiency, a genetic diagnosis was identified in 3 of 18 patients (17%). In contrast to patients with mild COVID-19, patients with defects in SOCS1, XIAP, or CYBB exhibit an inflammatory immune cell transcriptome with enrichment of differentially expressed genes in pathways downstream of IL-18, oncostatin M, and nuclear factor κB, even after recovery. CONCLUSIONS: Although inflammatory disorders are rare in the general population, our cohort of patients with MIS-C was enriched for monogenic susceptibility to inflammation. Our results support the use of next-generation sequencing in previously healthy children who develop MIS-C.


Asunto(s)
COVID-19/etiología , COVID-19/metabolismo , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Biomarcadores , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/virología , Niño , Preescolar , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico
8.
Ann Plast Surg ; 87(5): 542-546, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34699433

RESUMEN

BACKGROUND: Reinforcement of the abdominal wall with synthetic mesh in autologous breast reconstruction using abdominal free tissue transfer decreases the risk of bulging and herniation. However, the impact of the plane of mesh placement on donor site complications has not yet been investigated. METHODS: We performed a retrospective analysis of 312 patients who had undergone autologous breast reconstruction with muscle-sparing transverse rectus abdominis myocutaneous (MS-TRAM) flaps or deep inferior epigastric perforator (DIEP) flaps as well as polypropylene mesh implantation at the donor site. Donor site complications were compared among patients with different flap types and different mesh positions including overlay (n = 90), inlay and overlay (I-O; n = 134), and sublay (n = 88). RESULTS: Abdominal hernias occurred in 2.86% of patients who had undergone MS-TRAM reconstructions and in 2.63% of patients who had undergone DIEP reconstructions. When comparing patients with different mesh positions, donor site complications occurred in 14.4% of patients with overlay mesh, 13.4% of patients with I-O mesh, and 10.2% of patients with sublay mesh (P = 0.68). Abdominal hernias occurred in 4.44% of patients with overlay mesh, 2.24% of patients with I-O mesh, and 2.27% of patients with sublay mesh (P = 0.69). Multivariable logistic regression analysis did not identify a significant association between mesh position and hernia rates as well as wound complications. CONCLUSIONS: Our data indicate that the plane of synthetic mesh placement in relation to the rectus abdominis muscle does not impact the rate of postoperative donor site complications in patients undergoing breast reconstruction with MS-TRAM or DIEP flaps.


Asunto(s)
Pared Abdominal , Mamoplastia , Colgajo Perforante , Arterias Epigástricas/cirugía , Humanos , Mamoplastia/efectos adversos , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Recto del Abdomen/trasplante , Estudios Retrospectivos , Mallas Quirúrgicas/efectos adversos
9.
Pediatr Phys Ther ; 33(1): E7-E9, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337780

RESUMEN

PURPOSE: The purpose of this report is to demonstrate the successful application of virtual reality to improve physical therapy in the pediatric cardiovascular intensive care unit. Early mobilization and cognitive stimulation improve morbidity of critically ill children. However, maintaining child engagement with these therapies can be challenging, especially during extended intensive care stays. SUMMARY OF KEY POINTS: While virtual reality has been successfully used as an analgesic and anxiolytic in the cardiovascular intensive care unit, this report demonstrates its novel use as a tool to augment physical therapy for a child who had been debilitated after heart transplantation. Virtual reality encouraged the child to engage in physical therapy sessions, participate for greater durations, and directly address barriers to discharge. CONCLUSIONS AND RECOMMENDATIONS FOR CLINICAL PRACTICE: While further studies are needed to define best practice, this report demonstrates that virtual reality can be safely used for carefully selected and monitored children in critical care.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/rehabilitación , Unidades de Cuidado Intensivo Pediátrico/organización & administración , Modalidades de Fisioterapia , Realidad Virtual , Adolescente , Ambulación Precoz/métodos , Femenino , Humanos
10.
Clin Immunol ; 216: 108459, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32418917

RESUMEN

The COVID-19 pandemic is one of the greatest infectious challenges in recent history. Presently, few treatment options exist and the availability of effective vaccines is at least one year away. There is an urgent need to find currently available, effective therapies in the treatment of patients with COVID-19 infection. In this review, we compare and contrast the use of intravenous immunoglobulin and hyperimmune globulin in the treatment of COVID-19 infection.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/patología , Inmunoglobulinas Intravenosas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Pandemias , Neumonía Viral/tratamiento farmacológico , Inmunidad Adaptativa/efectos de los fármacos , Enzima Convertidora de Angiotensina 2 , Acrecentamiento Dependiente de Anticuerpo/efectos de los fármacos , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/prevención & control , Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunización Pasiva/efectos adversos , Inmunización Pasiva/métodos , Inmunoglobulinas Intravenosas/efectos adversos , Factores Inmunológicos/efectos adversos , Terapia Molecular Dirigida , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/inmunología , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Neumonía Viral/virología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología , Sueroterapia para COVID-19
11.
Proc Natl Acad Sci U S A ; 114(41): E8685-E8694, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973887

