Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Emerg Nurs ; 48(6): 666-677, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36075769

RESUMEN

INTRODUCTION: The average length of stay of a fast-track area of a large urban hospital was excessively long, which affected the patient experience and the rate at which patients left without being seen. One approach to reducing average length of stay is to create nurse standard work. Nurse standard work was a defined set of process and procedures that reduce variability within a nurse's workflow. METHODS: Nurse standard work was created by a team of nurses assisted by management engineering using lean methodology and A3 problem solving. Data were gathered about average length of stay and left without being seen for patients in the emergency department fast-track area of an urban emergency department from October 2018 to June 2020. This period includes 5 months before the intervention start, 4 months during nurse standard work implementation, 9 months using nurse standard work before the unit was repurposed during COVID-19, and 3 months during COVID-19. RESULTS: Nurse standard work helped reduce average length of stay in the emergency department fast-track area from 205 minutes before project initiation to 150.4 minutes in the 7 months after implementing nurse standard work. The time spent walking for supplies was reduced from 422 and 272 seconds before nurse standard work to 25 and 30 seconds for the nurse technician and nurse, respectively, after nurse standard work. Left without being seen was decreased from 4.7% in October of 2018 to 0.7% by March of 2020. DISCUSSION: Nurse standard work reduced the amount of time that nurses spent performing support tasks and reduced delays in providing patient care, which then allowed more time for nurses to interact directly with patients. Nurse standard work provides a clear task sequence that eliminates delays in treating patients, but it also allows for fast identification of delays that do occur and simplifies problem solving to eliminate reoccurrence of delays. Therefore, nurse standard work is an essential component of efforts to reduce patient average length of stay in health care processes and reduce left without being seen to the national standard of less than 2%.


Asunto(s)
COVID-19 , Mejoramiento de la Calidad , Humanos , Tiempo de Internación , Servicio de Urgencia en Hospital , Flujo de Trabajo
2.
J Immunol ; 182(10): 6401-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414793

RESUMEN

Progenitor mast cells (prMCs), derived from CD34(+) precursors are CD4(+)/CCR5(+)/CXCR4(+) and susceptible to CCR5(R5)-tropic virus but only marginally susceptible to CXCR4(X4)-tropic HIV. As infected prMCs mature within extravascular compartments, they become both latently infected and HIV-infection resistant, and thus capable of establishing an inducible reservoir of CCR5-tropic infectious clones. In this report we provide the first evidence that IgE-FcepsilonRI interactions, occurring during a unique period of mast cell (MC) ontogeny, enhance prMC susceptibility to X4 and R5X4 virus. IgE-FcepsilonRI interactions significantly increased expression of CXCR4 mRNA ( approximately 400- to 1800-fold), enhanced prMC susceptibility to X4 and R5X4 virus ( approximately 3000- to 16,000-fold), but had no significant effect on CD4, CCR3, or CCR5 expression, susceptibility to R5 virus, or degranulation. Enhanced susceptibility to infection with X4 virus occurred during the first 3-5 wk of MC ontogeny and was completely inhibited by CXCR4-specific peptide antagonists and omalizumab, a drug that inhibits IgE-FcepsilonRI interactions. IgE-FcepsilonRI coaggregation mediated by HIVgp120 or Schistosoma mansoni soluble egg Ag accelerated maximal CXCR4 expression and susceptibility to X4 virus by prMCs. Our findings suggest that for HIV-positive individuals with atopic or helminthic diseases, elevated IgE levels could potentially influence the composition of CXCR4-tropic and R5X4-tropic variants archived within the long-lived tissue MC reservoir created during infection.


