Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cytotherapy ; 24(7): 691-698, 2022 07.
Article in English | MEDLINE | ID: mdl-35279374

ABSTRACT

BACKGROUND AIMS: The final harvest or wash of a cell therapy product is an important step in manufacturing, as viable cell recovery is critical to the overall success of a cell therapy. Most harvest/wash approaches in the clinical lab involve centrifugation, which can lead to loss of cells and decreased viability of the final product. Here the authors report on a multi-center assessment of the LOVO Cell Processing System (Fresenius Kabi, Bad Homburg, Germany), a cell processing device that uses a spinning filtration membrane instead of centrifugation. METHODS: Four National Institutes of Health Production Assistance for Cellular Therapies cell processing facilities (CPFs) assessed the LOVO Cell Processing System for final harvest and/or wash of the following three different cell products: activated T cells (ATCs), tumor-infiltrating lymphocytes (TILs) and bone marrow-derived mesenchymal stromal cells (MSCs). Each site compared their current in-house, routinely used method of final cell harvest and/or wash with that of the LOVO device. RESULTS: Final harvest and/or wash of ATCs, TILs and MSCs using the LOVO system resulted in satisfactory cell viability and recovery with some substantial improvement over the in-house methods of CPFs. Processing time was variable among cell types/facilities. CONCLUSIONS: The LOVO Cell Processing System provides an alternative to centrifuge-based technologies. The system employs a spinning membrane filter, exposing cells to minimal g-forces compared with centrifugation, and is automated and closed. This small multi-center study demonstrated the ability of the LOVO device to yield satisfactory cell viability and recovery of T cells and MSCs.


Subject(s)
Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Centrifugation
2.
Clin Cancer Res ; 25(24): 7340-7350, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31558475

ABSTRACT

PURPOSE: Current protocols for CD19 chimeric antigen receptor-expressing T cells (CD19.CAR-T cells) require recipients to tolerate preinfusion cytoreductive chemotherapy, and the presence of sufficient target antigen on normal or malignant B cells. PATIENTS AND METHODS: We investigated whether additional stimulation of CD19.CAR-T cells through their native receptors can substitute for cytoreductive chemotherapy, inducing expansion and functional persistence of CD19.CAR-T even in patients in remission of B-cell acute lymphocytic leukemia. We infused a low dose of CD19.CAR-modified virus-specific T cells (CD19.CAR-VST) without prior cytoreductive chemotherapy into 8 patients after allogeneic stem cell transplant. RESULTS: Absent virus reactivation, we saw no CD19.CAR-VST expansion. In contrast, in patients with viral reactivation, up to 30,000-fold expansion of CD19.CAR-VSTs was observed, with depletion of CD19+ B cells. Five patients remain in remission at 42-60+ months. CONCLUSIONS: Dual T-cell receptor and CAR stimulation can thus potentiate effector cell expansion and CAR-target cell killing, even when infusing low numbers of effector cells without cytoreduction.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Adenoviridae/physiology , Adolescent , Antigens, CD19/metabolism , Child , Child, Preschool , Genetic Vectors , Herpesvirus 4, Human/physiology , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/virology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Retroviridae/physiology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Young Adult
3.
Virol J ; 16(1): 22, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30786885

