Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Blood ; 143(3): 272-278, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37879046

ABSTRACT

ABSTRACT: Megakaryocytes (MKs) generate thousands of platelets over their lifespan. The roles of platelets in infection and inflammation has guided an interest to the study of extramedullary thrombopoiesis and therefore MKs have been increasingly reported within the spleen and lung. However, the relative abundance of MKs in these organs compared to the bone marrow and the scale of their contribution to the platelet pool in a steady state remain controversial. We investigated the relative abundance of MKs in the adult murine bone marrow, spleen, and lung using whole-mount light-sheet and quantitative histological imaging, flow cytometry, intravital imaging, and an assessment of single-cell RNA sequencing (scRNA-seq) repositories. Flow cytometry revealed significantly higher numbers of hematopoietic stem and progenitor cells and MKs in the murine bone marrow than in spleens or perfused lungs. Two-photon intravital and light-sheet microscopy, as well as quantitative histological imaging, confirmed these findings. Moreover, ex vivo cultured MKs from the bone marrow subjected to static or microfluidic platelet production assays had a higher capacity for proplatelet formation than MKs from other organs. Analysis of previously published murine and human scRNA-seq data sets revealed that only a marginal fraction of MK-like cells can be found within the lung and most likely only marginally contribute to platelet production in the steady state.


Subject(s)
Bone Marrow , Thrombopoiesis , Mice , Humans , Animals , Thrombopoiesis/genetics , Blood Platelets , Megakaryocytes , Spleen
2.
Curr Opin Hematol ; 31(1): 1-5, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37910197

ABSTRACT

PURPOSE OF REVIEW: Single-cell RNA sequencing studies have revealed transcriptional heterogeneity within the megakaryocytic lineage and the identified unique subsets. In this review, we discuss the functional and phenotypic plasticity of these subpopulations as well as the impacts on health and disease. RECENT FINDINGS: Megakaryocytes (MKs) can be transcriptionally categorized into platelet generating, niche supporting, immune, and cycling cells, which are distinguished by their unique gene expression patterns and cellular markers. Additionally, a significant population of these cells has been established to reside in the nonhematopoietic tissues and they display enhanced immune-related characteristics. Combined with the location in which the megakaryocytes exist, these cells can play unique roles dictated by their current environment and biological needs, including responding to changes in pathogen exposure. SUMMARY: Advances in megakaryocyte research has elucidated the existence of multiple subpopulations of MKs that serve different functions. These subpopulations implicate a greater potential for MKs to be regulators of health and suggest new avenues for treatments and therapies in related diseases.


Subject(s)
Blood Platelets , Megakaryocytes , Humans , Megakaryocytes/metabolism , Blood Platelets/metabolism
4.
Front Immunol ; 12: 772408, 2021.
Article in English | MEDLINE | ID: mdl-34804065

ABSTRACT

The immune microenvironment is a critical driver and regulator of leukemic progression and hematological disease. Recent investigations have demonstrated that multiple immune components play a central role in regulating hematopoiesis, and dysfunction at the immune cell level significantly contributes to neoplastic disease. Immune cells are acutely sensitive to remodeling by leukemic inflammatory cytokine exposure. Importantly, immune cells are the principal cytokine producers in the hematopoietic system, representing an untapped frontier for clinical interventions. Due to a proinflammatory cytokine environment, dysregulation of immune cell states is a hallmark of hematological disease and neoplasia. Malignant immune adaptations have profound effects on leukemic blast proliferation, disease propagation, and drug-resistance. Conversely, targeting the immune landscape to restore hematopoietic function and limit leukemic expansion may have significant therapeutic value. Despite the fundamental role of the immune microenvironment during the initiation, progression, and treatment response of hematological disease, a detailed examination of how leukemic cytokines alter immune cells to permit, promote, or inhibit leukemia growth is lacking. Here we outline an immune-based model of leukemic transformation and highlight how the profound effect of immune alterations on the trajectory of malignancy. The focus of this review is to summarize current knowledge about the impacts of pro- and anti-inflammatory cytokines on immune cells subsets, their modes of action, and immunotherapeutic approaches with the potential to improve clinical outcomes for patients suffering from hematological myeloid malignancies.