Asunto(s)
Infecciones por VIH/inmunología , VIH/inmunología , Inmunoglobulina E/inmunología , Mastocitos/virología , Receptores CXCR4/inmunología , Receptores de IgE/inmunología , Animales , Antígenos Helmínticos/inmunología , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Citometría de Flujo , Expresión Génica , Infecciones por VIH/complicaciones , Helmintiasis/complicaciones , Helmintiasis/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Hipersensibilidad Inmediata/complicaciones , Hipersensibilidad Inmediata/inmunología , Inmunoglobulina E/metabolismo , Inmunofenotipificación , Macaca mulatta , Mastocitos/inmunología , Receptores CCR3/inmunología , Receptores CCR3/metabolismo , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Receptores de IgE/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Am J Obstet Gynecol ; 197(4): 371.e1-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17904964

RESUMEN

OBJECTIVE: The purpose of this study was to determine levels of intrauterine infection and transcriptional activity in cord blood mononuclear cells that were collected at term from fetuses who were born to women who were infected with human immunodeficiency virus (HIV) and who received highly active antiretroviral therapy (HAART). STUDY DESIGN: RNA and DNA were isolated from maternal placental tissues and fetal cord blood specimens that were obtained at term from pregnant women who were infected with HIV and who received HAART. Levels of integrated HIV provirus and messenger RNA transcripts were determined by real-time polymerase chain reaction. RESULTS: Detectable levels of transcriptionally active integrated provirus were present in approximately 27% of cord blood samples (n = 22) that were collected from fetuses who born to HIV-positive mothers who received HAART. Levels of HIV-p24 antigen in cultures that were detected in randomly selected cord blood samples confirmed the presence of inducible infectious virus. CONCLUSION: These findings suggest that some fetuses from HIV-infected mothers who receive HAART and who may be HIV-negative infants after delivery can harbor circulating leukocytes that are infected productively by intrauterine transmission of HIV.


Asunto(s)
Sangre Fetal/virología , Infecciones por VIH/sangre , Infecciones por VIH/transmisión , VIH-1/genética , Transmisión Vertical de Enfermedad Infecciosa , Leucocitos Mononucleares/virología , Complicaciones Infecciosas del Embarazo/virología , Adulto , ADN Viral/sangre , Femenino , Infecciones por VIH/virología , Humanos , Recién Nacido , Modelos Lineales , Masculino , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/sangre , Provirus/genética , ARN Mensajero/sangre , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Integración Viral/genética
4.
J Bone Miner Res ; 17(3): 406-14, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11874232

RESUMEN

Rat LIM mineralization protein 1 (LMP-1, an LIM domain protein) mediates bone morphogenetic protein 6 (BMP-6) induction of bone nodule formation in fetal rat calvarial osteoblast (ROB) cultures. We have isolated the complementary DNA (cDNA) for the human homologue of LMP-1 from an adult human heart cDNA library and showed that when overexpressed it is osteoinductive in the same culture system. The recently revised cDNA sequence of Enigma, the protein product of which binds to the insulin receptor and the tyrosine kinase receptor ret, now matches the nucleotide sequence of human LMP-1 (hLMP-1). A truncated, 223 amino acid (AA) LMP-1(t) protein has identical effects as the full-length protein, despite the deletion of the LIM domains. Two splice variants of human LMP-1 have been detected. Human LMP-2 has a 119-base pair (bp) deletion between bp 325 and 444 and a 17-bp insertion at bp 444. The resulting derived protein contains 423 AA with the LIM domains intact and does not induce bone formation when overexpressed in ROB cultures. Human LMP-3 has the same 17 nucleotide insertion at bp 444, resulting in a shift in the reading frame that causes a stop codon to occur at bp 505-507. The resulting 153 AA protein does not have the LIM domains, but overexpression of hLMP-3 induces bone formation in osteoblast cultures. These findings suggest that the LIM domains are not required for LMPs to induce bone formation. In addition, a small region (36 AA) of the LMP-1 protein may be required for bone formation.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Proteínas Adaptadoras Transductoras de Señales , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/química , Diferenciación Celular , Células Cultivadas , Clonación Molecular , Medios de Cultivo Condicionados , Proteínas del Citoesqueleto , ADN Complementario/genética , Expresión Génica , Variación Genética , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Datos de Secuencia Molecular , Osteoblastos/citología , Osteoblastos/fisiología , Estructura Terciaria de Proteína , Ratas , Homología de Secuencia de Aminoácido , Distribución Tisular , Transfección
5.
Bone ; 35(3): 673-81, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15336603