ABSTRACT

BACKGROUND: The regulatory cyclin, Cyclin T1 (CycT1), is a host factor essential for HIV-1 replication in CD4 T cells and macrophages. The importance of CycT1 and the Positive Transcription Elongation Factor b (P-TEFb) complex for HIV replication is well-established, but regulation of CycT1 expression and protein levels during HIV replication and latency establishment in CD4 T cells is less characterized. METHODS: To better define the regulation of CycT1 levels during HIV replication in CD4 T cells, multiparameter flow cytometry was utilized to study the interaction between HIV replication (intracellular p24) and CycT1 of human peripheral blood memory CD4 T cells infected with HIV in vitro. CycT1 was further examined in CD4 T cells of human lymph nodes. RESULTS: In activated (CD3+CD28 costimulation) uninfected blood memory CD4 T cells, CycT1 was most significantly upregulated in maximally activated (CD69+CD25+ and HLA.DR+CD38+) cells. In memory CD4 T cells infected with HIV in vitro, two distinct infected populations of p24+CycT1+ and p24+CycT1- cells were observed during 7 days infection, suggestive of different phases of productive HIV replication and subsequent latency establishment. Intriguingly, p24+CycT1- cells were the predominant infected population in activated CD4 T cells, raising the possibility that productively infected cells may transition into latency subsequent to CycT1 downregulation. Additionally, when comparing infected p24+ cells to bystander uninfected p24- cells (after bulk HIV infections), HIV replication significantly increased T cell activation (CD69, CD25, HLA.DR, CD38, and Ki67) without concomitantly increasing CycT1 protein levels, possibly due to hijacking of P-TEFb by the viral Tat protein. Lastly, CycT1 was constitutively expressed at higher levels in lymph node CD4 T cells compared to blood T cells, potentially enhancing latency generation in lymphoid tissues. CONCLUSIONS: CycT1 is most highly upregulated in maximally activated memory CD4 T cells as expected, but may become less associated with T cell activation during HIV replication. The progression into latency may further be predicated by substantial generation of p24+CycT1- cells during HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cyclin T/genetics , HIV Infections/immunology , Virus Latency/physiology , Virus Replication/physiology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Gene Expression Regulation , HIV-1/physiology , Host Microbial Interactions , Humans , Positive Transcriptional Elongation Factor B/genetics , Transcriptional Activation
4.
Cancer ; 125(8): 1267-1280, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30620384

ABSTRACT

BACKGROUND: Re-expression of the imprinted tumor suppressor gene DIRAS family GTPase 3 (DIRAS3) (aplysia ras homology member I [ARHI]) induces autophagy and tumor dormancy in ovarian cancer xenografts, but drives autophagic cancer cell death in cell culture. The current study explored the tumor and host factors required to prevent autophagic cancer cell death in xenografts and the use of antibodies against those factors or their receptors to eliminate dormant autophagic ovarian cancer cells. METHODS: Survival factors (insulinlike growth factor 1 [IGF-1], vascular endothelial growth factor [VEGF], and interleukin 8 [IL-8]) were detected with growth factor arrays and measured using enzyme-linked immunoadsorbent assay analysis. Phosphorylation of protein kinase B (AKT), phosphorylation of extracellular signal-regulated kinase (ERK), nuclear localization of translocation factor EB (TFEB) or forkhead box O3a (FOXo3a), and expression of microtubule-associated proteins 1A/1B light chain 3B (MAPLC3B; LC3B) were examined using Western blot analysis. The effect of treatment with antibodies against survival factors or their receptors was studied using DIRAS3-induced dormant xenograft models. RESULTS: Ovarian cancer cells grown subcutaneously in nude mice exhibited higher levels of phosphorylated ERK/AKT activity and lower levels of nuclear TFEB/FOXo3a, MAPLC3B, and autophagy compared with cells grown in culture. Induction of autophagy and dormancy with DIRAS3 was associated with decreased ERK/AKT signaling. The addition of VEGF, IGF-1, and IL-8 weakened the inhibitory effect of DIRAS3 on ERK/AKT activity and reduced DIRAS3-mediated TFEB or FOXo3a nuclear localization and MAPLC3B expression in ovarian cancer cells. Treatment with antibodies against VEGF, IL-8, and IGF receptor inhibited the growth of dormant xenografts, thereby prolonging survival from 99 to >220 days (P < .05) and curing a percentage of mice. CONCLUSIONS: Treatment with a combination of anti-VEGF, anti-IL-8, and anti-IGF receptor antibodies prevented the outgrowth of dormant cells and prolonged survival in a preclinical model.