Subject(s)
Cytokines/immunology , Hematologic Neoplasms/immunology , Myeloproliferative Disorders/immunology , Animals , Humans , Immunity, Innate , Signal Transduction
5.
Leukemia ; 35(12): 3371-3382, 2021 12.
Article in English | MEDLINE | ID: mdl-34120146

ABSTRACT

Leukemic stem cells (LSCs) can acquire non-mutational resistance following drug treatment leading to therapeutic failure and relapse. However, oncogene-independent mechanisms of drug persistence in LSCs are incompletely understood, which is the primary focus of this study. We integrated proteomics, transcriptomics, and metabolomics to determine the contribution of STAT3 in promoting metabolic changes in tyrosine kinase inhibitor (TKI) persistent chronic myeloid leukemia (CML) cells. Proteomic and transcriptional differences in TKI persistent CML cells revealed BCR-ABL-independent STAT3 activation in these cells. While knockout of STAT3 inhibited the CML cells from developing drug-persistence, inhibition of STAT3 using a small molecule inhibitor sensitized the persistent CML cells to TKI treatment. Interestingly, given the role of phosphorylated STAT3 as a transcription factor, it localized uniquely to genes regulating metabolic pathways in the TKI-persistent CML stem and progenitor cells. Subsequently, we observed that STAT3 dysregulated mitochondrial metabolism forcing the TKI-persistent CML cells to depend on glycolysis, unlike TKI-sensitive CML cells, which are more reliant on oxidative phosphorylation. Finally, targeting pyruvate kinase M2, a rate-limiting glycolytic enzyme, specifically eradicated the TKI-persistent CML cells. By exploring the role of STAT3 in altering metabolism, we provide critical insight into identifying potential therapeutic targets for eliminating TKI-persistent LSCs.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Metabolome , Neoplastic Stem Cells/drug effects , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/pharmacology , Transcriptome , Animals , Apoptosis , Female , Glycolysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/genetics
6.
JCI Insight ; 5(22)2020 11 19.
Article in English | MEDLINE | ID: mdl-33208555

ABSTRACT

The nonimmune roles of Tregs have been described in various tissues, including the BM. In this study, we comprehensively phenotyped marrow Tregs, elucidating their key features and tissue-specific functions. We show that marrow Tregs are migratory and home back to the marrow. For trafficking, marrow Tregs use S1P gradients, and disruption of this axis allows for specific targeting of the marrow Treg pool. Following Treg depletion, the function and phenotype of both mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) was impaired. Transplantation also revealed that a Treg-depleted niche has a reduced capacity to support hematopoiesis. Finally, we found that marrow Tregs are high producers of IL-10 and that Treg-secreted IL-10 has direct effects on MSC function. This is the first report to our knowledge revealing that Treg-secreted IL-10 is necessary for stromal cell maintenance, and our work outlines an alternative mechanism by which this cytokine regulates hematopoiesis.


Subject(s)
Bone Marrow Cells/physiology , Hematopoiesis , Hematopoietic Stem Cells/physiology , Interleukin-10/metabolism , Mesenchymal Stem Cells/physiology , Stromal Cells/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Communication , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , Stromal Cells/cytology , Stromal Cells/immunology
7.
Immunity ; 52(6): 1105-1118.e9, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553173

ABSTRACT

The challenges in recapitulating in vivo human T cell development in laboratory models have posed a barrier to understanding human thymopoiesis. Here, we used single-cell RNA sequencing (sRNA-seq) to interrogate the rare CD34+ progenitor and the more differentiated CD34- fractions in the human postnatal thymus. CD34+ thymic progenitors were comprised of a spectrum of specification and commitment states characterized by multilineage priming followed by gradual T cell commitment. The earliest progenitors in the differentiation trajectory were CD7- and expressed a stem-cell-like transcriptional profile, but had also initiated T cell priming. Clustering analysis identified a CD34+ subpopulation primed for the plasmacytoid dendritic lineage, suggesting an intrathymic dendritic specification pathway. CD2 expression defined T cell commitment stages where loss of B cell potential preceded that of myeloid potential. These datasets delineate gene expression profiles spanning key differentiation events in human thymopoiesis and provide a resource for the further study of human T cell development.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Lymphopoiesis/genetics , T-Lymphocytes/metabolism , Thymocytes/metabolism , Animals , Biomarkers , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing/methods , Humans , Immunophenotyping , Mice , Single-Cell Analysis , T-Lymphocytes/cytology , Thymocytes/cytology , Transcriptome
8.
Cell Rep ; 28(2): 302-311.e5, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291568

ABSTRACT

The bone marrow microenvironment is composed of heterogeneous cell populations of non-hematopoietic cells with complex phenotypes and undefined trajectories of maturation. Among them, mesenchymal cells maintain the production of stromal, bone, fat, and cartilage cells. Resolving these unique cellular subsets within the bone marrow remains challenging. Here, we used single-cell RNA sequencing of non-hematopoietic bone marrow cells to define specific subpopulations. Furthermore, by combining computational prediction of the cell state hierarchy with the known expression of key transcription factors, we mapped differentiation paths to the osteocyte, chondrocyte, and adipocyte lineages. Finally, we validated our findings using lineage-specific reporter strains and targeted knockdowns. Our analysis reveals differentiation hierarchies for maturing stromal cells, determines key transcription factors along these trajectories, and provides an understanding of the complexity of the bone marrow microenvironment.


Subject(s)
Bone Marrow/metabolism , Stem Cell Niche/physiology , Cell Differentiation , Humans
9.
J Allergy Clin Immunol ; 142(6): 1894-1908.e7, 2018 12.
Article in English | MEDLINE | ID: mdl-29470999

ABSTRACT

BACKGROUND: Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown. OBJECTIVE: The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation. METHODS: Isolated dendritic cells and bone marrow-derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC-/-, MHC class II-/-, Wiskott-Aldrich syndrome protein (WASP)-/-, 5C.C7 recombination-activating gene 2 (Rag2)-/-, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection. RESULTS: Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes. CONCLUSIONS: Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Mast Cells/drug effects , NAD/pharmacology , Adult , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Line , Humans , Listeria monocytogenes , Listeriosis/drug therapy , Listeriosis/immunology , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Transgenic , NAD/therapeutic use
10.
Nature ; 551(7682): 590-595, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29168504

ABSTRACT

Small, approximately 10-kilobase microhomology-mediated tandem duplications are abundant in the genomes of BRCA1-linked but not BRCA2-linked breast cancer. Here we define the mechanism underlying this rearrangement signature. We show that, in primary mammalian cells, BRCA1, but not BRCA2, suppresses the formation of tandem duplications at a site-specific chromosomal replication fork barrier imposed by the binding of Tus proteins to an array of Ter sites. BRCA1 has no equivalent role at chromosomal double-stranded DNA breaks, indicating that tandem duplications form specifically at stalled forks. Tandem duplications in BRCA1 mutant cells arise by a replication restart-bypass mechanism terminated by end joining or by microhomology-mediated template switching, the latter forming complex tandem duplication breakpoints. Solitary DNA ends form directly at Tus-Ter, implicating misrepair of these lesions in tandem duplication formation. Furthermore, BRCA1 inactivation is strongly associated with ~10 kilobase tandem duplications in ovarian cancer. This tandem duplicator phenotype may be a general signature of BRCA1-deficient cancer.


Subject(s)
DNA End-Joining Repair/genetics , DNA Replication/genetics , Tandem Repeat Sequences/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Animals , BRCA1 Protein , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells , Female , Genes, Reporter , Homologous Recombination , Humans , Mice , Ovarian Neoplasms/genetics , Sequence Deletion , Tumor Suppressor Proteins/metabolism
11.
Methods Mol Biol ; 1660: 175-190, 2017.
Article in English | MEDLINE | ID: mdl-28828656

ABSTRACT

Here, we describe a comprehensive methodology for the setup and standardization of EV analysis using nanoscale flow cytometry. Controls of different size ranges, fluorescent intensities, and materials can be used to set up distribution curves that are then used for instrument optimization and as a reference guide. Using these controls, flow cytometry instruments can be primed for the detection, analysis, and sorting of specific EV populations. This allows for cross platform comparison and the ability to monitor both quality control (QC) and quality assurance (QA). The method here describes the use of nanoparticles to optimize a flow cytometer for small particle detection. It also outlines the procedures necessary to recover EVs for downstream applications.


Subject(s)
Extracellular Vesicles/metabolism , Flow Cytometry , Flow Cytometry/methods , Microscopy, Fluorescence , Nanoparticles/chemistry , Polystyrenes/chemistry , Reproducibility of Results , Sensitivity and Specificity , Spectrum Analysis
12.
Int J Hematol ; 105(5): 566-577, 2017 May.
Article in English | MEDLINE | ID: mdl-28176225

ABSTRACT

Leukemias depend on transformed stem cells for their growth and thus these cells represent important therapeutic targets. However, leukemic stem cells resemble normal hematopoietic stem cells (HSCs) with respect to most surface markers, gene expression patterns, and ability to be transplanted. Furthermore, the microenvironment that supports healthy HSCs non-hematopoietic populations, and immune cells correspondingly, the cytokines, adhesion molecules and signal transduction pathways are also impaired during leukemogenesis. This altered environment promotes leukemic growth specifically through pro-inflammatory cytokines. Here, we characterize normal and leukemic signaling, as well as the instructive cues from the neighboring hematopoietic cells and the microenvironment that promote stem cell self-renewal and differentiation.


Subject(s)
Cytokines/physiology , Leukemia/pathology , Neoplastic Stem Cells/pathology , Signal Transduction/physiology , Tumor Microenvironment , Cell Adhesion Molecules/physiology , Cell Transformation, Neoplastic/genetics , Hematopoietic Stem Cells , Tumor Microenvironment/genetics
13.
Front Cell Dev Biol ; 4: 117, 2016.
Article in English | MEDLINE | ID: mdl-27833910

ABSTRACT

A key function of human eosinophils is to secrete cytokines, chemokines and cationic proteins, trafficking, and releasing these mediators for roles in inflammation and other immune responses. Eosinophil activation leads to secretion of pre-synthesized granule-stored mediators through different mechanisms, but the ability of eosinophils to secrete extracellular vesicles (EVs), very small vesicles with preserved membrane topology, is still poorly understood. In the present work, we sought to identify and characterize EVs released from human eosinophils during different conditions: after a culturing period or after isolation and stimulation with inflammatory stimuli, which are known to induce eosinophil activation and secretion: CCL11 (eotaxin-1) and tumor necrosis factor alpha (TNF-α). EV production was investigated by nanoscale flow cytometry, conventional transmission electron microscopy (TEM) and pre-embedding immunonanogold EM. The tetraspanins CD63 and CD9 were used as EV biomarkers for both flow cytometry and ultrastructural immunolabeling. Nanoscale flow cytometry showed that human eosinophils produce EVs in culture and that a population of EVs expressed detectable CD9, while CD63 was not consistently detected. When eosinophils were stimulated immediately after isolation and analyzed by TEM, EVs were clearly identified as microvesicles (MVs) outwardly budding off the plasma membrane. Both CCL11 and TNF-α induced significant increases of MVs compared to unstimulated cells. TNF-α induced amplified release of MVs more than CCL11. Eosinophil MV diameters varied from 20 to 1000 nm. Immunonanogold EM revealed clear immunolabeling for CD63 and CD9 on eosinophil MVs, although not all MVs were labeled. Altogether, we identified, for the first time, that human eosinophils secrete MVs and that this production increases in response to inflammatory stimuli. This is important to understand the complex secretory activities of eosinophils underlying immune responses. The contribution of the eosinophil-derived MVs to the regulation of immune responses awaits further investigation.

14.
Sci Rep ; 6: 22325, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928119

ABSTRACT

CD4(+) CD25(+) Foxp3(+) Tregs have been shown to play a central role in immune homeostasis while preventing from fatal inflammatory responses, while Th17 cells have traditionally been recognized as pro-inflammatory mediators implicated in a myriad of diseases. Studies have shown the potential of Tregs to convert into Th17 cells, and Th17 cells into Tregs. Increasing evidence have pointed out CD25 as a key molecule during this transdifferentiation process, however molecules that allow such development remain unknown. Here, we investigated the impact of NAD(+) on the fate of CD4(+) CD25(+) Foxp3(+) Tregs in-depth, dissected their transcriptional signature profile and explored mechanisms underlying their conversion into IL-17A producing cells. Our results demonstrate that NAD(+) promotes Treg conversion into Th17 cells in vitro and in vivo via CD25 cell surface marker. Despite the reduced number of Tregs, known to promote homeostasis, and an increased number of pro-inflammatory Th17 cells, NAD(+) was able to promote an impressive allograft survival through a robust systemic IL-10 production that was CD4(+) CD25(+) Foxp3(+) independent. Collectively, our study unravels a novel immunoregulatory mechanism of NAD(+) that regulates Tregs fate while promoting allograft survival that may have clinical applications in alloimmunity and in a wide spectrum of inflammatory conditions.


Subject(s)
Forkhead Transcription Factors/metabolism , Graft Survival/drug effects , Interleukin-10/metabolism , NAD/therapeutic use , Skin Transplantation , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Allografts/transplantation , Animals , CD4 Antigens/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Forkhead Transcription Factors/genetics , Homeostasis/drug effects , Humans , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
15.
PLoS One ; 11(1): e0144678, 2016.
Article in English | MEDLINE | ID: mdl-26745887

ABSTRACT

The identification of extracellular vesicles (EVs) as intercellular conveyors of biological information has recently emerged as a novel paradigm in signaling, leading to the exploitation of EVs and their contents as biomarkers of various diseases. However, whether there are diurnal variations in the size, number, and tissue of origin of blood EVs is currently not known, and could have significant implications when using EVs as biomarkers for disease progression. Currently available technologies for the measurement of EV size and number are either time consuming, require specialized equipment, or lack sufficient accuracy across a range of EV sizes. Flow cytometry represents an attractive alternative to these methods; however, traditional flow cytometers are only capable of measuring particles down to 500 nm, which is significantly larger than the average and median sizes of plasma EVs. Utilizing a Beckman Coulter MoFlo XDP flow cytometer with NanoView module, we employed nanoscale flow cytometry (termed nanoFCM) to examine the relative number and scatter distribution of plasma EVs at three different time points during the day in 6 healthy adults. Analysis of liposomes and plasma EVs proved that nanoFCM is capable of detecting biologically-relevant vesicles down to 100 nm in size. With this high resolution configuration, we observed variations in the relative size (FSC/SSC distributions) and concentration (proportions) of EVs in healthy adult plasma across the course of a day, suggesting that there are diurnal variations in the number and size distribution of circulating EV populations. The use of nanoFCM provides a valuable tool for the study of EVs in both health and disease; however, additional refinement of nanoscale flow cytometric methods is needed for use of these instruments for quantitative particle counting and sizing. Furthermore, larger scale studies are necessary to more clearly define the diurnal variations in circulating EVs, and thus further inform their use as biomarkers for disease.


Subject(s)
Extracellular Vesicles/physiology , Flow Cytometry , Adult , Extracellular Vesicles/chemistry , Humans , Liposomes/chemical synthesis , Liposomes/chemistry , Microscopy, Atomic Force , Particle Size
16.
Nat Commun ; 5: 5101, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25290058

ABSTRACT

CD4(+) T cells are involved in the development of autoimmunity, including multiple sclerosis (MS). Here we show that nicotinamide adenine dinucleotide (NAD(+)) blocks experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, by inducing immune homeostasis through CD4(+)IFNγ(+)IL-10(+) T cells and reverses disease progression by restoring tissue integrity via remyelination and neuroregeneration. We show that NAD(+) regulates CD4(+) T-cell differentiation through tryptophan hydroxylase-1 (Tph1), independently of well-established transcription factors. In the presence of NAD(+), the frequency of T-bet(-/-) CD4(+)IFNγ(+) T cells was twofold higher than wild-type CD4(+) T cells cultured in conventional T helper 1 polarizing conditions. Our findings unravel a new pathway orchestrating CD4(+) T-cell differentiation and demonstrate that NAD(+) may serve as a powerful therapeutic agent for the treatment of autoimmune and other diseases.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myelin Sheath/drug effects , NAD/pharmacology , Regeneration/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Homeostasis/drug effects , Mice , Tryptophan Hydroxylase/drug effects , Tryptophan Hydroxylase/metabolism
17.
Rev. centroam. obstet. ginecol ; 18(2): 38-46, abr.-jun. 2013. tab
Article in Spanish | LILACS | ID: lil-734120

ABSTRACT

Las causas de mortalidad materna en Latino América y el Caribe estan lideradas por los trastornos hipertensivos del embarazo, en donde una de cada 4 muertes maternas es por esta patología. En términos generales podemos discutir 5 grandes razones para explicar nuestras altas tasas de muerte materna...


Subject(s)
Humans , Hypertension/complications , Hypertension/diagnosis , Hypertension/prevention & control , Pre-Eclampsia/diagnosis , Pre-Eclampsia/mortality , Pre-Eclampsia/prevention & control
18.
Montevideo; CLAP/SMR; 2007. 211 p. ilus.(Publicación CLAP/SMR, 1562).
Monography in Spanish | LILACS | ID: lil-488759

ABSTRACT

La guía esta orientada al cuidado de la salud reproductiva y sexual a través de la atención permanente que comienza con los cuidados precocepcionales y continúa con el antenatal, la atención del aborto o del parto, el puerperio, la atención del recién nacido y culmina con la planficación familiar. Se pretende que los profesionales de salud tengan a su disposición en una misma publicación las herramientas clínicas necesarias para brindar una atención de calidad en los servicios de salud, dentro del marco de la estrategia de la APS renovada, a través intervenciones de probada eficacia.


Subject(s)
Humans , Female , Pregnancy , Infant, Newborn , Delivery of Health Care , Maternal Welfare , Postnatal Care , Primary Health Care , Reproductive Health Services , Reproductive Health , Maternal and Child Health , Women's Health
19.
La Paz; MPSSP; 1999. 32 p. tab.
Monography in Spanish | LILACS, LIBOCS, LIBOE, LIBOSP | ID: lil-276326

ABSTRACT

La presente evaluación ejecutado en tres hospitales de Bolivia dan a conocer que el aborto es una realidad a nivel mundial; en América Latina las tasas de aborto son tres veces más altas que en el resto del mundo siendo sus complicaciones la causa más importante de muertes maternas y de enfermedades crónicas. En nuestro país se realizan aproximadamente 115 abortos inducidos por día, haciendo un total de 40 mil a 50 mil por año. Las estadísticas oficiales reconocen que las complicaciones del aborto son responsables de un 27 a 35 por ciento del total de muertes maternas, y por lo tanto es muy importante capacitar a los prestadores de servicios para proveer a la mujer orientación con empatía que evite prejuzgarlas, brindandoles la oportunidad, a través de la planificación familiar, y evitar embarazos no deseados y por ende el aborto en condiciones de riesgo


Subject(s)
Humans , Health Services , Hospitals , Abortion, Spontaneous , Contraception , Quality of Health Care , Bolivia
20.
La Paz; s.n; mar. 1994. 20 p.
Monography in Spanish | LILACS, LIBOCS, LIBOE | ID: lil-408908

ABSTRACT

Mejorar la calidad de atención de los servicios de salud significa revertir muchos conceptos y prejuicios, tanto institucionales, como individuales del personal de salud, en especial sus ctitudes y esquemas rígidos apegados a una formación académica occidentalizada y prponderadamente biologiscista y un modelo de atención con tendencias cada vez mayores hacia la privatización. En este contexto los servicios de salud reproductiva son aún más sensibles, ya que afectan a la pareja, especialmente a la mujer y tacan un aspecto sumamente delicado y complejo como es el de la sexualidad humana...


Subject(s)
Humans , Male , Female , Pregnancy , Uterine Neoplasms , Maternal Mortality , Maternal Health Services , Women's Health Services , Bolivia , Quality of Health Care
SELECTION OF CITATIONS
SEARCH DETAIL
...