Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Front Immunol ; 14: 1275937, 2023.
Article in English | MEDLINE | ID: mdl-37920467

ABSTRACT

Background: Human perinatal life is characterized by a period of extraordinary change during which newborns encounter abundant environmental stimuli and exposure to potential pathogens. To meet such challenges, the neonatal immune system is equipped with unique functional characteristics that adapt to changing conditions as development progresses across the early years of life, but the molecular characteristics of such adaptations remain poorly understood. The application of single cell genomics to birth cohorts provides an opportunity to investigate changes in gene expression programs elicited downstream of innate immune activation across early life at unprecedented resolution. Methods: In this study, we performed single cell RNA-sequencing of mononuclear cells collected from matched birth cord blood and 5-year peripheral blood samples following stimulation (18hrs) with two well-characterized innate stimuli; lipopolysaccharide (LPS) and Polyinosinic:polycytidylic acid (Poly(I:C)). Results: We found that the transcriptional response to LPS was constrained at birth and predominantly partitioned into classical proinflammatory gene upregulation primarily by monocytes and Interferon (IFN)-signaling gene upregulation by lymphocytes. Moreover, these responses featured substantial cell-to-cell communication which appeared markedly strengthened between birth and 5 years. In contrast, stimulation with Poly(I:C) induced a robust IFN-signalling response across all cell types identified at birth and 5 years. Analysis of gene regulatory networks revealed IRF1 and STAT1 were key drivers of the LPS-induced IFN-signaling response in lymphocytes with a potential developmental role for IRF7 regulation. Conclusion: Additionally, we observed distinct activation trajectory endpoints for monocytes derived from LPS-treated cord and 5-year blood, which was not apparent among Poly(I:C)-induced monocytes. Taken together, our findings provide new insight into the gene regulatory landscape of immune cell function between birth and 5 years and point to regulatory mechanisms relevant to future investigation of infection susceptibility in early life.


Subject(s)
Lipopolysaccharides , Transcriptome , Infant, Newborn , Humans , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Monocytes , Signal Transduction , Gene Expression Regulation , Poly I-C/pharmacology , Poly I-C/metabolism
2.
Leukemia ; 37(1): 61-71, 2023 01.
Article in English | MEDLINE | ID: mdl-36380143

ABSTRACT

Infants with KMT2A-rearranged B-cell acute lymphoblastic leukemia (ALL) have a dismal prognosis. Survival outcomes have remained static in recent decades despite treatment intensification and novel therapies are urgently required. KMT2A-rearranged infant ALL cells are characterized by an abundance of promoter hypermethylation and exhibit high BCL-2 expression, highlighting potential for therapeutic targeting. Here, we show that hypomethylating agents exhibit in vitro additivity when combined with most conventional chemotherapeutic agents. However, in a subset of samples an antagonistic effect was seen between several agents. This was most evident when hypomethylating agents were combined with methotrexate, with upregulation of ATP-binding cassette transporters identified as a potential mechanism. Single agent treatment with azacitidine and decitabine significantly prolonged in vivo survival in KMT2A-rearranged infant ALL xenografts. Treatment of KMT2A-rearranged infant ALL cell lines with azacitidine and decitabine led to differential genome-wide DNA methylation, changes in gene expression and thermal proteome profiling revealed the target protein-binding landscape of these agents. The selective BCL-2 inhibitor, venetoclax, exhibited in vitro additivity in combination with hypomethylating or conventional chemotherapeutic agents. The addition of venetoclax to azacitidine resulted in a significant in vivo survival advantage indicating the therapeutic potential of this combination to improve outcome for infants with KMT2A-rearranged ALL.