RESUMEN

Addition of dexamethasone (Dex) to human mesenchymal stem cells (hMSCs) resulted in a 16-fold increase in human bone morphogenetic protein-6 (hBMP-6) mRNA levels 24 h after treatment. Evaluation of luciferase expression after transfection of HeLa cells with hBMP-6 promoter/luciferase reporter constructs indicated that the hBMP-6 promoter activity was contained in a 268-bp region (-1051 to -784 where +1 is the translation start site) over 600 bases 5' to that previously published. It further showed that the promoter activity is regulated by glucocorticoid treatment. Analysis of RNA from hMSCs and HeLa cells by primer extension, RNase protection, and 5' RACE further narrowed the location of the transcription start site to an 84-bp region (-940 to -857). To determine whether this start site was regulated in hMSCs, hBMP-6 mRNA levels in control and Dex-treated cells were quantitated by RT-PCR using one primer set in the translated region of the gene and one located just 3' of the 84-bp region. Both primer sets showed hBMP-6 mRNA levels approximately 16- to 22-fold higher in the Dex-treated cells, demonstrating that hBMP-6 transcription is being regulated by glucocorticoids in the pluripotent hMSCs at the upstream transcription start site.


Asunto(s)
Proteínas Morfogenéticas Óseas/biosíntesis , Glucocorticoides/farmacología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Secuencia de Bases/genética , Proteína Morfogenética Ósea 6 , Proteínas Morfogenéticas Óseas/genética , Células HeLa , Humanos , Datos de Secuencia Molecular
6.
Chem Commun (Camb) ; (3): 424-5, 2003 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-12613644

RESUMEN

Novel supramolecular architectures are observed in the solid state structures of [AlMe(C6F5)(mu-Me)]2 (1) and Ga(C6F5)2Me (2) via pi-pi stacking between C6F5 rings and intermolecular aryl-F-->Ga interactions, respectively.

7.
J Bone Joint Surg Am ; 85(6): 1030-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12783998

RESUMEN

BACKGROUND: LIM mineralization protein-1 (LMP-1), an intracellular protein, is thought to induce secretion of soluble factors that convey its osteoinductive activity. Although evidence suggests that LMP-1 may be a critical regulator of osteoblast differentiation in vitro and in vivo, little is known about its mechanism of action. The purpose of the present study was to identify candidates for the induced secreted factors and to describe the time sequence of histological changes during bone formation induced by LMP-1. METHODS: Human lung carcinoma (A549) cells were used to determine if LMP-1 overexpression would induce expression of bone morphogenetic proteins (BMPs) in vitro. Cultured A549 cells were infected with recombinant replication-deficient human type-5 adenovirus containing the LMP-1 or LacZ cDNA. Cells were subjected to immunohistochemical analysis after forty-eight hours. Finally, sixteen athymic rats received subcutaneous implants consisting of collagen disks loaded with human buffy-coat cells that were infected with one of the above two viruses. Rats were killed at intervals, and explants were studied with histological and immunohistochemical analyses. RESULTS: In vitro experiments with A549 cells showed that AdLMP-1-infected cells express elevated levels of BMP-2, BMP-4, BMP-6, BMP-7, and TGF-beta1 (transforming growth factor-beta 1) protein. Human buffy-coat cells infected with AdLMP-1 also demonstrated increased levels of BMP-4 and BMP-7 protein seventy-two hours after ectopic implantation in athymic rats, confirming the in vitro hypothesis. CONCLUSIONS: The osteoinductive properties of LMP-1 involve synthesis of several BMPs and the recruitment of host cells that differentiate and participate in direct membranous bone formation.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas Portadoras/genética , ADN Complementario/genética , Técnicas de Transferencia de Gen , Osteogénesis/genética , Dedos de Zinc/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Portadoras/biosíntesis , Proteínas del Citoesqueleto , Vectores Genéticos/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Operón Lac/genética , Ratas , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas/fisiología
8.
Blood ; 109(12): 5293-300, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17351109

