Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Nurs Care Qual ; 37(3): 206-212, 2022.
Article in English | MEDLINE | ID: mdl-34775422

ABSTRACT

BACKGROUND: A collaborative and multidisciplinary shared responsibility with providing discharge information to pediatric patients/families can support successful reassimilation to life posthospitalization. PROBLEM: An analysis of the current discharge process at an urban pediatric hospital identified variations in discharge practices and instructions. APPROACH: A multidisciplinary taskforce used the Plan-Do-Study-Act methodology to standardize the discharge process including creating a discharge template, adopting a new education platform, enhancing the electronic health record, and implementing strategies to improve discharge. OUTCOMES: There was a reduction in 7-day readmission rate from preintervention 4.58 to postintervention for 4 consecutive quarters of 3.92, 4.20, 3.44, and 3.41, respectively. Percentile ranking and top box scores of the patient satisfaction measure of discharge preparation increased from 12th-62nd to 65th-95th percentile postimplementation and 81% to 88%, respectively. CONCLUSIONS: A local improvement initiative related to standardization of the discharge process resulted in a reduction in 7-day readmission rate and improved patient satisfaction scores.


Subject(s)
Patient Discharge , Quality Improvement , Child , Hospitals, Pediatric , Hospitals, Urban , Humans , Patient Readmission , Patient Satisfaction
2.
Curr Opin Hematol ; 27(1): 11-17, 2020 01.
Article in English | MEDLINE | ID: mdl-31652152

ABSTRACT

PURPOSE OF REVIEW: WHIM syndrome (warts, hypogammaglobulinemia, immunodeficiency, myelokathexis, or WHIMs) is a very rare autosomal dominant immunodeficiency disorder attributable to mutations in CXCR4. We reviewed clinical manifestations in 24 patients in 9 families to expand understanding of this syndrome. RECENT FINDINGS: Warts, cellulitis and respiratory infections are common in patients with WHIMs. Less commonly these patients have congenital heart disease, human papilloma virus-associated malignancies (cervical and vulvular) and lymphomas. Hearing loss because of recurrent otitis media is another important complication. Treatment with granulocyte colony-stimulating factor is controversial; this review indicates that it is effective to prevent and treat infections based upon long-term observations of patients enrolled in the Severe Chronic Neutropenia International Registry. Understanding the natural history and diversity of this syndrome are important for ongoing clinical trials of novel agents to treat WHIMs. SUMMARY: WHIM syndrome has diverse manifestations; some features occur consistently in almost all patients, for example, neutropenia, lymphocytopenia and mild hypogammaglobulinemia. However, the clinical consequences are quite variable across patient cohorts and within families. Each complication is important as a cause for morbidity and a source for patient and family concerns.


Subject(s)
Agammaglobulinemia , Family , Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Registries , Warts , Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Agammaglobulinemia/therapy , Female , Humans , Male , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/therapy , Risk Factors , Warts/diagnosis , Warts/genetics , Warts/pathology , Warts/therapy
3.
Cancer Immunol Res ; 3(9): 1052-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25943534

ABSTRACT

Programmed cell-death 1 ligand 1 (PD-L1) is a member of the B7/CD28 family of proteins that control T-cell activation. Many tumors can upregulate expression of PD-L1, inhibiting antitumor T-cell responses and avoiding immune surveillance and elimination. We have identified and characterized MEDI4736, a human IgG1 monoclonal antibody that binds with high affinity and specificity to PD-L1 and is uniquely engineered to prevent antibody-dependent cell-mediated cytotoxicity. In vitro assays demonstrate that MEDI4736 is a potent antagonist of PD-L1 function, blocking interaction with PD-1 and CD80 to overcome inhibition of primary human T-cell activation. In vivo MEDI4736 significantly inhibits the growth of human tumors in a novel xenograft model containing coimplanted human T cells. This activity is entirely dependent on the presence of transplanted T cells, supporting the immunological mechanism of action for MEDI4736. To further determine the utility of PD-L1 blockade, an anti-mouse PD-L1 antibody was investigated in immunocompetent mice. Here, anti-mouse PD-L1 significantly improved survival of mice implanted with CT26 colorectal cancer cells. The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Taken together, our results demonstrate that inhibition of PD-L1 function can have potent antitumor activity when used as monotherapy or in combination in preclinical models, and suggest it may be a promising therapeutic approach for the treatment of cancer. MEDI4736 is currently in several clinical trials both alone and in combination with other agents, including anti-CTLA-4, anti-PD-1, and inhibitors of IDO, MEK, BRAF, and EGFR.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Binding, Competitive , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Melanoma/immunology , Melanoma/pathology , Melanoma/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Stem Cells Dev ; 22(20): 2714-24, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23829870

ABSTRACT

With an incidence of ∼1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harboring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ∼30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.


Subject(s)
Dystrophin/genetics , Gene Expression/drug effects , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation , Myocytes, Cardiac/drug effects , Oligonucleotides, Antisense/pharmacology , Base Sequence , Cell Differentiation , Child , Dystrophin/metabolism , Exons , Gene Transfer Techniques , Genetic Therapy , Humans , Induced Pluripotent Stem Cells/pathology , Male , Molecular Sequence Data , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture
5.
Stem Cells Dev ; 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23786351

ABSTRACT

With an incidence of ~1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harbouring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ~30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.

6.
Mol Ther ; 19(9): 1695-703, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21694703

ABSTRACT

The limited ability of the heart to regenerate has prompted development of new systems to produce cardiomyocytes for therapeutics. While differentiation of human embryonic stem cells (hESCs) into cardiomyocytes has been well documented, the process remains inefficient and/or expensive, and progress would be facilitated by better understanding the early genetic events that cause cardiac specification. By maintaining a transgenic cardiac-specific MYH6-monomeric red fluorescent protein (mRFP) reporter hESC line in conditions that promote pluripotency, we tested the ability of combinations of 15 genes to induce cardiac specification. Screening identified GATA4 plus TBX5 as the minimum requirement to activate the cardiac gene regulatory network and produce mRFP(+) cells, while a combination of GATA4, TBX5, NKX2.5, and BAF60c (GTNB) was necessary to generate beating cardiomyocytes positive for cTnI and α-actinin. Including the chemotherapeutic agent, Ara-C, from day 10 of induced differentiation enriched for cTnI/α-actinin double positive cells to 45%. Transient expression of GTNB for 5-7 days was necessary to activate the cardiogenesis through progenitor intermediates in a manner consistent with normal heart development. This system provides a route to test the effect of different factors on human cardiac differentiation and will be useful in understanding the network failures that underlie disease phenotypes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Gene Regulatory Networks , Actinin/metabolism , Cell Line , Chromosomal Proteins, Non-Histone , Cloning, Molecular , Cytarabine/pharmacology , Embryonic Stem Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Vectors , Heart/embryology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lentivirus/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Myocytes, Cardiac/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Red Fluorescent Protein
7.
Nat Protoc ; 6(6): 701-14, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21637193

ABSTRACT

The protocols described here address methods used in two crucial stages in the retroviral reprogramming of somatic cells to produce human induced pluripotent stem cell (hiPSC) lines. The first is an optimized method for producing lentivirus at an efficiency 600-fold greater than previously published, and it includes conjugation of the lentivirus to streptavidin superparamagnetic particles; this process takes 8 d. The second method enables the isolation of true hiPSCs immediately after somatic cell reprogramming and involves column-based positive selection of cells expressing the pluripotency marker TRA-1-81. This process takes 2 h and, as it is directly compatible with feeder-free culture, the time burden of manually identifying and mechanically propagating hiPSC colonies is reduced drastically. Taken together, these methods accelerate the production of hiPSCs and enable lines to be isolated, expanded to approxiamtely 107 cells and cryopreserved within 6-8 weeks.


Subject(s)
Cell Culture Techniques , Cell Line , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Animals , Humans , Mice , Virus Cultivation
8.
J Virol ; 85(15): 7912-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21593174

ABSTRACT

Rhinoviral infection is an important trigger of acute inflammatory exacerbations in patients with underlying airway disease. We have previously established that interleukin-1ß (IL-1ß) is central in the communication between epithelial cells and monocytes during the initiation of inflammation. In this study we explored the roles of IL-1ß and its signaling pathways in the responses of airway cells to rhinovirus-1B (RV-1B) and further determined how responses to RV-1B were modified in a model of bacterial coinfection. Our results revealed that IL-1ß dramatically potentiated RV-1B-induced proinflammatory responses, and while monocytes did not directly amplify responses to RV-1B alone, they played an important role in the responses observed with our coinfection model. MyD88 is the essential signaling adapter for IL-1ß and most Toll-like receptors. To examine the role of MyD88 in more detail, we created stable MyD88 knockdown epithelial cells using short hairpin RNA (shRNA) targeted to MyD88. We determined that IL-1ß/MyD88 plays a role in regulating RV-1B replication and the inflammatory response to viral infection of airway cells. These results identify central roles for IL-1ß and its signaling pathways in the production of CXCL8, a potent neutrophil chemoattractant, in viral infection. Thus, IL-1ß is a viable target for controlling the neutrophilia that is often found in inflammatory airway disease and is exacerbated by viral infection of the airways.


Subject(s)
Interleukin-1beta/physiology , Myeloid Differentiation Factor 88/metabolism , Picornaviridae Infections/metabolism , Rhinovirus/isolation & purification , Signal Transduction , Blotting, Western , Cell Communication , Cell Line , Cytopathogenic Effect, Viral , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Humans , Myeloid Differentiation Factor 88/genetics , Picornaviridae Infections/virology , Polymerase Chain Reaction , Rhinovirus/pathogenicity
9.
Eur Heart J ; 32(8): 952-62, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21367833

ABSTRACT

AIMS: Congenital long QT syndromes (LQTSs) are associated with prolonged ventricular repolarization and sudden cardiac death. Limitations to existing clinical therapeutic management strategies prompted us to develop a novel human in vitro drug-evaluation system for LQTS type 2 (LQT2) that will complement the existing in vitro and in vivo models. METHODS AND RESULTS: Skin fibroblasts from a patient with a KCNH2 G1681A mutation (encodes I(Kr) potassium ion channel) were reprogrammed to human induced pluripotent stem cells (hiPSCs), which were subsequently differentiated to functional cardiomyocytes. Relative to controls (including the patient's mother), multi-electrode array and patch-clamp electrophysiology of LQT2-hiPSC cardiomyocytes showed prolonged field/action potential duration. When LQT2-hiPSC cardiomyocytes were exposed to E4031 (an I(Kr) blocker), arrhythmias developed and these presented as early after depolarizations (EADs) in the action potentials. In contrast to control cardiomyocytes, LQT2-hiPSC cardiomyocytes also developed EADs when challenged with the clinically used stressor, isoprenaline. This effect was reversed by ß-blockers, propranolol, and nadolol, the latter being used for the patient's therapy. Treatment of cardiomyocytes with experimental potassium channel enhancers, nicorandil and PD118057, caused action potential shortening and in some cases could abolish EADs. Notably, combined treatment with isoprenaline (enhancers/isoprenaline) caused EADs, but this effect was reversed by nadolol. CONCLUSIONS: Findings from this paper demonstrate that patient LQT2-hiPSC cardiomyocytes respond appropriately to clinically relevant pharmacology and will be a valuable human in vitro model for testing experimental drug combinations.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Ether-A-Go-Go Potassium Channels/genetics , Induced Pluripotent Stem Cells/physiology , Long QT Syndrome/genetics , Point Mutation/genetics , Adolescent , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Drug Evaluation, Preclinical/methods , ERG1 Potassium Channel , Electrocardiography , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Isoproterenol/pharmacology , Long QT Syndrome/drug therapy , Myocytes, Cardiac/physiology , Nicorandil/pharmacology
10.
Stem Cell Res ; 6(2): 158-67, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21095172

ABSTRACT

There are two critical stages in the retroviral reprogramming of somatic cells to produce human induced pluripotent stem cell (hiPSC) lines. One is the production of high titer virus required to reprogram somatic cells; the other is identification of true hiPSC colonies from heterogeneous cell populations, and their isolation and expansion to generate a sustainable, pluripotent stem cell line. Here we describe simple, time-saving methods to address the current difficulties at these two critical junctures. First, we have developed a method to increase the number of infectious viral units 600-fold. Second, we have developed a TRA-1-81-based positive selection column method for isolating "true" hiPSCs from the heterogeneous cell populations, which overcomes the labor-intensive and highly subjective method of manual selection of hiPSC colonies. We have used these techniques to produce 8 hiPSC lines from human fibroblasts and we believe that they are of considerable utility to researchers in the hiPSC field.


Subject(s)
Cell Culture Techniques/methods , Cell Line , Cell Separation/methods , Induced Pluripotent Stem Cells/physiology , Animals , Biomarkers/metabolism , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation
11.
Biochem Soc Trans ; 38(4): 1037-45, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20659000

ABSTRACT

Functional cardiomyocytes can now be derived routinely from hPSCs (human pluripotent stem cells), which collectively include embryonic and induced pluripotent stem cells. This technology presents new opportunities to develop pharmacologically relevant in vitro screens to detect cardiotoxicity, with a view to improving patient safety while reducing the economic burden to industry arising from high drug attrition rates. In the present article, we consider the need for human cardiomyocytes in drug-screening campaigns and review the strategies used to differentiate hPSCs towards the cardiac lineage. During early stages of differentiation, hPSC-cardiomyocytes display gene expression profiles, ultra-structures, ion channel functionality and pharmacological responses reminiscent of an embryonic phenotype, but maturation during extended time in culture has been demonstrated convincingly. Notably, hPSC-cardiomyocytes have been shown to respond in a highly predictable manner to over 40 compounds that have a known pharmacological effect on the human heart. This suggests that further development and validation of the hPSC-cardiomyocyte model as a tool for assessing cardiotoxicity is warranted.


Subject(s)
Drug Evaluation, Preclinical/methods , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Gene Expression Profiling , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/isolation & purification , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
12.
Immunology ; 127(2): 249-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19175800

ABSTRACT

Human neutrophils express Toll-like receptor 4 (TLR4) at low levels, and the role of this receptor in neutrophil responses to microbial stimuli has been questioned. Genetic manipulation of these cells to enable the study of the role of proteins such as TLR4 in their function is challenging. Here, we show that primary human neutrophils rapidly express novel proteins such as enhanced green fluorescent protein (eGFP) after transduction with lentivirus. Stimulation of transduced neutrophils with lipopolysaccharide (LPS) resulted in increased cell survival, which was inhibited when neutrophils were transduced with a lentivirus encoding a dominant negative (dn) TLR4 protein. LPS-induced survival was also inhibited by lentiviruses encoding dnMyD88 or a truncated TRIF (Toll/interleukin-1R homologous domain-containing adapter protein inducing interferon-beta) molecule, whilst, in contrast, neutrophil survival was enhanced by overexpression of kinase-mutated interleukin-1 receptor-associated kinase 1 (kmIRAK-1), which activated nuclear factor (NF)-kappaB. These studies provide proof of the role of TLR4 in human neutrophil biology, have begun to elucidate TLR-dependent pathways regulating neutrophil survival, and demonstrate that neutrophils can be genetically manipulated to enhance or inhibit survival.


Subject(s)
Lipopolysaccharides/immunology , Neutrophils/immunology , Toll-Like Receptor 4/immunology , Cell Survival/immunology , Cells, Cultured , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Neutrophil Activation/immunology , Signal Transduction/immunology , Transduction, Genetic
13.
Stem Cells ; 26(10): 2552-63, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18669908

ABSTRACT

Neutropenia as a consequence of bone marrow failure, severe infections, or intensive chemotherapy is frequently associated with life-threatening sepsis. Ex vivo expansion of CD34(+) stem cells has been shown to generate apparently functional neutrophils, and the use of autologous ex vivo-expanded cells can reduce the duration of neutropenia. Nonetheless, the principal antimicrobial capabilities of such cells, and thus their true therapeutic potential, is unknown. Using established protocols, we derived mature neutrophils from normal human adult bone marrow (BM) CD34(+) cells and compared them with freshly isolated peripheral blood neutrophils (PBN). Despite functional similarities between ex vivo-differentiated neutrophils (EDN) and PBN in assays of respiratory burst and phagocytosis, EDN showed marked impairment in their ability to kill both Escherichia coli and Streptococcus pneumoniae compared with PBN. We found that EDN were able to detect (through Toll-like receptor 2 [TLR2], TLR4, and CD14 expression), phagocytose, and mount a respiratory burst to microorganisms. EDN, however, were unable to release neutrophil elastase in response to formyl-met-leu-phe and showed a significantly reduced expression of neutrophil elastase, cathepsin G myeloperoxidase, and LL-37/human cathelicidin protein 18 (hCAP18) as determined by Western blotting. Ultrastructural analysis was consistent with a failure of normal granule development in EDN. Neutrophils derived from BM CD34(+) cells may therefore provide apparently functional cells as assessed by common methodologies; however, important deficiencies may still limit their therapeutic potential. The results presented here suggest additional key tests that such cells may need to undergo prior to clinical use and highlight the potential challenges of using ex vivo modified stem cells in therapeutic settings. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Microbial Viability , Neutrophils/cytology , Adult , Antimicrobial Cationic Peptides/metabolism , Cell Differentiation , Cell Proliferation , Cell Separation , Chemotaxis , Cytokines/biosynthesis , Cytoplasmic Granules/ultrastructure , Escherichia coli/cytology , Humans , Leukocyte Elastase/metabolism , Neutrophils/enzymology , Neutrophils/ultrastructure , Phagocytosis , Respiratory Burst , Stem Cells/cytology , Streptococcus pneumoniae/cytology , Toll-Like Receptors/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...