Subject(s)
Antibodies/administration & dosage , Interleukin-8/metabolism , Ovarian Neoplasms/drug therapy , Somatomedins/metabolism , Vascular Endothelial Growth Factor A/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Antibodies/pharmacology , Autophagy/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-8/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Phosphorylation , Somatomedins/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , rho GTP-Binding Proteins/genetics
5.
Basic Res Cardiol ; 112(1): 3, 2017 01.
Article in English | MEDLINE | ID: mdl-27882430

ABSTRACT

Autologous bone marrow mononuclear cell (BM-MNC) therapy for patients with ST-segment elevation myocardial infarction (STEMI) has produced inconsistent results, possibly due to BM-MNC product heterogeneity. Patient-specific cardiovascular risk factors (CRFs) may contribute to variations in BM-MNC composition. We sought to identify associations between BM-MNC subset frequencies and specific CRFs in STEMI patients. Bone marrow was collected from 191 STEMI patients enrolled in the CCTRN TIME and LateTIME trials. Relationships between BM-MNC subsets and CRFs were determined with multivariate analyses. An assessment of CRFs showed that hyperlipidemia and hypertension were associated with a higher frequency of CD11b+ cells (P = 0.045 and P = 0.016, respectively). In addition, we found that females had lower frequencies of CD11b+ (P = 0.018) and CD45+CD14+ (P = 0.028) cells than males, age was inversely associated with the frequency of CD45+CD31+ cells (P = 0.001), smoking was associated with a decreased frequency of CD45+CD31+ cells (P = 0.013), glucose level was positively associated with the frequency of CD45+CD3+ cells, and creatinine level (an indicator of renal function) was inversely associated with the frequency of CD45+CD3+ cells (P = 0.015). In conclusion, the frequencies of monocytic, lymphocytic, and angiogenic BM-MNCs varied in relation to patients' CRFs. These phenotypic variations may affect cell therapy outcomes and might be an important consideration when selecting patients for and reviewing results from autologous cell therapy trials.


Subject(s)
Bone Marrow Cells/cytology , Cardiovascular Diseases , Adult , Aged , Bone Marrow Transplantation , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Phenotype , Retrospective Studies , Risk Factors
6.
Am Heart J ; 179: 142-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27595689

ABSTRACT

BACKGROUND: Although several preclinical studies have shown that bone marrow cell (BMC) transplantation promotes cardiac recovery after myocardial infarction, clinical trials with unfractionated bone marrow have shown variable improvements in cardiac function. METHODS: To determine whether in a population of post-myocardial infarction patients, functional recovery after BM transplant is associated with specific BMC subpopulation, we examined the association between BMCs with left ventricular (LV) function in the LateTIME-CCTRN trial. RESULTS: In this population, we found that older individuals had higher numbers of BM CD133(+) and CD3(+) cells. Bone marrow from individuals with high body mass index had lower CD45(dim)/CD11b(dim) levels, whereas those with hypertension and higher C-reactive protein levels had higher numbers of CD133(+) cells. Smoking was associated with higher levels of CD133(+)/CD34(+)/VEGFR2(+) cells and lower levels of CD3(+) cells. Adjusted multivariate analysis indicated that CD11b(dim) cells were negatively associated with changes in LV ejection fraction and wall motion in both the infarct and border zones. Change in LV ejection fraction was positively associated with CD133(+), CD34(+), and CD45(+)/CXCR4(dim) cells as well as faster BMC growth rates in endothelial colony forming assays. CONCLUSIONS: In the LateTIME population, BM composition varied with patient characteristics and treatment. Irrespective of cell therapy, recovery of LV function was greater in patients with greater BM abundance of CD133(+) and CD34(+) cells and worse in those with higher levels of CD11b(dim) cells. Bone marrow phenotype might predict clinical response before BMC therapy and administration of selected BM constituents could potentially improve outcomes of other future clinical trials.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Recovery of Function , Ventricular Dysfunction, Left/therapy , AC133 Antigen/metabolism , Adult , Aged , Antigens, CD34/metabolism , Body Mass Index , Bone Marrow Cells/metabolism , C-Reactive Protein/metabolism , CD11b Antigen/metabolism , Cohort Studies , Female , Humans , Hypertension/metabolism , Leukocyte Common Antigens/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Obesity/metabolism , Prospective Studies , Receptors, CXCR4/metabolism , Smoking/metabolism , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
7.
J Clin Invest ; 126(9): 3363-76, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27482888