Subject(s)
Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Infant , Azacitidine/pharmacology , Azacitidine/therapeutic use , Decitabine/pharmacology , Decitabine/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-2 , Leukemia, Myeloid, Acute/genetics
3.
J Telemed Telecare ; 28(10): 740-749, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36346933

ABSTRACT

INTRODUCTION: A prior study examining perceptions of Allied Health Professions (AHP) telehealth services at a metropolitan hospital highlighted multiple issues impacting service uptake, operationalisation, and delivery. Concept mapping methodology was utilised to address these issues and prioritise actionable telehealth service improvements. METHODS: Representatives (n = 22) from seven AHP departments and consumers generated statements addressing the question: 'What do we need to do to enhance and sustain telehealth services?' Statements were synthesised and then clinicians and managers sorted them into similar groups and assigned each statement a ranking of perceived (a) importance and (b) changeability. Multivariate and multidimensional scaling was undertaken to develop a final prioritised set of goals for change. RESULTS: Ninety-six unique statements were generated as actionable goals for change. Statements were grouped into 13 clusters relating to improvements in staff support, infrastructure, consumer support and organisational processes. All clusters were rated >50% for importance (range 3.3-2.4 out of 4) and changeability (range 2.6-2.1 out of 4). Twenty-six statements were ranked highest for importance and changeability. Key prioritised areas were staff training, consumer advocacy and engagement, telehealth operations and workflow. CONCLUSION: Concept mapping was an effective process for generating a prioritised list of actions to enhance AHP telehealth services.


Subject(s)
Telemedicine , Humans , Health Services , Hospitals, Urban
4.
Front Immunol ; 13: 876654, 2022.
Article in English | MEDLINE | ID: mdl-35990635

ABSTRACT

Appropriate innate immune function is essential to limit pathogenesis and severity of severe lower respiratory infections (sLRI) during infancy, a leading cause of hospitalization and risk factor for subsequent asthma in this age group. Employing a systems biology approach to analysis of multi-omic profiles generated from a high-risk cohort (n=50), we found that the intensity of activation of an LPS-induced interferon gene network at birth was predictive of sLRI risk in infancy (AUC=0.724). Connectivity patterns within this network were stronger among susceptible individuals, and a systems biology approach identified IRF1 as a putative master regulator of this response. These findings were specific to the LPS-induced interferon response and were not observed following activation of viral nucleic acid sensing pathways. Comparison of responses at birth versus age 5 demonstrated that LPS-induced interferon responses but not responses triggered by viral nucleic acid sensing pathways may be subject to strong developmental regulation. These data suggest that the risk of sLRI in early life is in part already determined at birth, and additionally that the developmental status of LPS-induced interferon responses may be a key determinant of susceptibility. Our findings provide a rationale for the identification of at-risk infants for early intervention aimed at sLRI prevention and identifies targets which may be relevant for drug development.


Subject(s)
Asthma , Nucleic Acids , Respiratory Tract Infections , Antiviral Agents , Child, Preschool , Humans , Infant , Infant, Newborn , Interferons , Lipopolysaccharides/pharmacology
5.
Nat Commun ; 13(1): 4501, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042201

ABSTRACT

KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) represents the most refractory type of childhood leukemia. To uncover the molecular heterogeneity of this disease, we perform RNA sequencing, methylation array analysis, whole exome and targeted deep sequencing on 84 infants with KMT2A-rearranged leukemia. Our multi-omics clustering followed by single-sample and single-cell inference of hematopoietic differentiation establishes five robust integrative clusters (ICs) with different master transcription factors, fusion partners and corresponding stages of B-lymphopoietic and early hemato-endothelial development: IRX-type differentiated (IC1), IRX-type undifferentiated (IC2), HOXA-type MLLT1 (IC3), HOXA-type MLLT3 (IC4), and HOXA-type AFF1 (IC5). Importantly, our deep mutational analysis reveals that the number of RAS pathway mutations predicts prognosis and that the most refractory subgroup of IC2 possesses 100% frequency and the heaviest burden of RAS pathway mutations. Our findings highlight the previously under-appreciated intra- and inter-patient heterogeneity of KMT2A-rearranged infant ALL and provide a rationale for the future development of genomics-guided risk stratification and individualized therapy.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Gene Fusion , Humans , Infant , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics
6.
J Telemed Telecare ; 27(10): 615-624, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34726993