RESUMEN

We have proposed that, unlike other HIV-vulnerable cell lineages, progenitor mast cells (prMCs), cultured in vitro from undifferentiated bone marrow-derived CD34(+) pluripotent progenitors (PPPs), are susceptible to infection during a limited period of their ontogeny. As infected prMCs mature in culture, they lose expression of viral chemokine coreceptors necessary for viral entry and develop into long-lived, latently infected mature tissue mast cells (MCs), resistant to new infection. In vivo recruitment of prMCs to different tissue compartments occurs in response to tissue injury, growth, and remodeling or allergic inflammation, allowing populations of circulating and potentially HIV-susceptible prMCs to spread persistent infection to diverse tissue compartments. In this report, we provide in vivo evidence to confirm this model by demonstrating that HIV-infected women have both circulating prMCs and placental tissue MCs (PLMCs) that harbor inducible infectious HIV even after highly active antiretroviral therapy (HAART) during pregnancy. Furthermore, infectious virus, capable of infecting alloactivated fetal cord blood mononuclear cells (CBMCs), could be induced in isolated latently infected PLMCs after weeks in culture in vitro. These data provide the first in vivo evidence that tissue MCs, developed from infected circulating prMCs, comprise a long-lived inducible reservoir of persistent HIV in infected persons during HAART.


Asunto(s)
Infecciones por VIH/patología , Mastocitos/virología , Adulto , Terapia Antirretroviral Altamente Activa , Células Cultivadas , Femenino , Sangre Fetal/virología , Infecciones por VIH/transmisión , Humanos , Placenta/virología , Embarazo , Receptores del VIH/análisis
9.
Blood ; 107(1): 187-9, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16123213

RESUMEN

Increased rates of graft rejection after bone marrow transplantation (BMT) are observed in patients whose illnesses--such as sickle cell disease, thalassemia, and aplastic anemia--necessitate chronic transfusion before BMT. Because BM transplants in these patients are routinely HLA matched, any immunization responsible for increased rejection is likely against minor histocompatibility antigens (mHAs). It has been assumed that contaminating leukocytes in red blood cell (RBC) units are the main sources of immunization to mHAs. However, in this report, we demonstrate that antigens on donor RBCs are presented in the major histocompatibility complex (MHC) class I pathway of recipient antigen-presenting cells, resulting in activation and expansion of recipient CD8+ T cells specific for donor mHAs. Given that human hematopoietic progenitor cells express many of the known mHAs, this observation provides a mechanism by which chronic transfusion of even stringently leukoreduced RBCs may result in sufficient mHA immunization to increase the frequency of BMT rejection.


Asunto(s)
Reactividad Cruzada , Transfusión de Eritrocitos , Eritrocitos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunización , Antígenos de Histocompatibilidad Menor/inmunología , Animales , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Pollos , Rechazo de Injerto/etiología , Rechazo de Injerto/inmunología , Procedimientos de Reducción del Leucocitos , Ratones , Ovalbúmina
10.
Blood ; 106(3): 1105-12, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15831698

RESUMEN

Transfusion of red blood cells (RBCs) into patients with anti-donor RBC antibodies (crossmatch-incompatible transfusion) can result in lethal antibody-mediated hemolysis. Less well appreciated is the ability of anti-RBC antibodies to specifically remove their target antigen from donor RBCs without compromising cell survival or adversely affecting the transfusion recipient. In an effort to elucidate the mechanistic details of this process, we describe the first animal model of nonhemolytic antibody-induced RBC antigen loss. RBCs from transgenic mHEL mice express surface hen egg lysozyme (HEL) as a transmembrane protein. Transfusion of mHEL RBCs into mice immunized with HEL results in selective loss of HEL antigen from donor RBCs without affecting other blood group antigens or reducing the circulatory life span of the transfused RBCs. While this process does not require the presence of a spleen, it requires both anti-RBC immunoglobulin G (IgG) antibodies and the FcgammaIII receptor. These studies provide mechanistic insight into the phenomenon of antigen loss during incompatible transfusion in humans.