RESUMEN

The molecular underpinnings of invasion, a hallmark of cancer, have been defined in terms of individual mediators but crucial interactions between these mediators remain undefined. In xenograft models and patient specimens, we identified a c-Met/ß1 integrin complex that formed during significant invasive oncologic processes: breast cancer metastases and glioblastoma invasive resistance to antiangiogenic VEGF neutralizing antibody, bevacizumab. Inducing c-Met/ß1 complex formation through an engineered inducible heterodimerization system promoted features crucial to overcoming stressors during metastases or antiangiogenic therapy: migration in the primary site, survival under hypoxia, and extravasation out of circulation. c-Met/ß1 complex formation was up-regulated by hypoxia, while VEGF binding VEGFR2 sequestered c-Met and ß1 integrin, preventing their binding. Complex formation promoted ligand-independent receptor activation, with integrin-linked kinase phosphorylating c-Met and crystallography revealing the c-Met/ß1 complex to maintain the high-affinity ß1 integrin conformation. Site-directed mutagenesis verified the necessity for c-Met/ß1 binding of amino acids predicted by crystallography to mediate their extracellular interaction. Far-Western blotting and sequential immunoprecipitation revealed that c-Met displaced α5 integrin from ß1 integrin, creating a complex with much greater affinity for fibronectin (FN) than α5ß1. Thus, tumor cells adapt to microenvironmental stressors induced by metastases or bevacizumab by coopting receptors, which normally promote both cell migration modes: chemotaxis, movement toward concentrations of environmental chemoattractants, and haptotaxis, movement controlled by the relative strengths of peripheral adhesions. Tumor cells then redirect these receptors away from their conventional binding partners, forming a powerful structural c-Met/ß1 complex whose ligand-independent cross-activation and robust affinity for FN drive invasive oncologic processes.


Asunto(s)
Neoplasias de la Mama/secundario , Resistencia a Antineoplásicos , Glioblastoma/secundario , Integrina beta1/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Bevacizumab/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Femenino , Fibronectinas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Integrina beta1/genética , Ratones , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Ann Plast Surg ; 84(5S Suppl 4): S318-S322, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32187065

RESUMEN

INTRODUCTION: Individualized postsurgical risk assessment models provide surgeons and patients with information that is vital to the surgical decision-making process. One such tool, the Breast Reconstruction Risk Assessment (BRA) score, uses a limited selection of patient-specific factors to predict 30-day postsurgical risk of surgical site infection, seroma, dehiscence, reoperation and explantation associated with immediate submuscular tissue expander breast reconstruction. This model's performance in prepectoral tissue expander reconstruction has not been previously reported. Here, we evaluate the performance of the BRA score model in a population of patients who underwent immediate prepectoral tissue expander breast reconstruction. MATERIALS AND METHODS: A retrospective chart review was conducted of prepectoral breast reconstructions performed in 2 institutions between January 2017 and December 2018. Complications occurring within 30 days postoperatively were documented and compared with the BRA score predicted risk for each patient. RESULTS: Overall 247 patients (average age, 49.2 years) were included in the study. The mean BRA score predicted 30-day risk of a complication was 13.0% (7.5-41.5%). The observed rate of 30-day postoperative complications was 31.2% (77 patients), though only 36 (14.6%) patients had complications included in the model. The remaining patients experienced skin necrosis or hematoma as their only early complication. The 30-day BRA score model demonstrated good fit for the overall occurrence of any of the BRA score predicted complications (Hosmer-Lemeshow 0.7167), though the model discrimination was poor (C statistic <0.60). Notably, half of the 30-day postsurgical complications observed in this study were due to skin necrosis, a complication not currently included in the 30-day BRA score model. CONCLUSIONS: Our results indicate that the current 30-day BRA score model may have poor predictive value in prepectoral breast reconstruction. The most common early complication observed, skin necrosis, is not currently included in the model, suggesting that caution should be applied when using this risk predictive calculator as an adjunct to patient evaluation and counseling.


