Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37115584

ABSTRACT

Hematopoietic stem cells (HSC) and downstream lineage-biased multipotent progenitors (MPP) tailor blood production and control myelopoiesis on demand. Recent lineage tracing analyses revealed MPPs to be major functional contributors to steady-state hematopoiesis. However, we still lack a precise resolution of myeloid differentiation trajectories and cellular heterogeneity in the MPP compartment. Here, we found that myeloid-biased MPP3 are functionally and molecularly heterogeneous, with a distinct subset of myeloid-primed secretory cells with high endoplasmic reticulum (ER) volume and FcγR expression. We show that FcγR+/ERhigh MPP3 are a transitional population serving as a reservoir for rapid production of granulocyte/macrophage progenitors (GMP), which directly amplify myelopoiesis through inflammation-triggered secretion of cytokines in the local bone marrow (BM) microenvironment. Our results identify a novel regulatory function for a secretory MPP3 subset that controls myeloid differentiation through lineage-priming and cytokine production and acts as a self-reinforcing amplification compartment in inflammatory stress and disease conditions.


Subject(s)
Hematopoiesis , Receptors, IgG , Cell Differentiation , Cell Lineage , Myeloid Cells , Guanylate Kinases/metabolism , Membrane Proteins/metabolism
2.
Blood Adv ; 2(19): 2462-2466, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30266821

ABSTRACT

Although the role of promyelocytic leukemia/retinoic acid receptor α (PML/RARA) fusion protein is well recognized in acute promyelocytic leukemia (APL), its contribution to initiation and maintenance of leukemogenesis is not completely understood. Transcriptome analysis in the murine MRP8-PML/RARA APL model has demonstrated modest alterations in gene expression accompanied by expansion of the promyelocyte compartment. Of particular interest, mice expressing PML/RARA showed downregulation of the transcription factor Irf8 mRNA. Interferon regulatory factor 8 (IRF8) is a known regulator of hematopoiesis. Previous research had implicated IRF8 as a tumor suppressor for myeloid neoplasia, and mice lacking IRF8 develop a well-differentiated myeloproliferative neoplasm characterized by expansion of neutrophilic lineage cells. We hypothesized that PML/RARA-mediated downregulation of Irf8 transcript levels contributes to the initiation of APL. We observed significant downregulation of IRF8 protein levels in highly purified promyelocyte populations of PML/RARA transgenic mice. We also found that loss of IRF8 results in expansion of promyelocytes in vivo, partially phenocopying the impact of PML/RARA expression. Moreover, survival experiments showed that complete loss of IRF8 leads to acceleration of APL onset in our PML/RARA mice. Collectively, these data identify IRF8 downregulation as an important factor in APL initiation and highlight a tumor-suppressor role for IRF8 in this acute leukemia.


Subject(s)
Genes, Tumor Suppressor , Interferon Regulatory Factors/genetics , Leukemia, Promyelocytic, Acute/genetics , Animals , Biomarkers, Tumor , Bone Marrow/pathology , Disease Models, Animal , Flow Cytometry , Humans , Interferon Regulatory Factors/metabolism , Leukemia, Promyelocytic, Acute/mortality , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
3.
Drug Metab Dispos ; 46(3): 237-247, 2018 03.
Article in English | MEDLINE | ID: mdl-29311136

ABSTRACT

Momelotinib (MMB), a small-molecule inhibitor of Janus kinase (JAK)1/2 and of activin A receptor type 1 (ACVR1), is in clinical development for the treatment of myeloproliferative neoplasms. The pharmacokinetics and disposition of [14C]MMB were characterized in a single-dose, human mass-balance study. Metabolism and the pharmacologic activity of key metabolites were elucidated in multiple in vitro and in vivo experiments. MMB was rapidly absorbed following oral dosing with approximately 97% of the radioactivity recovered, primarily in feces with urine as a secondary route. Mean blood-to-plasma [14C] area under the plasma concentration-time curve ratio was 0.72, suggesting low association of MMB and metabolites with blood cells. [14C]MMB-derived radioactivity was detectable in blood for ≤48 hours, suggesting no irreversible binding of MMB or its metabolites. The major circulating human metabolite, M21 (a morpholino lactam), is a potent inhibitor of JAK1/2 and ACVR1 in vitro. Estimation of pharmacological activity index suggests M21 contributes significantly to the pharmacological activity of MMB for the inhibition of both JAK1/2 and ACVR1. M21 was observed in disproportionately higher amounts in human plasma than in rat or dog, the rodent and nonrodent species used for the general nonclinical safety assessment of this molecule. This discrepancy was resolved with additional nonclinical studies wherein the circulating metabolites and drug-drug interactions were further characterized. The human metabolism of MMB was mediated primarily by multiple cytochrome P450 enzymes, whereas M21 formation involved initial P450 oxidation of the morpholine ring followed by metabolism via aldehyde oxidase.


Subject(s)
Benzamides/pharmacokinetics , Pyrimidines/pharmacokinetics , Adolescent , Adult , Animals , Cell Line , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions/physiology , Female , Hep G2 Cells , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Rats , Young Adult
4.
Blood ; 129(13): 1823-1830, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28188131

ABSTRACT

Patients with myelofibrosis (MF) often develop anemia and frequently become dependent on red blood cell transfusions. Results from a phase 2 study for the treatment of MF with the Janus kinase 1/2 (JAK1/2) inhibitor momelotinib (MMB) demonstrated that MMB treatment ameliorated anemia, which was unexpected for a JAK1/2 inhibitor, because erythropoietin-mediated JAK2 signaling is essential for erythropoiesis. Using a rat model of anemia of chronic disease, we demonstrated that MMB treatment can normalize hemoglobin and red blood cell numbers. We found that this positive effect is driven by direct inhibition of the bone morphogenic protein receptor kinase activin A receptor, type I (ACVR1), and the subsequent reduction of hepatocyte hepcidin production. Of note, ruxolitinib, a JAK1/2 inhibitor approved for the treatment of MF, had no inhibitory activity on this pathway. Further, we demonstrated the effect of MMB is not mediated by direct inhibition of JAK2-mediated ferroportin (FPN1) degradation, because neither MMB treatment nor myeloid-specific deletion of JAK2 affected FPN1 expression. Our data support the hypothesis that the improvement of inflammatory anemia by MMB results from inhibition of ACVR1-mediated hepcidin expression in the liver, which leads to increased mobilization of sequestered iron from cellular stores and subsequent stimulation of erythropoiesis.


Subject(s)
Anemia/drug therapy , Benzamides/therapeutic use , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Hepcidins/biosynthesis , Pyrimidines/therapeutic use , Activin Receptors, Type I/antagonists & inhibitors , Animals , Benzamides/pharmacology , Chronic Disease , Hepatocytes/metabolism , Iron/metabolism , Primary Myelofibrosis/complications , Pyrimidines/pharmacology , Rats
5.
Nature ; 543(7644): 205-210, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28241143

ABSTRACT

With age, haematopoietic stem cells lose their ability to regenerate the blood system, and promote disease development. Autophagy is associated with health and longevity, and is critical for protecting haematopoietic stem cells from metabolic stress. Here we show that loss of autophagy in haematopoietic stem cells causes accumulation of mitochondria and an activated metabolic state, which drives accelerated myeloid differentiation mainly through epigenetic deregulations, and impairs haematopoietic stem-cell self-renewal activity and regenerative potential. Strikingly, most haematopoietic stem cells in aged mice share these altered metabolic and functional features. However, approximately one-third of aged haematopoietic stem cells exhibit high autophagy levels and maintain a low metabolic state with robust long-term regeneration potential similar to healthy young haematopoietic stem cells. Our results demonstrate that autophagy actively suppresses haematopoietic stem-cell metabolism by clearing active, healthy mitochondria to maintain quiescence and stemness, and becomes increasingly necessary with age to preserve the regenerative capacity of old haematopoietic stem cells.


Subject(s)
Autophagy , Cell Self Renewal , Cellular Senescence , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Autophagy/genetics , Cell Self Renewal/genetics , Cellular Senescence/genetics , Epigenesis, Genetic , Female , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Male , Mice , Mitochondria/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism
7.
Nature ; 494(7437): 323-7, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23389440

ABSTRACT

Blood production is ensured by rare, self-renewing haematopoietic stem cells (HSCs). How HSCs accommodate the diverse cellular stresses associated with their life-long activity remains elusive. Here we identify autophagy as an essential mechanism protecting HSCs from metabolic stress. We show that mouse HSCs, in contrast to their short-lived myeloid progeny, robustly induce autophagy after ex vivo cytokine withdrawal and in vivo calorie restriction. We demonstrate that FOXO3A is critical to maintain a gene expression program that poises HSCs for rapid induction of autophagy upon starvation. Notably, we find that old HSCs retain an intact FOXO3A-driven pro-autophagy gene program, and that ongoing autophagy is needed to mitigate an energy crisis and allow their survival. Our results demonstrate that autophagy is essential for the life-long maintenance of the HSC compartment and for supporting an old, failing blood system.


Subject(s)
Autophagy/genetics , Energy Metabolism/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Stress, Physiological/genetics , Aging , Animals , Apoptosis , Caloric Restriction , Cell Survival/genetics , Cellular Senescence , Cytokines/deficiency , Cytokines/metabolism , Food Deprivation , Forkhead Box Protein O3 , Homeostasis , Mice , Mice, Inbred C57BL
8.
Cell Stem Cell ; 12(1): 1-3, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23290130

ABSTRACT

Hematopoietic stem cells (HSCs) must at times exit quiescence to divide and produce differentiated blood cells. In this issue of Cell Stem Cell, Takubo et al. (2013) and Yu et al. (2013) show that these opposing actions require distinct metabolic programs to meet the changing energy demands of self-renewing HSCs.

9.
J Cell Biol ; 195(5): 709-20, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22123859

ABSTRACT

Hematopoietic stem cells (HSCs) give rise to all lineages of blood cells. Because HSCs must persist for a lifetime, the balance between their proliferation and quiescence is carefully regulated to ensure blood homeostasis while limiting cellular damage. Cell cycle regulation therefore plays a critical role in controlling HSC function during both fetal life and in the adult. The cell cycle activity of HSCs is carefully modulated by a complex interplay between cell-intrinsic mechanisms and cell-extrinsic factors produced by the microenvironment. This fine-tuned regulatory network may become altered with age, leading to aberrant HSC cell cycle regulation, degraded HSC function, and hematological malignancy.


Subject(s)
Cell Cycle Proteins/physiology , Cell Cycle/physiology , Hematopoietic Stem Cells/cytology , Aging , Animals , Cell Proliferation , Embryo, Mammalian/cytology , Mice , Models, Biological , Signal Transduction
10.
Genes Dev ; 25(16): 1746-57, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21852537

ABSTRACT

MdmX, also known as Mdm4, is a critical negative regulator of p53, and its overexpression serves to block p53 tumor suppressor function in many cancers. Consequently, inhibiting MdmX has emerged as an attractive approach to restoring p53 function in those cancers that retain functional p53. However, the consequences of acute systemic MdmX inhibition in normal adult tissues remain unknown. To determine directly the effects of systemic MdmX inhibition in normal tissues and in tumors, we crossed mdmX(-/-) mice into the p53ER(TAM) knockin background. In place of wild-type p53, p53ER(TAM) knockin mice express a variant of p53, p53ER(TAM), that is completely dependent on 4-hydroxy-tamoxifen for its activity. MdmX inhibition was then modeled by restoring p53 function in these MdmX-deficient mice. We show that MdmX is continuously required to buffer p53 activity in adult normal tissues and their stem cells. Importantly, the effects of transient p53 restoration in the absence of MdmX are nonlethal and reversible, unlike transient p53 restoration in the absence of Mdm2, which is ineluctably lethal. We also show that the therapeutic impact of restoring p53 in a tumor model is enhanced in the absence of MdmX, affording a significant extension of life span over p53 restoration in the presence of MdmX. Hence, systemic inhibition of MdmX is both a feasible therapeutic strategy for restoring p53 function in tumors that retain wild-type p53 and likely to be significantly safer than inhibition of Mdm2.


Subject(s)
Lymphoma/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian/cytology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Immunoblotting , Kaplan-Meier Estimate , Liver/drug effects , Liver/metabolism , Lymphoma/drug therapy , Lymphoma/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
J Biol Chem ; 286(28): 25098-107, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21613222

ABSTRACT

Mcl-1, a pro-survival member of the Bcl-2 family located at the mitochondrial outer membrane, is subject to constitutive ubiquitylation by the Bcl-2 homology 3-only E3 ligase, Mule/Lasu1, resulting in rapid steady-state degradation via the proteasome. Insertion of newly synthesized Mcl-1 into the mitochondrial outer membrane is dependent on its C-terminal transmembrane segment, but once inserted, the N terminus of a portion of the Mcl-1 molecules can be subject to proteolytic processing. Remarkably, this processing requires an intact electrochemical potential across the inner membrane. Three lines of evidence directed at the endogenous protein, however, indicate that the resulting Mcl-1ΔN isoform resides in the outer membrane: (i) full-length Mcl-1 and Mcl-1ΔN resist extraction by alkali but are accessible to exogenous protease; (ii) almost the entire populations of Mcl-1 and Mcl-1ΔN are accessible to the membrane-impermeant Cys-reactive agent 4-acetamido-4'-[(iodoacetyl)amino]stilbene-2,2'-disulfonic acid; and (iii) Mcl-1 and Mcl-1ΔN exhibit equivalent chemical cross-linking to Bak in intact mitochondria, an Mcl-1 binding partner located in the outer membrane. In addition to the Mule Bcl-2 homology 3 domain, we show that interaction between Mcl-1 and Mule also requires the extreme N terminus of Mcl-1, which is lacking in Mcl-1ΔN. Thus, Mcl-1ΔN does not interact with Mule, exhibits reduced steady-state ubiquitylation, evades the hyper-rapid steady-state degradation that is observed for full-length Mcl-1 in response to treatments that limit global protein synthesis, and confers resistance to UV stress-induced cell death.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Proteolysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites , Cell Death/physiology , Cell Death/radiation effects , HeLa Cells , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Myeloid Cell Leukemia Sequence 1 Protein , NIH 3T3 Cells , Protein Biosynthesis/physiology , Protein Biosynthesis/radiation effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology , Ubiquitination/radiation effects , Ultraviolet Rays
12.
Article in English | MEDLINE | ID: mdl-21412991

ABSTRACT

Hematopoiesis, the process by which all mature blood cells are generated from multipotent hematopoietic stem cells (HSCs), is a finely tuned balancing act in which HSCs must constantly decide between different cell fates: to proliferate, to self-renew or differentiate, to stay quiescent in the bone marrow niche or migrate to the periphery, to live or die. These fates are regulated by a complex interplay between cell-extrinsic cues and cell-intrinsic regulatory pathways whose function is to maintain a homeostatic balance between HSC self-renewal and life-long replenishment of lost blood cells. Improper regulation of these competing cellular programs can transform HSCs and progenitor cells into disease-initiating leukemic stem cells (LSCs). Strikingly, many of the mechanisms required for maintenance of normal HSC fate decisions are equally critical for the aberrant functions of LSCs. Because of the inherent complexities of these molecular mechanisms, a systematic approach to understanding the regulatory networks underlying HSC self-renewal is critical for uncovering the similarities and differences between HSCs and LSCs. In this review, we focus on recent developments in elucidating the regulatory networks governing normal HSC self-renewal programs and their implications for leukemic transformation. We describe the current technical and methodological limitations in isolating and characterizing HSCs and LSCs, and the emerging approaches that may afford a better understanding of the regulation of normal and leukemic hematopoiesis. Finally, we discuss how such basic mechanistic information may be of use for the design of novel therapies that will selectively reprogram and/or eliminate LSCs.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Cycle Proteins/metabolism , Cellular Reprogramming , Epigenesis, Genetic , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Signal Transduction
13.
Cell Stem Cell ; 7(2): 174-85, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20619762

ABSTRACT

Most adult stem cells, including hematopoietic stem cells (HSCs), are maintained in a quiescent or resting state in vivo. Quiescence is widely considered to be an essential protective mechanism for stem cells that minimizes endogenous stress caused by cellular respiration and DNA replication. We demonstrate that HSC quiescence can also have detrimental effects. We found that HSCs have unique cell-intrinsic mechanisms ensuring their survival in response to ionizing irradiation (IR), which include enhanced prosurvival gene expression and strong activation of p53-mediated DNA damage response. We show that quiescent and proliferating HSCs are equally radioprotected but use different types of DNA repair mechanisms. We describe how nonhomologous end joining (NHEJ)-mediated DNA repair in quiescent HSCs is associated with acquisition of genomic rearrangements, which can persist in vivo and contribute to hematopoietic abnormalities. Our results demonstrate that quiescence is a double-edged sword that renders HSCs intrinsically vulnerable to mutagenesis following DNA damage.


Subject(s)
DNA Repair , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutagenesis/genetics , Animals , Cell Death/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , DNA Damage , DNA Repair/radiation effects , Hematopoietic Stem Cells/radiation effects , Mice , Mice, Inbred C57BL , Mutagenesis/radiation effects , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/radiation effects , Radiation Tolerance/radiation effects , Radiation, Ionizing , Recombination, Genetic/radiation effects , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism
14.
Curr Mol Med ; 8(2): 138-47, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18336294

ABSTRACT

Accumulating evidence suggests that Mcl-1 plays a critical pro-survival role in the development and maintenance of both normal and malignant tissues. Regulation of Mcl-1 expression occurs at multiple levels, allowing for either the rapid induction or elimination of the protein in response to different cellular events. This suggests that Mcl-1 can play an early role in response to signals directing either cell survival or cell death. Deregulation of pathways regulating Mcl-1 that result in its over-expression likely contribute to a cell's inability to properly respond to death signals possibly leading to cell immortalization and tumorigenic conversion. Correspondingly, Mcl-1 has been shown to be up-regulated in numerous hematological and solid tumor malignancies. Moreover, this up-regulation appears to be a factor in the resistance of some cancer types to conventional cancer therapies. Mechanisms that abrogate the pro-survival function of Mcl-1 either by diminishing its levels or inactivating its functional BH3 groove have shown promise for the combinational treatment with existing cancer therapies and as single agents in certain malignancies. Here we review the various pathways that regulate Mcl-1 expression and describe agents that are currently under development to modulate Mcl-1 activity for therapeutic benefit in oncology.


Subject(s)
Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Humans , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction
15.
FEBS Lett ; 579(25): 5603-8, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16213503

ABSTRACT

A genome wide search for new BH3-containing Bcl-2 family members was conducted using position weight matrices (PWM) and identified a large (480kDa), novel BH3-only protein, originally called LASU1 (now also known as Ureb-1, E3(histone), ARF-BP1, and Mule). We demonstrated that LASU1 is an E3 ligase that ubiquitinated Mcl-1 in vitro and was required for its proteasome-dependent degradation in HeLa cells. Of note, the BH3 domain of LASU1 interacted with Mcl-1 but not with Bcl-2 or Bcl-Xl. A competing BH3-ligand derived from Bim interacted with Mcl-1 and prevented its interaction with LASU1 in HeLa cells, causing elevation of the steady-state levels of Mcl-1. This suggests that the unliganded form of Mcl-1 is sensitive to LASU1-mediated degradation of Mcl-1.


Subject(s)
Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cells, Cultured , Computational Biology , Humans , Ligands , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitins/metabolism
16.
Catheter Cardiovasc Interv ; 62(4): 519-25, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15274165

ABSTRACT

The objective of this study was to determine the feasibility of device closure of patent foramen ovale (PFO) for presumed paradoxical emboli without echocardiographic guidance or balloon sizing and the clinical outcome after device closure. Closure of the PFO has been proposed as an alternative to anticoagulation in patients with presumed paradoxical emboli. At present, most centers perform device closure with transesophageal echocardiographic guidance and balloon sizing of the defect. Between May 1998 and April 2002, 92 consecutive patients underwent device closure for a PFO using fluoroscopic monitoring only. Procedural success and major complications were recorded. Follow-up outcomes were recurrence rate and residual atrial shunting on transthoracic echocardiography. All patients (mean age, 45 +/- 13 years; 52% male) had successful device deployment using either the CardioSeal (n = 78) and Amplatzer (n = 14) PFO occluders with no major complications. Mean procedure time and fluoroscopy time was 27 +/- 13 and 6 +/- 4 min, respectively. One patient had a residual shunt on echocardiography at 1 year. Cumulative event-free survival for recurrence of paradoxical embolus at 1 year was 97.3% +/- 1.8%. This study provides a basis for device closure of PFO becoming a safe, day-case procedure, resulting in a low rate of residual shunting and recurrent thromboembolic events.


Subject(s)
Cardiac Catheterization/instrumentation , Embolism, Paradoxical/etiology , Foramen Ovale, Patent/therapy , Ischemic Attack, Transient/etiology , Stroke/etiology , Adult , Aged , Anticoagulants/therapeutic use , Cardiac Catheterization/adverse effects , Disease-Free Survival , Echocardiography, Doppler, Color , Echocardiography, Transesophageal , Embolism, Paradoxical/complications , Embolism, Paradoxical/diagnostic imaging , Embolism, Paradoxical/therapy , Equipment Design , Feasibility Studies , Female , Fluoroscopy , Follow-Up Studies , Foramen Ovale, Patent/complications , Foramen Ovale, Patent/diagnostic imaging , Humans , Ischemic Attack, Transient/diagnostic imaging , Ischemic Attack, Transient/therapy , Male , Middle Aged , Recurrence , Research Design , Retrospective Studies , Stroke/diagnostic imaging , Stroke/therapy , Time Factors , Treatment Outcome
17.
Catheter Cardiovasc Interv ; 61(1): 131-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14696173

ABSTRACT

Outcomes of device closure of large and small secundum atrial septal defects (ASDs) as related to rim anatomy with the Amplatzer atrial septal occluder were compared. Rim adequacy (> or = 5mm) of the anterior, inferior, posterior, and superior rims was determined using transesophageal echocardiography. Balloon-stretched defect size defined patients into two groups: group 1, < or = 25 mm (n = 138); group 2, > 25 mm (n = 34). Rim deficiency (n = 62) was more frequent in group 2 compared to group 1 (50% vs. 33%; P = 0.07), especially inferior rim deficiency (35% vs. 2%; P = 0.005). Device deployment was successful in group 1 and group 2 (100% vs. 91%; P = 0.007). Unsuccessful deployment was associated with an ASD of > 25 mm (P = 0.007) and inferior rim deficiency (P = 0.001). At first follow-up (54 +/- 16 days), right ventricular systolic pressure had improved in both groups (P < 0.001). Closure of a large ASD associated with a lack of support in the inferior rim may warrant alternative strategies to position the device successfully.


Subject(s)
Heart Septal Defects, Atrial/therapy , Prosthesis Implantation/methods , Adult , Body Weights and Measures , Cardiac Catheterization/methods , Echocardiography, Transesophageal , Female , Heart Septal Defects, Atrial/diagnostic imaging , Humans , Male , Middle Aged , Prostheses and Implants , Treatment Outcome
18.
J Am Coll Cardiol ; 41(12): 2252-8, 2003 Jun 18.
Article in English | MEDLINE | ID: mdl-12821256

ABSTRACT

OBJECTIVES: The study was done to determine the prevalence of pulmonary emboli (PE) in asymptomatic adult Fontan patients and to identify the risk factors associated with PE. BACKGROUND: Right atrial thrombi and systemic thromboembolic complications have been reported after the Fontan procedure. However, the frequency of silent PE in this patient population is not known. METHODS: All consecutive adult Fontan patients attending the adult congenital clinic over a six-month period underwent ventilation-perfusion (VQ) scanning and blood testing for thrombophilia tendency. If the VQ scan showed an intermediate or high probability for PE, a computerized tomography (CT) pulmonary angiogram was performed to confirm the presence of PE. RESULTS: Thirty patients (mean age 26 +/- 7 years, 57% men) were included in this study. Five (17%) adult Fontan patients had an intermediate or high probability for PE on VQ scan, all of which were confirmed on CT pulmonary angiography. No patient had a thrombophilia tendency. Pulmonary emboli were not present in any patients (30%) taking warfarin. Late age at time of Fontan operation (19 +/- 6 years vs. 11 +/- 6 years, p = 0.012) and type of Fontan anatomy (p = 0.001) were associated with increased risk of silent PE. CONCLUSIONS: Seventeen percent of adult patients with Fontan procedure have clinically silent PE. The long-term hemodynamic implications of this with respect to Fontan attrition over time are unknown. Large randomized prospective studies looking at anticoagulation therapy in all Fontan patients are urgently needed.


Subject(s)
Fontan Procedure/adverse effects , Heart Defects, Congenital/surgery , Pulmonary Embolism/diagnosis , Pulmonary Embolism/etiology , Adolescent , Adult , Echocardiography, Doppler , Female , Follow-Up Studies , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/epidemiology , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Prevalence , Pulmonary Embolism/epidemiology , Risk Factors , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...