ABSTRACT

The patient, clinician and administration staff perspectives of telehealth (specifically videoconferencing) services provided by Allied Health Professions (AHP) at a large quaternary hospital were explored. The purpose was to understand stakeholders' perceptions of the service during initial COVID-19 restrictions and examine factors that influenced the implementation and sustained use of telehealth. A sequential mixed-methods approach was undertaken. Stage 1 involved surveys completed by patients (n = 109) and clinicians (n = 66) who received and provided care via telehealth, respectively, across six AHP departments. Stage 2 involved focus groups with clinicians (n = 24) and administrative staff (n = 13) to further examine implementation and sustainability factors.All participant groups confirmed that telehealth was a valid service model and valued the benefits it afforded, particularly during COVID-19 restrictions. Both patients and clinicians reported that not all AHP services could be delivered via telehealth and preferred a blended model of telehealth and in-person care. Increased administrative staff assistance was needed to support growing telehealth demand. Main factors to address are the need to expand AHP telehealth models and workforce/patient training, improve workflow processes and enhance technical support.Despite rapid implementation, telehealth experiences were overall positive. Study findings are being used to generate solutions to enhance and sustain AHP telehealth services.


Subject(s)
COVID-19 , Telemedicine , Hospitals , Humans , SARS-CoV-2 , Videoconferencing
7.
Front Immunol ; 12: 735133, 2021.
Article in English | MEDLINE | ID: mdl-34552594

ABSTRACT

Cancer vaccination drives the generation of anti-tumor T cell immunity and can be enhanced by the inclusion of effective immune adjuvants such as type I interferons (IFNs). Whilst type I IFNs have been shown to promote cross-priming of T cells, the role of individual subtypes remains unclear. Here we systematically compared the capacity of distinct type I IFN subtypes to enhance T cell responses to a whole-cell vaccination strategy in a pre-clinical murine model. We show that vaccination in combination with IFNß induces significantly greater expansion of tumor-specific CD8+ T cells than the other type I IFN subtypes tested. Optimal expansion was dependent on the presence of XCR1+ dendritic cells, CD4+ T cells, and CD40/CD40L signaling. Therapeutically, vaccination with IFNß delayed tumor progression when compared to vaccination without IFN. When vaccinated in combination with anti-PD-L1 checkpoint blockade therapy (CPB), the inclusion of IFNß associated with more mice experiencing complete regression and a trend in increased overall survival. This work demonstrates the potent adjuvant activity of IFNß, highlighting its potential to enhance cancer vaccination strategies alone and in combination with CPB.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/pharmacology , Interferon-beta/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Immune Checkpoint Inhibitors/pharmacology , Interferon-beta/genetics , Interferon-beta/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Transgenic , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Vaccination
8.
Biomolecules ; 11(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-34200313

ABSTRACT

Notch signaling forms an evolutionarily conserved juxtacrine pathway crucial for cellular development. Initially identified in Drosophila wing morphogenesis, Notch signaling has since been demonstrated to play pivotal roles in governing mammalian cellular development in a large variety of cell types. Indeed, abolishing Notch constituents in mouse models result in embryonic lethality, demonstrating that Notch signaling is critical for development and differentiation. In this review, we focus on the crucial role of Notch signaling in governing embryogenesis and differentiation of multiple progenitor cell types. Using hematopoiesis as a diverse cellular model, we highlight the role of Notch in regulating the cell fate of common lymphoid progenitors. Additionally, the influence of Notch through microenvironment interplay with lymphoid cells and how dysregulation influences disease processes is explored. Furthermore, bi-directional and lateral Notch signaling between ligand expressing source cells and target cells are investigated, indicating potentially novel therapeutic options for treatment of Notch-mediated diseases. Finally, we discuss the role of cis-inhibition in regulating Notch signaling in mammalian development.