Asunto(s)
Reacciones Antígeno-Anticuerpo/inmunología , Antígenos de Superficie/inmunología , Eritrocitos/inmunología , Isoanticuerpos/inmunología , Animales , Inmunización , Inmunoglobulina G , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Muramidasa/administración & dosificación , Muramidasa/genética , Muramidasa/inmunología , Receptores de IgG/inmunología , Reacción a la Transfusión
11.
Spine (Phila Pa 1976) ; 28(3): 219-26, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12567021

RESUMEN

STUDY DESIGN: An animal study in immune competent rabbits and athymic rats was conducted. OBJECTIVES: To develop an animal model for simulation of previous human Type 5 adenovirus (Ad5) exposure, to determine the impact of adenoviral pre-exposure on spine fusion induced with ex vivo Ad5-LMP-1, and to test strategies for overcoming any potential immune response. SUMMARY OF BACKGROUND DATA: Cells transduced with adenovirus containing the osteoinductive LMP-1 cDNA (Ad5-LMP-1) can induce spine fusion in rabbits. Because up to 80% of the human population has been exposed to adenovirus, immune responses to the vector may limit this strategy in humans. Few studies have modeled previous adenoviral exposure and tested strategies to circumvent it. METHODS: Adult New Zealand white rabbits were injected with 10 or 10 viral particles of Ad5-LacZ. At 4 or 16 weeks after Ad5 injection, autologous buffy coats were prepared from peripheral blood, and 4 million cells per side were infected ex vivo for 10 minutes with Ad5-LMP-1 (multiplicity of infection = 4). Cells were implanted on a collagen matrix instead of an autograft for posterolateral lumbar arthrodesis. Unimmunized rabbits served as control subjects. Additional immunized rabbits underwent arthrodesis at 4 weeks with increased cell number (10 million) and viral dose (multiplicity of infection = 10), or with both parameters increased. The rabbits were killed at 4 weeks, and the spines were assessed by palpation and radiograph. A parallel study was performed in athymic rats using immunized rabbits for the donor cells. RESULTS: All the unimmunized rabbits had solid spine fusions. None of the rabbits arthrodesed 4 weeks after Ad5 pre-exposure achieved fusion. At 4 weeks after Ad5 exposure, increasing the multiplicity of infection to 10 did not overcome the immune response (0/3 fused), but increasing the cell number to 10 million (2/3 fused) or increasing both cell number and multiplicity of infection (3/3 fused) did overcome the immune effects. Delaying arthrodesis until 16 weeks after Ad5 pre-exposure also overcame the immune response (3/3 fused). Similar results were seen in the athymic rat ectopic implant model, suggesting that the immune effect was mediated by humoral antibodies rather than a T-cell response. CONCLUSIONS: Two model systems were developed that simulate previous exposure to human Ad5 and could separate the cellular and humoral components of the response. There was a dose-dependent inhibition of ex vivo Ad5-LMP-1 gene transfer to cells from animals previously exposed to human Ad5. Data suggested that the inhibition of Ad5 infection was caused by humoral antibodies rather than a T-cell-based response. Minor modifications in the gene transfer protocol, such as doubling the viral dose or number of cells infected, or increasing the infection time, could overcome the immune response for an ex vivo approach.


Asunto(s)
Adenoviridae/inmunología , Proteínas Portadoras/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/inmunología , Fusión Vertebral/métodos , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae/genética , Animales , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Calcificación Fisiológica/efectos de los fármacos , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Línea Celular , Proteínas del Citoesqueleto , ADN Complementario/administración & dosificación , ADN Complementario/inmunología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/antagonistas & inhibidores , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Región Lumbosacra , Conejos , Radiografía , Ratas , Ratas Desnudas , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/cirugía , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA