Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Subst Use Addctn J ; : 29767342241249870, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38804580

ABSTRACT

BACKGROUND: For structurally marginalized populations, including people who use drugs (PWUD), equitable access to healthcare can be achieved through healthcare access supports. However, few studies characterized utilization of formal (eg, outreach workers, healthcare professionals) and informal (eg, friends/family) supports. Therefore, we sought to estimate the prevalence of and factors associated with receiving each type of support among PWUD. METHODS: We used data from 2 prospective cohort studies of PWUD in Vancouver, Canada, in 2017 to 2020. We constructed separate multivariable generalized linear mixed-effects models to identify factors associated with receiving each of the 3 types of supports (ie, healthcare professionals, outreach workers/peer navigators, and informal supports) compared to no supports. RESULTS: Of 996 participants, 350 (35.1%) reported receiving supports in the past 6 months at baseline, through informal supports (6.2%), outreach workers (14.1%), and healthcare professionals (20.9%). In multivariable analyses, HIV positivity, chronic pain, and avoiding healthcare due to the past mistreatment were positively associated with receiving supports from each of healthcare professionals and outreach workers. Men were less likely to receive any types of the supports (all P < .05). CONCLUSIONS: Utilization of healthcare access supports was relatively low in this sample. However, formal supports appeared to have reached PWUD exhibiting more comorbidities and experiencing discrimination in healthcare. Further efforts to make formal supports more available would benefit PWUD with unmet healthcare needs, particularly men.

2.
Nat Cell Biol ; 20(6): 710-720, 2018 06.
Article in English | MEDLINE | ID: mdl-29802403

ABSTRACT

Elucidation of the identity and diversity of mechanisms that sustain long-term human blood cell production remains an important challenge. Previous studies indicate that, in adult mice, this property is vested in cells identified uniquely by their ability to clonally regenerate detectable, albeit highly variable levels and types, of mature blood cells in serially transplanted recipients. From a multi-parameter analysis of the molecular features of very primitive human cord blood cells that display long-term cell outputs in vitro and in immunodeficient mice, we identified a prospectively separable CD33+CD34+CD38-CD45RA-CD90+CD49f+ phenotype with serially transplantable, but diverse, cell output profiles. Single-cell measurements of the mitogenic response, and the transcriptional, DNA methylation and 40-protein content of this and closely related phenotypes revealed subtle but consistent differences both within and between each subset. These results suggest that multiple regulatory mechanisms combine to maintain different cell output activities of human blood cell precursors with high regenerative potential.


Subject(s)
Cell Proliferation , Cell Separation/methods , Fetal Blood/cytology , Mitosis , Sialic Acid Binding Ig-like Lectin 3/metabolism , Single-Cell Analysis/methods , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cord Blood Stem Cell Transplantation , DNA Methylation , Female , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genotype , Humans , Male , Mice, Transgenic , Phenotype , Time Factors , Transcriptome
3.
Stem Cell Reports ; 8(1): 152-162, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076756

ABSTRACT

The role of growth factors (GFs) in controlling the biology of human hematopoietic stem cells (HSCs) remains limited by a lack of information concerning the individual and combined effects of GFs directly on the survival, Mitogenesis, and regenerative activity of highly purified human HSCs. We show that the initial input HSC activity of such a purified starting population of human cord blood cells can be fully maintained over a 21-day period in serum-free medium containing five GFs alone. HSC survival was partially supported by any one of these GFs, but none were essential, and different combinations of GFs variably stimulated HSC proliferation. However, serial transplantability was not detectably compromised by many conditions that reduced human HSC proliferation and/or survival. These results demonstrate the dissociated control of these three human HSC bio-responses, and set the stage for future improvements in strategies to modify and expand human HSCs ex vivo.


Subject(s)
Cell Differentiation , Cell Proliferation , Cell Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , In Vitro Techniques , Integrin alpha6/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Phenotype
4.
Exp Hematol ; 48: 41-49, 2017 04.
Article in English | MEDLINE | ID: mdl-28087429

ABSTRACT

Xenograft models are transforming our understanding of the output capabilities of primitive human hematopoietic cells in vivo. However, many variables that affect posttransplantation reconstitution dynamics remain poorly understood. Here, we show that an equivalent level of human chimerism can be regenerated from human CD34+ cord blood cells transplanted intravenously either with or without additional radiation-inactivated cells into 2- to 6-month-old NOD-Rag1-/--IL2Rγc-/- (NRG) mice given a more radioprotective conditioning regimen than is possible in conventionally used, repair-deficient NOD-Prkdcscid/scid-IL2Rγc-/- (NSG) hosts. Comparison of sublethally irradiated and non-irradiated NRG mice and W41/W41 derivatives showed superior chimerism in the W41-deficient recipients, with some differential effects on different lineage outputs. Consistently superior outputs were observed in female recipients regardless of their genotype, age, or pretransplantation conditioning, with greater differences apparent later after transplantation. These results define key parameters for optimizing the sensitivity and minimizing the intraexperimental variability of human hematopoietic xenografts generated in increasingly supportive immunodeficient host mice.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins c-kit/genetics , Age Factors , Animals , Biomarkers , Cell Count , Female , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Sex Factors , Transplantation Chimera , Transplantation, Heterologous
5.
Biol Blood Marrow Transplant ; 22(11): 1945-1952, 2016 11.
Article in English | MEDLINE | ID: mdl-27496214

ABSTRACT

Plerixafor (P) together with granulocyte colony-stimulating factor (G) is now recognized as an important strategy for mobilizing hematopoietic cells for use in patients given myelosuppressive therapies. However, quantitative comparisons of their ability to mobilize human cells with different hematopoietic activities in vitro or in vivo (in immunodeficient mice) and their interrelationships have not been investigated. To address these questions, we collected samples from 5 normal adult volunteers before and after administering P alone and from another 5 before and after a 4-day course of G and again after a subsequent injection of P. Measurements of their blood content of CD34+ cells, in vitro myeloid colony-forming cells, 3- and 6-week long-term culture (LTC) cell outputs, and levels of circulating human platelets, as well as myeloid and lymphoid cells obtained in immunodeficient mice that received transplants, showed all activities were maximal 4 hours after P preceded by G, and 3-week LTC outputs showed the highest concordance with the 3-week circulating human neutrophil levels obtained in mice that received transplants. Thus, human cells capable of producing neutrophils rapidly in vivo were optimally mobilized by the G + P protocol, and the 3-week LTC assay appears to offer a more specific predictor of their levels than conventional CD34+ cell or colony-forming cell counts.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/administration & dosage , Animals , Antigens, CD34/blood , Antigens, CD34/drug effects , Benzylamines , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Count , Cell Culture Techniques , Cyclams , Heterografts , Humans , Mice , Mice, SCID , Neutrophils/cytology , Neutrophils/drug effects
6.
Exp Hematol ; 44(7): 635-40, 2016 07.
Article in English | MEDLINE | ID: mdl-27090409

ABSTRACT

The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.


Subject(s)
Blood Platelets/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myelopoiesis , Neutrophils/metabolism , Thrombopoiesis , Animals , Cell Differentiation/drug effects , Cells, Cultured , Graft Survival , Hematopoietic Cell Growth Factors/metabolism , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Myelopoiesis/drug effects , Thrombopoiesis/drug effects
7.
Blood ; 125(3): 504-15, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25370416

ABSTRACT

Without effective therapy, chronic-phase chronic myeloid leukemia (CP-CML) evolves into an acute leukemia (blast crisis [BC]) that displays either myeloid or B-lymphoid characteristics. This transition is often preceded by a clinically recognized, but biologically poorly characterized, accelerated phase (AP). Here, we report that IKAROS protein is absent or reduced in bone marrow blasts from most CML patients with advanced myeloid disease (AP or BC). This contrasts with primitive CP-CML cells and BCR-ABL1-negative acute myeloid leukemia blasts, which express readily detectable IKAROS. To investigate whether loss of IKAROS contributes to myeloid disease progression in CP-CML, we examined the effects of forced expression of a dominant-negative isoform of IKAROS (IK6) in CP-CML patients' CD34(+) cells. We confirmed that IK6 disrupts IKAROS activity in transduced CP-CML cells and showed that it confers on them features of AP-CML, including a prolonged increased output in vitro and in xenografted mice of primitive cells with an enhanced ability to differentiate into basophils. Expression of IK6 in CD34(+) CP-CML cells also led to activation of signal transducer and activator of transcription 5 and transcriptional repression of its negative regulators. These findings implicate loss of IKAROS as a frequent step and potential diagnostic harbinger of progressive myeloid disease in CML patients.