Subject(s)
Cell Lineage/physiology , Embryonic Development/physiology , Lymphopoiesis/physiology , Receptors, Notch/physiology , Animals , Humans , Lymphocytes/physiology , Signal Transduction/physiology
9.
Mol Immunol ; 128: 150-164, 2020 12.
Article in English | MEDLINE | ID: mdl-33129017

ABSTRACT

During mammalian lymphoid development, Notch signaling is necessary at multiple stages of T lymphopoiesis, including lineage commitment, and later stages of T cell effector differentiation. In contrast, outside of a defined role in the development of splenic marginal zone B cells, there is conflicting evidence regarding whether Notch signaling plays functional roles in other B cell sub-populations. Complement receptor 2 (CR2) modulates BCR-signaling and is tightly regulated throughout differentiation. During B lymphopoiesis, CR2 is detected on immature and mature B cells with high surface expression on marginal zone B cells. Here, we have explored the possibility that Notch regulates human CR2 transcriptional activity using in vitro models including a co-culture system, co-transfection gene reporters and chromatin accessibility assays. We provide evidence that Notch signaling regulates CR2 promoter activity in a mature B cell line, as well as the induction of endogenous CR2 mRNA in a non-expressing pre-B cell line. The dynamics of endogenous gene activation suggests additional unidentified factors are required to mediate surface CR2 expression on immature and mature B lineage cells.


Subject(s)
Complement C3d/genetics , Precursor Cells, B-Lymphoid/physiology , Promoter Regions, Genetic/genetics , Receptors, Complement 3d/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , B-Lymphocytes/physiology , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Chromatin/genetics , Coculture Techniques/methods , Humans , K562 Cells , Lymphocyte Activation/genetics , Lymphopoiesis/genetics
10.
Clin Transl Immunology ; 9(7): e1151, 2020.
Article in English | MEDLINE | ID: mdl-32695339

ABSTRACT

OBJECTIVES: Natural killer (NK) cells are an attractive source of cells for an 'off the shelf' cellular therapy because of their innate capacity to target malignant cells, and ability to be transferred between donors and patients. However, since not all NK cells are equally effective at targeting cancer, selecting the right donor for cellular therapy is critical for the success of the treatment. Recently, cellular therapies utilising NK cells from cytomegalovirus (CMV)-seropositive donors have been explored. However, whether these NK cells are the best source to treat paediatric acute lymphoblastic leukaemia (ALL) remains unclear. METHODS: Using a panel of patient-derived paediatric B- and T-ALL, we assessed the ability of NK cells from 49 healthy donors to mount an effective functional response against these two major subtypes of ALL. RESULTS: From this cohort, we have identified a pool of donors with superior activity against multiple ALL cells. While these donors were more likely to be CMV+, we identified multiple CMVneg donors within this group. Furthermore, NK cells from these donors recognised B- and T-ALL through different activating receptors. Dividing functional NK cells into 29 unique subsets, we observed that within each individual the same NK cell subsets dominated across all ALL cells. Intriguingly, this occurred despite the ALL cells in our panel expressing different combinations of NK cell ligands. Finally, we can demonstrate that cellular therapy products derived from these superior donors significantly delayed leukaemia progression in preclinical models of ALL. CONCLUSIONS: We have identified a pool of superior donors that are effective against a range of ALL cells, representing a potential pool of donors that can be used as an adoptive NK cell therapy to treat paediatric ALL.

11.
Epigenetics ; 15(1-2): 134-144, 2020.
Article in English | MEDLINE | ID: mdl-31423932

ABSTRACT

Activation of naïve CD8+ T cells stimulates proliferation and differentiation into cytotoxic T-lymphocytes (CTLs). Adoptive T Cell Therapy (ACT) involves multiple rounds of ex vivo activation to generate enough CTLs for reinfusion into patients, but this drives differentiation into terminal effector T cells. Less differentiated CTL populations, such as stem cell memory T cells, are more ideal candidates for ACT because of increased self-renewal and persistent properties. Ex vivo targeting of T cell differentiation with epigenetic modifiers is a potential strategy to improve cytotoxic T-lymphocyte (CTL) generation for ACT. We established a pipeline to assess the effects of epigenetic modifiers on CD8+ T cell proliferation, differentiation, and efficacy in a preclinical melanoma model. Single treatment with epigenetic modifiers inhibited T cell proliferation in vitro, producing CD44hiCD62Lhi effector-like T cells rather than a stem cell memory T cell phenotype. Most epigenetic modifying agents had no significant effect on ACT efficacy with the notable exception of the bromodomain and extraterminal (BET)-inhibitor JQ1 which was associated with a decrease in efficacy compared to unmodified T cells. These findings reveal the complexity of epigenetic targeting of T cell differentiation, highlighting the need to precisely define the epigenetic targeting strategies to improve CTL generation for ACT.


Subject(s)
Cell Proliferation , Epigenesis, Genetic , Immunotherapy, Adoptive/methods , Melanoma, Experimental/therapy , T-Lymphocytes/drug effects , Animals , Azepines/pharmacology , Benzodiazepines/pharmacology , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Indolizines/pharmacology , Mice , Mice, Inbred C57BL , Sulfones/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Triazoles/pharmacology
12.
Oncoimmunology ; 8(8): 1609874, 2019.
Article in English | MEDLINE | ID: mdl-31413920

ABSTRACT

Immunotherapies such as adoptive cell therapy (ACT) are promising treatments for solid cancers. However, relapsing disease remains a problem and the molecular mechanisms underlying resistance are poorly defined. We postulated that the deregulated epigenetic landscape in cancer cells could underpin the acquisition of resistance to immunotherapy. To address this question, two preclinical models of ACT were employed to study transcriptional and epigenetic regulatory processes within ACT-treated cancer cells. In these models ACT consistently causes robust tumor regression, but resistance develops and tumors relapse. We identified down-regulated expression of immunogenic antigens at the mRNA level correlated with escape from immune control. To determine whether this down-regulation was under epigenetic control, we treated escaped tumor cells with DNA demethylating agents, azacytidine (AZA) and decitabine (DEC). AZA or DEC treatment restored antigen expression in a proportion of the tumor population. To explore the importance of other epigenetic modifications we isolated tumor cells refractory to DNA demethylation and screened clones against a panel of 19 different epigenetic modifying agents (EMAs). The library of EMAs included inhibitors of a range of chromosomal and transcription regulatory protein complexes, however, when tested as single agents none restored further antigen expression. These findings suggest that tumor cells employ multiple epigenetic and genetic mechanisms to evade immune control, and a combinatorial approach employing several EMAs targeting transcription and genome stability may be required to overcome tumor resistance to immunotherapy.

14.
Oncogene ; 38(1): 140-150, 2019 01.
Article in English | MEDLINE | ID: mdl-30076412

ABSTRACT

Overexpression of MYC oncogene is highly prevalent in many malignancies such as aggressive triple-negative breast cancers (TNBCs) and it is associated with very poor outcome. Despite decades of research, attempts to effectively inhibit MYC, particularly with small molecules, still remain challenging due to the featureless nature of its protein structure. Herein, we describe the engineering of the dominant-negative MYC peptide (OmoMYC) linked to a functional penetrating 'Phylomer' peptide (FPPa) as a therapeutic strategy to inhibit MYC in TNBC. We found FPPa-OmoMYC to be a potent inducer of apoptosis (with IC50 from 1-2 µM) in TNBC cells with negligible effects in non-tumorigenic cells. Transcriptome analysis of FPPa-OmoMYC-treated cells indicated that the fusion protein inhibited MYC-dependent networks, inducing dynamic changes in transcriptional, metabolic, and apoptotic processes. We demonstrated the efficacy of FPPa-OmoMYC in inhibiting breast cancer growth when injected orthotopically in TNBC allografts. Lastly, we identified strong pharmacological synergisms between FPPa-OmoMYC and chemotherapeutic agents. This study highlights a novel therapeutic approach to target highly aggressive and chemoresistant MYC-activated cancers.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Molecular Targeted Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Female , Genes, myc , Humans , Inhibitory Concentration 50 , Leucine Zippers/genetics , Mice , Models, Molecular , Mutation , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Peptide Library , Protein Conformation , Protein Engineering , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics
15.
Front Immunol ; 9: 2990, 2018.
Article in English | MEDLINE | ID: mdl-30700986