Asunto(s)
Neoplasias de la Mama , Mamoplastia , Humanos , Mamoplastia/efectos adversos , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Dispositivos de Expansión Tisular
13.
Am J Pathol ; 188(3): 715-727, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29294300

RESUMEN

Fractures are common, with an incidence of 13.7 per 1000 adults annually. Systemic agents have been widely used for enhancing bone regeneration; however, the efficacy of these therapeutics for the management and prevention of fracture remains unclear. NEL-like protein 1 (NELL-1) is a potent pro-osteogenic cytokine that has been modified with polyethylene glycol (PEG)ylation [PEGylated NELL-1 (NELL-PEG)] to enhance its pharmacokinetics for systemic therapy. Our aim was to investigate the effects of systemic administration of NELL-PEG on fracture healing in mice and on overall bone properties in uninjured bones. Ten-week-old CD-1 mice were subjected to an open osteotomy of bilateral radii and treated with weekly injections of NELL-PEG or PEG phosphate-buffered saline as control. Systemic injection of NELL-PEG resulted in improved bone mineral density of the fracture site and accelerated callus union. After 4 weeks of treatment, mice treated with NELL-PEG exhibited substantially enhanced callus volume, callus mineralization, and biomechanical properties. NELL-PEG injection significantly augmented bone regeneration, as confirmed by high expression of bone turnover rate, bone formation rate, and mineral apposition rate. Consistently, the immunohistochemistry results also confirmed a high bone remodeling activity in the NELL-PEG-treated group. Our findings suggest that weekly injection of NELL-PEG may have the clinical potential to accelerate fracture union and enhance overall bone properties, which may help prevent subsequent fractures.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Proteínas de Unión al Calcio/uso terapéutico , Curación de Fractura/efectos de los fármacos , Fracturas Óseas/tratamiento farmacológico , Glicoproteínas/uso terapéutico , Radio (Anatomía)/lesiones , Animales , Proteínas de Unión al Calcio/farmacología , Femenino , Glicoproteínas/farmacología , Ratones , Modelos Animales , Osteotomía , Radio (Anatomía)/efectos de los fármacos , Resultado del Tratamiento
14.
Int J Mol Sci ; 20(8)2019 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-31013709

RESUMEN

The skin is a complex organ that has devised numerous strategies, such as physical, chemical, and microbiological barriers, to protect the host from external insults. In addition, the skin contains an intricate network of immune cells resident to the tissue, crucial for host defense as well as tissue homeostasis. In the event of an insult, the skin-resident immune cells are crucial not only for prevention of infection but also for tissue reconstruction. Deregulation of immune responses often leads to impaired healing and poor tissue restoration and function. In this review, we will discuss the defensive components of the skin and focus on the function of skin-resident immune cells in homeostasis and their role in wound healing.


Asunto(s)
Sistema Inmunológico , Fenómenos Fisiológicos de la Piel , Piel/inmunología , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Microbiota , Piel/citología , Piel/inervación , Piel/metabolismo , Cicatrización de Heridas
15.
Am J Pathol ; 187(7): 1485-1495, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28502475

RESUMEN

Multiple case reports using recombinant human bone morphogenetic protein-2 (rhBMP-2) have reported complications. However, the local adverse effects of rhBMP-2 application are not well documented. In this report we show that, in addition to promoting lumbar spinal fusion through potent osteogenic effects, rhBMP-2 augmentation promotes local cyst-like osteolytic formations in sheep trabecular bones that have undergone anterior lumbar interbody fusion. Three months after operation, conventional computed tomography showed that the trabecular bones of the rhBMP-2 application groups could fuse, whereas no fusion was observed in the control group. Micro-computed tomography analysis revealed that the core implant area's bone volume fraction and bone mineral density increased proportionately with rhBMP-2 dose. Multiple cyst-like bone voids were observed in peri-implant areas when using rhBMP-2 applications, and these sites showed significant bone mineral density decreases in relation to the unaffected regions. Biomechanically, these areas decreased in strength by 32% in comparison with noncystic areas. Histologically, rhBMP-2-affected void sites had an increased amount of fatty marrow, thinner trabecular bones, and significantly more adiponectin- and cathepsin K-positive cells. Despite promoting successful fusion, rhBMP-2 use in clinical applications may result in local adverse structural alterations and compromised biomechanical changes to the bone.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Vértebras Lumbares/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fusión Vertebral/métodos , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Densidad Ósea/efectos de los fármacos , Proteína Morfogenética Ósea 2/efectos adversos , Proteína Morfogenética Ósea 2/genética , Femenino , Humanos , Vértebras Lumbares/diagnóstico por imagen , Vértebras Lumbares/cirugía , Modelos Animales , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/genética , Ovinos , Fusión Vertebral/efectos adversos , Tomografía Computarizada por Rayos X , Factor de Crecimiento Transformador beta/efectos adversos , Factor de Crecimiento Transformador beta/genética
16.
Neurosurg Focus ; 44(6): E16, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29852776