Subject(s)
Basophils/pathology , Cell Differentiation/drug effects , Eosinophils/pathology , Ikaros Transcription Factor/antagonists & inhibitors , Leukemia, Myeloid, Chronic-Phase/pathology , STAT5 Transcription Factor/metabolism , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Basophils/drug effects , Basophils/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Disease Progression , Eosinophils/drug effects , Eosinophils/metabolism , Flow Cytometry , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Immunoenzyme Techniques , Leukemia, Myeloid, Chronic-Phase/genetics , Leukemia, Myeloid, Chronic-Phase/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics
8.
Stem Cell Reports ; 3(5): 841-57, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418728

ABSTRACT

Disrupted IKAROS activity is a recurrent feature of some human leukemias, but effects on normal human hematopoietic cells are largely unknown. Here, we used lentivirally mediated expression of a dominant-negative isoform of IKAROS (IK6) to block normal IKAROS activity in primitive human cord blood cells and their progeny. This produced a marked (10-fold) increase in serially transplantable multipotent IK6(+) cells as well as increased outputs of normally differentiating B cells and granulocytes in transplanted immunodeficient mice, without producing leukemia. Accompanying T/natural killer (NK) cell outputs were unaltered, and erythroid and platelet production was reduced. Mechanistically, IK6 specifically increased human granulopoietic progenitor sensitivity to two growth factors and activated CREB and its targets (c-FOS and Cyclin B1). In more primitive human cells, IK6 prematurely initiated a B cell transcriptional program without affecting the hematopoietic stem cell-associated gene expression profile. Some of these effects were species specific, thus identifying novel roles of IKAROS in regulating normal human hematopoietic cells.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Ikaros Transcription Factor/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blotting, Western , Cell Differentiation , Cells, Cultured , Fetal Blood/cytology , Gene Expression Profiling , Granulocytes/cytology , Granulocytes/metabolism , HeLa Cells , Hematopoietic Stem Cell Transplantation/methods , Humans , Ikaros Transcription Factor/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Confocal , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transplantation, Heterologous
9.
Blood ; 121(5): e1-4, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23233660

ABSTRACT

UNLABELLED: Better methods to characterize normal human hematopoietic cells with short-term repopulating activity cells (STRCs) are needed to facilitate improving recovery rates in transplanted patients.We now show that 5-fold more human myeloid cells are produced in sublethally irradiated NOD/SCID-IL-2Receptor-γchain-null (NSG) mice engineered to constitutively produce human interleukin-3, granulocyte-macrophage colony-stimulating factor and Steel factor (NSG-3GS mice) than in regular NSG mice 3 weeks after an intravenous injection of CD34 human cord blood cells. Importantly, the NSG-3GS mice also show a concomitant and matched increase in circulating mature human neutrophils. Imaging NSG-3GS recipients of lenti-luciferase-transduced cells showed that human cells being produced 3 weeks posttransplant were heterogeneously distributed, validating the blood as a more representative measure of transplanted STRC activity. Limiting dilution transplants further demonstrated that the early increase in human granulopoiesis in NSG-3GS mice reflects an expanded output of differentiated cells per STRC rather than an increase in STRC detection. KEY POINTS: NSG-3GS mice support enhanced clonal outputs from human short-term repopulating cells (STRCs) without affecting their engrafting efficiency. Increased human STRC clone sizes enable their more precise and efficient measurement by peripheral blood monitoring.


Subject(s)
Cord Blood Stem Cell Transplantation , Graft Survival , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-3/biosynthesis , Myelopoiesis , Neutrophils/metabolism , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-3/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neutrophils/cytology , Time Factors , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...