ABSTRACT

BACKGROUND: T cells expressing antigen-specific chimeric antigen receptors (CARs) improve outcomes for CD19-expressing B cell malignancies. We evaluated a human application of T cells that were genetically modified using the Sleeping Beauty (SB) transposon/transposase system to express a CD19-specific CAR. METHODS: T cells were genetically modified using DNA plasmids from the SB platform to stably express a second-generation CD19-specific CAR and selectively propagated ex vivo with activating and propagating cells (AaPCs) and cytokines. Twenty-six patients with advanced non-Hodgkin lymphoma and acute lymphoblastic leukemia safely underwent hematopoietic stem cell transplantation (HSCT) and infusion of CAR T cells as adjuvant therapy in the autologous (n = 7) or allogeneic settings (n = 19). RESULTS: SB-mediated genetic transposition and stimulation resulted in 2,200- to 2,500-fold ex vivo expansion of genetically modified T cells, with 84% CAR expression, and without integration hotspots. Following autologous HSCT, the 30-month progression-free and overall survivals were 83% and 100%, respectively. After allogeneic HSCT, the respective 12-month rates were 53% and 63%. No acute or late toxicities and no exacerbation of graft-versus-host disease were observed. Despite a low antigen burden and unsupportive recipient cytokine environment, CAR T cells persisted for an average of 201 days for autologous recipients and 51 days for allogeneic recipients. CONCLUSIONS: CD19-specific CAR T cells generated with SB and AaPC platforms were safe, and may provide additional cancer control as planned infusions after HSCT. These results support further clinical development of this nonviral gene therapy approach. TRIAL REGISTRATION: Autologous, NCT00968760; allogeneic, NCT01497184; long-term follow-up, NCT01492036. FUNDING: National Cancer Institute, private foundations, and institutional funds. Please see Acknowledgments for details.


Subject(s)
Antigens, CD19/metabolism , DNA Transposable Elements , Lymphoma, Non-Hodgkin/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/cytology , Adult , Antigen-Presenting Cells/immunology , Cytokines/metabolism , Disease-Free Survival , Female , Follow-Up Studies , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Humans , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Male , Middle Aged , Patient Safety , Plasmids/metabolism , Receptors, Antigen, T-Cell/metabolism , Transplantation, Homologous , Treatment Outcome , Young Adult
8.
Cell Transplant ; 25(9): 1675-1687, 2016.
Article in English | MEDLINE | ID: mdl-26590374

ABSTRACT

In the current study, we sought to identify bone marrow-derived mononuclear cell (BM-MNC) subpopulations associated with a combined improvement in left ventricular ejection fraction (LVEF), left ventricular end-systolic volume (LVESV), and maximal oxygen consumption (VO2 max) in patients with chronic ischemic cardiomyopathy 6 months after receiving transendocardial injections of autologous BM-MNCs or placebo. For this prospectively planned analysis, we conducted an embedded cohort study comprising 78 patients from the FOCUS-Cardiovascular Cell Therapy Research Network (CCTRN) trial. Baseline BM-MNC immunophenotypes and progenitor cell activity were determined by flow cytometry and colony-forming assays, respectively. Previously stable patients who demonstrated improvement in LVEF, LVESV, and VO2 max during the 6-month course of the FOCUS-CCTRN study (group 1, n = 17) were compared to those who showed no change or worsened in one to three of these endpoints (group 2, n = 61) and to a subset of patients from group 2 who declined in all three functional endpoints (group 2A, n = 11). Group 1 had higher frequencies of B-cell and CXCR4+ BM-MNC subpopulations at study baseline than group 2 or 2A. Furthermore, patients in group 1 had fewer endothelial colony-forming cells and monocytes/macrophages in their bone marrow than those in group 2A. To our knowledge, this is the first study to show that in patients with ischemic cardiomyopathy, certain bone marrow-derived cell subsets are associated with improvement in LVEF, LVESV, and VO2 max at 6 months. These results suggest that the presence of both progenitor and immune cell populations in the bone marrow may influence the natural history of chronic ischemic cardiomyopathy-even in stable patients. Thus, it may be important to consider the bone marrow composition and associated regenerative capacity of patients when assigning them to treatment groups and evaluating the results of cell therapy trials.


Subject(s)
Stem Cells/cytology , Ventricular Dysfunction, Left/therapy , Bone Marrow Transplantation , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Female , Heart Failure/physiopathology , Heart Failure/therapy , Humans , Immunophenotyping , Male , Middle Aged , Myocardial Ischemia/physiopathology , Myocardial Ischemia/therapy , Prospective Studies , Stroke Volume/physiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/physiology
9.
Circ Res ; 116(1): 99-107, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25406300

ABSTRACT

RATIONALE: Despite significant interest in bone marrow mononuclear cell (BMC) therapy for ischemic heart disease, current techniques have resulted in only modest benefits. However, selected patients have shown improvements after autologous BMC therapy, but the contributing factors are unclear. OBJECTIVE: The purpose of this study was to identify BMC characteristics associated with a reduction in infarct size after ST-segment-elevation-myocardial infarction. METHODS AND RESULTS: This prospective study comprised patients consecutively enrolled in the CCTRN TIME (Cardiovascular Cell Therapy Research Network Timing in Myocardial Infarction Evaluation) trial who agreed to have their BMCs stored and analyzed at the CCTRN Biorepository. Change in infarct size between baseline (3 days after percutaneous coronary intervention) and 6-month follow-up was measured by cardiac MRI. Infarct-size measurements and BMC phenotype and function data were obtained for 101 patients (mean age, 56.5 years; mean screening ejection fraction, 37%; mean baseline cardiac MRI ejection fraction, 45%). At 6 months, 75 patients (74.3%) showed a reduction in infarct size (mean change, -21.0±17.6%). Multiple regression analysis indicated that infarct size reduction was greater in patients who had a larger percentage of CD31(+) BMCs (P=0.046) and in those with faster BMC growth rates in colony-forming unit Hill and endothelial-colony forming cell functional assays (P=0.033 and P=0.032, respectively). CONCLUSIONS: This study identified BMC characteristics associated with a better clinical outcome in patients with segment-elevation-myocardial infarction and highlighted the importance of endothelial precursor activity in regenerating infarcted myocardium. Furthermore, it suggests that for these patients with segment-elevation-myocardial infarction, myocardial repair was more dependent on baseline BMC characteristics than on whether the patient underwent intracoronary BMC transplantation. CLINICAL TRIAL REGISTRATION INFORMATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00684021.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Myocardial Infarction/diagnosis , Myocardial Infarction/therapy , Adult , Aged , Cohort Studies , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies
10.
J Leukoc Biol ; 91(5): 759-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22416257

ABSTRACT

Some human memory CD4(+) T cells have cytotoxic functions best understood in the context of viral infections; however, their possible role in pathologic processes is understudied. The novel discovery that mitogenic CD28 antibodies induced proliferation and expansion of Tregs offered therapeutic promise for autoimmune disorders. However, the failed TGN1412 trial forced reassessment of this concept. As memory CD4(+) T cells are known to produce toxic molecules, including granzyme B (GrzB) and FasL, we wondered whether mitogenic CD28 was able to induce these cytotoxic molecules. A commercially available mitogenic human CD28 mAb (clone ANC28.1) was used to determine whether mitogenic CD28 induces cytotoxic function from human memory CD4(+) T cells. We found that stimulation of memory CD4(+) T cells by ANC28.1, as well as by conventional costimulation (CD3/CD28 mAb), robustly induced enzymatically active GrzB, along with increased surface expression of FasL. These functional phenotypes were induced in association with increased expression of T cell activation markers CD69 and CD25, and elimination of target cells by ANC28.1-activated memory CD4(+) T cells involved both GrzB and FasL. Additionally, ANC28.1-activated memory CD4(+) T cells caused disruption of epithelial cell monolayer integrity, which was partially mediated by GrzB. These findings reveal functions of memory CD4(+) T cells previously unknown to be induced by mitogenic CD28, and suggest that these pathogenic mechanisms may have been responsible for some of the widespread tissue destruction that occurred in the TGN1412 trial recipients.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Fas Ligand Protein/metabolism , Granzymes/metabolism , Immunologic Memory/immunology , Mitogens/metabolism , Antibodies, Monoclonal/pharmacology , Flow Cytometry , Humans , Signal Transduction
11.
Cytometry A ; 77(6): 502-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20235276

ABSTRACT

Microparticles, which include exosomes, micro-vesicles, apoptotic bodies and apoptotic microparticles, are small (0.05 - 3 mum in diameter), membranous vesicles that can contain DNA, RNA, miRNA, intracellular proteins and express extracellular surface markers from the parental cells. They can be secreted from intracellular multivesicular bodies or released from the surface of blebbing membranes. Circulating microparticles are abundant in the plasma of normal individuals and can be derived from circulating blood cells such as platelets, red blood cells and leukocytes as well as from tissue sources, such as endothelial and placental tissues. Elevated levels of microparticles are associated with various diseases such as thrombosis (platelet microparticles), congestive heart failure (endothelial microparticles), breast cancer patients (leukocyte microparticles) and women with preeclampsia (syncytiotrophoblast microparticles). Although microparticles can be detected by microscopy, enzyme-linked immunoassays and functional assays, flow cytometry is the preferred method because of the ability to quantitate (fluorescent bead- or flow rate-based method) and because of polychromatic capabilities. However, standardization of pre-analytical and analytical modus operandi for isolating, enumerating and fluorescent labeling of microparticles remains a challenge. The primary focus of this article is to review the preliminary steps required to optimally study circulating in vivo microparticles which include: 1) centrifugation speed used, 2) quantitation of microparticles before antibody labeling, 3) levels of fluorescence intensity of antibody-labeled microparticles, 4) polychromatic flow cytometric analysis of microparticle sub-populations and 5) use of polyclonal antibodies designed for Western blotting for flow cytometry. These studies determine a roadmap to develop microparticles as biomarkers for a variety of conditions.


Subject(s)
Blood Chemical Analysis/methods , Cell-Derived Microparticles/chemistry , Flow Cytometry , Blotting, Western , Cell Line , Fluorescence , Humans , Staining and Labeling
12.
Am J Pathol ; 173(6): 1595-608, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18974299

ABSTRACT

Microparticles (MPs) that circulate in blood may be a source of DNA for molecular analyses, including prenatal genetic diagnoses. Because MPs are heterogeneous in nature, however, further characterization is important before use in clinical settings. One key question is whether DNA is either bound to aggregates of blood proteins and lipid micelles or intrinsically associated with MPs from dying cells. To test the latter hypothesis, we asked whether MPs derived in vitro from dying cells were similar to those in maternal plasma. JEG-3 cells model extravillous trophoblasts, which predominate during the first trimester of pregnancy when prenatal diagnosis is most relevant. MPs were derived from apoptosis and increased over 48 hours. Compared with necrotic MPs, DNA in apoptotic MPs was more fragmented and resistant to plasma DNases. Membrane-specific dyes indicated that apoptotic MPs had more membranous material, which protects nucleic acids, including RNA. Flow cytometry showed that MPs derived from dying cells displayed light scatter and DNA staining similar to MPs found in maternal plasma. Quantification of maternal MPs using characteristics defined by MPs generated in vitro revealed a significant increase of DNA(+) MPs in the plasma of women with preeclampsia compared with plasma from women with normal pregnancies. Apoptotic MPs are therefore a likely source of stable DNA that could be enriched for both early genetic diagnosis and monitoring of pathological pregnancies.


Subject(s)
Apoptosis/physiology , Cell Membrane , Cell-Derived Microparticles/chemistry , Nucleic Acids/metabolism , Pre-Eclampsia , Trophoblasts/pathology , Cell Hypoxia , Cell Line , Cell Membrane/metabolism , Cell Membrane/pathology , Cell-Derived Microparticles/metabolism , Female , Fluorescent Dyes/metabolism , Humans , Male , Membrane Lipids/chemistry , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Trophoblasts/cytology
13.
Ann N Y Acad Sci ; 1075: 57-62, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17108192

ABSTRACT

Fetal DNA is found in the plasma of pregnant women that appears to be stable for PCR amplification. Although the underlying mechanism giving rise to this DNA in plasma remains unclear, the source of these fragments may be from apoptotic bodies (Apo-Bodies) created from dying cells. Trophoblast apoptosis is essential for normal placental development, given the enormous amount of proliferation, differentiation, and migration during pregnancy. Through flow cytometric analysis coupled with real-time PCR, our lab has shown that aggregates of acridine orange (AO)-stained material (apoptotic particles) are resistant to DNase treatment, disrupted by sodium dodecyl sulfate (SDS), and contain fetal DNA. Because the placenta continuously re-models in an hypoxic environment, our hypothesis is that fetal DNA in maternal plasma comes from hypoxia-induced dying trophoblasts and that this DNA circulates predominantly in the form of Apo-Bodies. We have developed a model culture system for analysis of Apo-Bodies derived from JEG-3 cells, an extravillous trophoblastic cell line, undergoing various methods of cell death: hypoxia-induced, etoposide-induced, and heat stress (necrosis like)-induced cell death. Under conditions of similar propidium iodide (PI) uptake, suggesting comparable levels of death, both hypoxia- and etoposide-induced Apo-Bodies increase in concentration over time, whereas heat-induced levels of particles remain fairly constant, indicating that production of DNA-associated Apo-Bodies is a continuous process. Hypoxia, which is likely to be responsible for trophoblast cell death in vivo, produced membrane-bound Apo-Bodies containing DNA. Our results are consistent with the characteristics of membrane-bound particles containing fetal DNA found in maternal plasma.


Subject(s)
Apoptosis/physiology , Cell Membrane/metabolism , DNA/metabolism , Fetus/physiology , Hypoxia , Plasma/chemistry , DNA Fragmentation , Female , Humans , Maternal-Fetal Exchange , Pregnancy
14.
J Immunol Methods ; 317(1-2): 45-55, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17067632

ABSTRACT

Flow cytometry provides accurate relative cellular quantitation (percent abundance) of cells from diverse samples, but technical limitations of most flow cytometers preclude accurate absolute quantitation. Several quantitation standards are now commercially available which, when added to samples, permit absolute quantitation of CD4+ T cells. However, these reagents are limited by their cost, technical complexity, requirement for additional software and/or limited applicability. Moreover, few studies have validated the use of such reagents in complex biological samples, especially for quantitation of non-T cells. Here we show that addition to samples of known quantities of polystyrene fluorescence standardization beads permits accurate quantitation of CD4+ T cells from complex cell samples. This procedure, here termed single bead-enhanced cytofluorimetry (SBEC), was equally capable of enumerating eosinophils as well as subcellular fragments of apoptotic cells, moieties with very different optical and fluorescent characteristics. Relative to other proprietary products, SBEC is simple, inexpensive and requires no special software, suggesting that the method is suitable for the routine quantitation of most cells and other particles by flow cytometry.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Separation/methods , Flow Cytometry/methods , Adoptive Transfer , Animals , Cell Line , Female , Fluorescence , Humans , Inflammation/pathology , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Polystyrenes
15.
J Biol Chem ; 277(52): 50761-7, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12399450

ABSTRACT

Structural and biochemical studies have revealed that procaspases form dimers prior to proteolytic activation. How the two procaspases interact in the dimer is unclear. To study the mechanisms of dimer-dependent caspase activation we used a heterodimeric system so that two caspase molecules can be specifically brought together. Surprisingly, only one caspase partner in the dimer needs to be enzymatically active for caspase processing and activation to occur. Caspase activation is inefficient in the dimer in the absence of intramolecular processing, suggesting that caspase activation is initiated via intramolecular processing. Homodimerization of caspase-8 or caspase-9 leads to the formation of a stable dimeric complex. However, heterodimerization between caspase-8 and caspases-3, -9, or -10 failed to induce stable dimer formation or caspase activation. Our data suggest that the formation of a stable dimeric intermediate initiates caspase activation.


Subject(s)
Caspases/chemistry , Caspases/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Dimerization , Enzyme Activation , Enzyme Stability , HeLa Cells , Humans , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tacrolimus Binding Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...