RESUMEN

OBJECTIVE Glioblastoma (GBM) is an aggressive brain malignancy with a short overall patient survival, yet there remains significant heterogeneity in outcomes. Although access to health care has previously been linked to impact on prognosis in several malignancies, this question remains incompletely answered in GBM. METHODS This study was a retrospective analysis of 354 newly diagnosed patients with GBM who underwent first resection at the authors' institution (2007-2015). RESULTS Of the 354 patients (median age 61 years, and 37.6% were females), 32 (9.0%) had no insurance, whereas 322 (91.0%) had insurance, of whom 131 (40.7%) had Medicare, 45 (14%) had Medicaid, and 146 (45.3%) had private insurance. On average, insured patients survived almost 2-fold longer (p < 0.0001) than those who were uninsured, whereas differences between specific insurance types did not influence survival. The adjusted hazard ratio (HR) for death was higher in uninsured patients (HR 2.27 [95% CI 1.49-3.33], p = 0.0003). Age, mean household income, tumor size at diagnosis, and extent of resection did not differ between insured and uninsured patients, but there was a disparity in primary care physician (PCP) status-none of the uninsured patients had PCPs, whereas 72% of insured patients had PCPs. Postoperative adjuvant treatment rates with temozolomide (TMZ) and radiation therapy (XRT) were significantly less in uninsured (TMZ in 56.3%, XRT in 56.3%) than in insured (TMZ in 75.2%, XRT in 79.2%; p = 0.02 and p = 0.003) patients. Insured patients receiving both agents had better prognosis than uninsured patients receiving the same treatment (9.1 vs 16.34 months; p = 0.025), suggesting that the survival effect in insured patients could only partly be explained by higher treatment rates. Moreover, having a PCP increased survival among the insured cohort (10.7 vs 16.1 months, HR 1.65 [95% CI 1.27-2.15]; p = 0.0001), which could be explained by significant differences in tumor diameter at initial diagnosis between patients with and without PCPs (4.3 vs 4.8 cm, p = 0.003), and a higher rate of clinical trial enrollment, suggesting a critical role of PCPs for a timelier diagnosis of GBM and proactive cancer care management. CONCLUSIONS Access to health care is a strong determinant of prognosis in newly diagnosed patients with GBM. Any type of insurance coverage and having a PCP improved prognosis in this patient cohort. Higher rates of treatment with TMZ plus XRT, clinical trial enrollment, fewer comorbidities, and early diagnosis may explain survival disparities. Lack of health insurance or a PCP are major challenges within the health care system, which, if improved upon, could favorably impact the prognosis of patients with GBM.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Glioblastoma/diagnóstico , Accesibilidad a los Servicios de Salud/tendencias , Disparidades en Atención de Salud/tendencias , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/economía , Neoplasias Encefálicas/terapia , Femenino , Glioblastoma/economía , Glioblastoma/terapia , Accesibilidad a los Servicios de Salud/economía , Disparidades en Atención de Salud/economía , Humanos , Masculino , Pacientes no Asegurados , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Adulto Joven
17.
Clin Immunol ; 179: 66-76, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28279811

RESUMEN

Severe forms of chronic rhinosinusitis (CRS), a common upper airway inflammatory disorder, are associated with nasal polyps (NPs). NP disease is ameliorated by glucocorticoid (GC) treatment, whose cellular effects are poorly understood. We therefore assessed the influence of GC therapy on NPs in CRS patients, focusing on regulatory T (Treg) cells. Treg cell populations were analyzed by flow cytometry in NPs and control tissues from GC-treated CRS patients and controls. After GC exposure, selective expansion of Treg cells was seen within NPs, and not blood or adjacent ethmoid tissues. To confirm direct GC effects, NPs from the same patients were biopsied prior to, and following, 1week of oral GC exposure. Direct expansion of Tregs into the same NP bed was detected in 4/4 CRS patients following GC exposure. Treg cell spikes into NPs were secondary to cellular recruitment given limited Ki67 expression within these regulatory cells. Chemokine gene expression profiling identified several chemokines, notably CCL4, induced within NPs upon GC treatment. Neutralization of chemokine receptor/ligand interactions using CCR4 small molecule antagonists reduced Treg migration towards GC-treated NPs in an ex vivo migration assay. Our findings suggest that the common use of GCs in the treatment of NP disease leads to recruitment of Treg cells from peripheral sites into NP tissues, which may be critical to the anti-inflammatory effect of GCs. Mechanistically Treg expansion appears to be conferred, in part, by chemokine receptor/ligand interactions induced following corticosteroid therapy.