ABSTRACT

Conventional dendritic cells (cDC) resident in the lymphoid organs of mice have been classically divided into CD8+ and CD8neg subsets. It is well-established that CD8+ dendritic cells (DCs) and their migratory counterparts in the periphery comprise the cross-presenting cDC1 subset. In contrast, CD8neg DCs are grouped together in the heterogeneous cDC2 subset. CD8neg DCs are relatively poor cross-presenters and drive more prominent CD4+ T cell responses against exogenous antigens. The discovery of the X-C motif chemokine receptor 1 (XCR1) as a specific marker of cross-presenting DCs, has led to the identification of a divergent subset of CD8+ DCs that lacks the ability to cross-present. Here, we report that these poorly characterized CD8+XCR1neg DCs have a gene expression profile that is consistent with both plasmacytoid DCs (pDCs) and cDC2. Our data demonstrate that CD8+XCR1neg DCs possess a unique pattern of endocytic receptors and a restricted toll-like receptor (TLR) profile that is particularly enriched for TLR5, giving them a unique position within the DC immunosurveillance network.


Subject(s)
Cross-Priming , Dendritic Cells/metabolism , Toll-Like Receptor 5/metabolism , Animals , CD8 Antigens/metabolism , Cell Separation , Dendritic Cells/immunology , Endocytosis/immunology , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Receptors, Chemokine/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Toll-Like Receptor 5/immunology
16.
J Hosp Med ; 11(4): 289-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26797978

ABSTRACT

Low levels of activity in hospital inpatients contribute to functional decline. Previous studies have shown low levels of activity in older inpatients, but few have investigated younger inpatients (aged <65 years). This observational study measured activity in older (aged ≥65 years) and younger hospital inpatients on 3 wards (medical, surgical, oncology) in a major teaching hospital in Brisbane, Australia, as part of a quality-improvement intervention to enhance mobility. Using structured behavioral mapping protocols, participants were observed for 2-minute intervals throughout 4, 4-hour daytime observation periods. The proportion of time spent at different activity levels was calculated for each participant, and time spent standing, walking or wheeling was compared between age group and wards. There were 3272 observations collected on 132 participants (median, 30 per patient; range, 9-35). The most time was spent lying in bed (mean 57%), with 9% standing or walking. There were significant differences among wards, but no difference between older and younger subgroups. Low mobility is common in adult inpatients of all ages. Behavioral mapping provided measures suitable for use in quality improvement.


Subject(s)
Activities of Daily Living , Hospitalization , Mobility Limitation , Walking , Activities of Daily Living/psychology , Adult , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Walking/physiology , Walking/psychology
17.
Cell Mol Immunol ; 13(1): 119-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25640655

ABSTRACT

Complement receptor 2 (CR2/CD21) is predominantly expressed on the surface of mature B cells where it forms part of a coreceptor complex that functions, in part, to modulate B-cell receptor signal strength. CR2/CD21 expression is tightly regulated throughout B-cell development such that CR2/CD21 cannot be detected on pre-B or terminally differentiated plasma cells. CR2/CD21 expression is upregulated at B-cell maturation and can be induced by IL-4 and CD40 signaling pathways. We have previously characterized elements in the proximal promoter and first intron of CR2/CD21 that are involved in regulating basal and tissue-specific expression. We now extend these analyses to the CR2/CD21 core promoter. We show that in mature B cells, CR2/CD21 transcription proceeds from a focused TSS regulated by a non-consensus TATA box, an initiator element and a downstream promoter element. Furthermore, occupancy of the general transcriptional machinery in pre-B versus mature B-cell lines correlate with CR2/CD21 expression level and indicate that promoter accessibility must switch from inactive to active during the transitional B-cell window.


Subject(s)
CD40 Antigens/metabolism , Interleukin-4/metabolism , Precursor Cells, B-Lymphoid/metabolism , Promoter Regions, Genetic , Receptors, Complement 3d/metabolism , Transcription Initiation Site , Base Sequence , CD40 Antigens/genetics , CD40 Antigens/immunology , Cell Differentiation , Cell Line, Tumor , Exons , Gene Expression Regulation , Humans , Interleukin-4/genetics , Interleukin-4/immunology , Introns , K562 Cells , Molecular Sequence Data , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Receptors, Complement 3d/genetics , Receptors, Complement 3d/immunology , Signal Transduction , Transcription, Genetic
18.
Int J Biochem Cell Biol ; 64: 107-19, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25817480

ABSTRACT

Complement receptor 2 (CR2/CD21) plays an important role in the generation of normal B cell immune responses. As transcription appears to be the prime mechanism via which surface CR2/CD21 expression is controlled, understanding transcriptional regulation of this gene will have broader implications to B cell biology. Here we report opposing, cell-context specific control of CR2/CD21 promoter activity by tandem E-box elements, spaced 22 bp apart and within 70 bp of the transcription initiation site. We have identified E2A and USF transcription factors as binding to the distal and proximal E-box sites respectively in CR2-positive B-cells, at a site that is hypersensitive to restriction enzyme digestion compared to non-expressing K562 cells. However, additional unidentified proteins have also been found to bind these functionally important elements. By utilizing a proteomics approach we have identified a repressor protein, RP58, binding the distal E-box motif. Co-transfection experiments using RP58 overexpression constructs demonstrated a specific 10-fold repression of CR2/CD21 transcriptional activity mediated through the distal E-box repressor element. Taken together, our results indicate that repression of the CR2/CD21 promoter can occur through one of the E-box motifs via recruitment of RP58 and other factors to bring about a silenced chromatin context within CR2/CD21 non-expressing cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Receptors, Complement 3d/genetics , Repressor Proteins/physiology , Upstream Stimulatory Factors/metabolism , Base Sequence , Chromatin/physiology , E-Box Elements , Epigenesis, Genetic , Humans , K562 Cells , Molecular Sequence Data , Organ Specificity , Promoter Regions, Genetic , Receptors, Complement 3d/metabolism
19.
Am J Med Qual ; 30(1): 5-13, 2015.
Article in English | MEDLINE | ID: mdl-24270172

ABSTRACT

High-quality, efficient health care for older patients is a priority for health care systems. Acute Care for Elders units improve outcomes but there is a need for generalizable models of care that adopt the principles pioneered in these units. This report describes Eat Walk Engage, a collaborative care model on a general medical ward in Brisbane, Australia. The model focused on early mobilization, feeding assistance, and cognitive stimulation. Using the Promoting Action on Research Implementation in Health Services implementation framework, the facilitation team enabled the clinical team to recognize barriers and develop solutions. Challenges included unclear responsibility, workload concerns, and risk aversion. Implementation strategies included engaging champions, education, audit and feedback, task delineation and delegation, improving physical resources, and workforce redesign. During the first 18 months, audits showed improved nursing documentation in targeted domains and improved performance of mobilizing and cognitive strategies; length of stay for older inpatients fell by 3 days on the intervention ward.


Subject(s)
Cooperative Behavior , Geriatrics , Hospital Administration , Quality Improvement/organization & administration , Aged , Australia , Cognition , Feedback , Hospitalization , Humans , Inservice Training/organization & administration , Leadership , Nutrition Policy , Organizational Culture , Patient Care Team/organization & administration , Quality of Health Care/organization & administration , Recovery of Function , Workload
20.
Eur J Hum Genet ; 22(12): 1376-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24713664

ABSTRACT

The phenomenon of X chromosome inactivation in female mammals is well characterised and remains the archetypal example of dosage compensation via monoallelic expression. The temporal series of events that culminates in inactive X-specific gene silencing by DNA methylation has revealed a 'patchwork' of gene inactivation along the chromosome, with approximately 15% of genes escaping. Such genes are therefore potentially subject to sex-specific imbalance between males and females. Aside from XIST, the non-coding RNA on the X chromosome destined to be inactivated, very little is known about the extent of loci that may be selectively silenced on the active X chromosome (Xa). Using longitudinal array-based DNA methylation profiling of two human tissues, we have identified specific and widespread active X-specific DNA methylation showing stability over time and across tissues of disparate origin. Our panel of X-chromosome loci subject to methylation on Xa reflects a potentially novel mechanism for controlling female-specific X inactivation and sex-specific dimorphisms in humans. Further work is needed to investigate these phenomena.


Subject(s)
Chromosomes, Human, X/genetics , DNA Methylation , X Chromosome Inactivation/genetics , Databases, Genetic , Dosage Compensation, Genetic , Female , Gene Expression Regulation , Gene Silencing , Genetic Loci , Humans , Male , RNA, Untranslated/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...