Asunto(s)
Glucocorticoides/farmacología , Pólipos Nasales/inmunología , Prednisona/farmacología , Rinitis/inmunología , Sinusitis/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Biopsia , Quimiocinas/genética , Enfermedad Crónica , Factores de Transcripción Forkhead/inmunología , Perfilación de la Expresión Génica , Glucocorticoides/uso terapéutico , Humanos , Pólipos Nasales/tratamiento farmacológico , Pólipos Nasales/genética , Pólipos Nasales/patología , Prednisona/uso terapéutico , Rinitis/tratamiento farmacológico , Rinitis/genética , Rinitis/patología , Sinusitis/tratamiento farmacológico , Sinusitis/genética , Sinusitis/patología , Linfocitos T Reguladores/inmunología
18.
Am J Pathol ; 186(2): 419-34, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26772960

RESUMEN

The differentiation factor NEL-like molecule-1 (NELL-1) has been reported as osteoinductive in multiple in vivo preclinical models. Bone morphogenetic protein (BMP)-2 is used clinically for skeletal repair, but in vivo administration can induce abnormal, adipose-filled, poor-quality bone. We demonstrate that NELL-1 combined with BMP2 significantly optimizes osteogenesis in a rodent femoral segmental defect model by minimizing the formation of BMP2-induced adipose-filled cystlike bone. In vitro studies using the mouse bone marrow stromal cell line M2-10B4 and human primary bone marrow stromal cells have confirmed that NELL-1 enhances BMP2-induced osteogenesis and inhibits BMP2-induced adipogenesis. Importantly, the ability of NELL-1 to direct BMP2-treated cells toward osteogenesis and away from adipogenesis requires intact canonical Wnt signaling. Overall, these studies establish the feasibility of combining NELL-1 with BMP2 to improve clinical bone regeneration and provide mechanistic insight into canonical Wnt pathway activity during NELL-1 and BMP2 osteogenesis. The novel abilities of NELL-1 to stimulate Wnt signaling and to repress adipogenesis may highlight new treatment approaches for bone loss in osteoporosis.


Asunto(s)
Adipogénesis , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea/fisiología , Proteínas del Tejido Nervioso/metabolismo , Osteogénesis/fisiología , Animales , Proteínas de Unión al Calcio , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas Endogámicas Lew , Transducción de Señal/fisiología
19.
Am J Orthod Dentofacial Orthop ; 150(2): 313-23, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27476365

RESUMEN

INTRODUCTION: The aims of this study were to evaluate the effects of rapid palatal expansion on the craniofacial skeleton of a patient with unilateral cleft lip and palate (UCLP) and to predict the points of force application for optimal expansion using a 3-dimensional finite element model. METHODS: A 3-dimensional finite element model of the craniofacial complex with UCLP was generated from spiral computed tomographic scans with imaging software (Mimics, version 13.1; Materialise, Leuven, Belgium). This model was imported into the finite element solver (version 12.0; ANSYS, Canonsburg, Pa) to evaluate transverse expansion forces from rapid palatal expansion. Finite element analysis was performed with transverse expansion to achieve 5 mm of anterolateral expansion of the collapsed minor segment to simulate correction of the anterior crossbite in a patient with UCLP. RESULTS: High-stress concentrations were observed at the body of the sphenoid, medial to the orbit, and at the inferior area of the zygomatic process of the maxilla. The craniofacial stress distribution was asymmetric, with higher stress levels on the cleft side. When forces were applied more anteriorly on the collapsed minor segment and more posteriorly on the major segment, there was greater expansion of the anterior region of the minor segment with minimal expansion of the major segment. CONCLUSIONS: The transverse expansion forces from rapid palatal expansion are distributed to the 3 maxillary buttresses. Finite element analysis is an appropriate tool to study and predict the points of force application for better controlled expansion in patients with UCLP.


Asunto(s)
Fisura del Paladar/terapia , Técnica de Expansión Palatina , Síndrome de Bandas Amnióticas/complicaciones , Fenómenos Biomecánicos , Niño , Fisura del Paladar/fisiopatología , Femenino , Análisis de Elementos Finitos , Humanos , Imagenología Tridimensional , Tomografía Computarizada Espiral
20.
Cell Mol Life Sci ; 71(8): 1353-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24158496

RESUMEN

Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.


Asunto(s)
Biomarcadores/metabolismo , Vasos Sanguíneos/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/clasificación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Pericitos/citología , Técnicas de Cultivo de Célula , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA