Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Nat Immunol ; 16(12): 1282-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502406

ABSTRACT

To elucidate the transcriptional 'landscape' that regulates human lymphoid commitment during postnatal life, we used RNA sequencing to assemble the long non-coding transcriptome across human bone marrow and thymic progenitor cells spanning the earliest stages of B lymphoid and T lymphoid specification. Over 3,000 genes encoding previously unknown long non-coding RNAs (lncRNAs) were revealed through the analysis of these rare populations. Lymphoid commitment was characterized by lncRNA expression patterns that were highly stage specific and were more lineage specific than those of protein-coding genes. Protein-coding genes co-expressed with neighboring lncRNA genes showed enrichment for ontologies related to lymphoid differentiation. The exquisite cell-type specificity of global lncRNA expression patterns independently revealed new developmental relationships among the earliest progenitor cells in the human bone marrow and thymus.


Subject(s)
B-Lymphocytes/metabolism , Cell Lineage/genetics , Lymphoid Progenitor Cells/metabolism , RNA, Long Noncoding/genetics , T-Lymphocytes/metabolism , Transcriptome , Bayes Theorem , Bone Marrow Cells/metabolism , Cluster Analysis , Gene Expression Profiling/methods , Gene Ontology , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA/methods , Thymus Gland/cytology , Thymus Gland/metabolism
3.
Nat Immunol ; 13(10): 963-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22941246

ABSTRACT

Expression of the cell-surface antigen CD10 has long been used to define the lymphoid commitment of human cells. Here we report a unique lymphoid-primed population in human bone marrow that was generated from hematopoietic stem cells (HSCs) before onset of the expression of CD10 and commitment to the B cell lineage. We identified this subset by high expression of the homing molecule L-selectin (CD62L). CD10(-)CD62L(hi) progenitors had full lymphoid and monocytic potential but lacked erythroid potential. Gene-expression profiling placed the CD10(-)CD62L(hi) population at an intermediate stage of differentiation between HSCs and lineage-negative (Lin(-)) CD34(+)CD10(+) progenitors. CD62L was expressed on immature thymocytes, and its ligands were expressed at the cortico-medullary junction of the thymus, which suggested a possible role for this molecule in homing to the thymus. Our studies identify the earliest stage of lymphoid priming in human bone marrow.


Subject(s)
Bone Marrow Cells/immunology , Hematopoietic Stem Cells/metabolism , L-Selectin/biosynthesis , Neprilysin/biosynthesis , Antigens, CD34/immunology , Antigens, CD34/metabolism , Antigens, CD7/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Gene Expression Profiling , Hematopoietic Stem Cells/immunology , Humans , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/metabolism , Up-Regulation
4.
Int J Cancer ; 151(11): 1913-1924, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-35793239

ABSTRACT

Pediatric oncology patients are at risk for poor outcomes with respiratory viral infections. Outcome data for COVID-19 in children and young adults with cancer are needed; data are sparse for obese/overweight and adolescent and young adult subgroups. We conducted a single center cohort study of COVID-19 outcomes in patients younger than 25 years with cancer. Candidate hospitalization risk factors were analyzed via univariable and multivariable analyses. Eighty-seven patients with cancer and COVID-19 were identified. Most were Hispanic/Latinx (n = 63, 72%). Forty-two (48%) were overweight/obese. Anticancer therapy included chemotherapy only (n = 64, 74%), chimeric antigen receptor T-cells (CAR-T, n = 7), hematopoietic stem cell transplantation (HSCT, n = 12), or CAR-T and HSCT (n = 4). There was no COVID-19 related mortality. Twenty-six patients (30%) required COVID-19 related hospitalization; 4 required multiple hospitalizations. Nine (10%) had severe/critical infection; 6 needed intensive care. COVID-19 resulted in anticancer therapy delays in 22 (34%) of 64 patients on active therapy (median delay = 14 days). Factors associated with hospitalization included steroids within 2 weeks prior to infection, lymphopenia, previous significant non-COVID infection, and low COVID-19 PCR cycle threshold value. CAR-T recipients with B-cell aplasia tended to have severe/critical infection (3 of 7 patients). A COVID-19 antibody response was detected in 14 of 32 patients (44%). A substantial proportion of COVID-19 infected children and young adults with cancer require inpatient management; morbidity may be high in B-cell immunodeficiency. However, a majority of patients can be taken through chemotherapy without prolonged therapy delays. Viral load is a potential outcome predictor in COVID-19 in pediatric cancer.


Subject(s)
COVID-19 , Hematopoietic Stem Cell Transplantation , Neoplasms , Receptors, Chimeric Antigen , Adolescent , Child , Cohort Studies , Humans , Neoplasms/complications , Neoplasms/therapy , Obesity , Overweight , Young Adult
5.
Blood ; 136(2): 210-223, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32219444

ABSTRACT

Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking Ab P5G10 induces apoptosis in primary ALL cells in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-cell ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL.


Subject(s)
Drug Resistance, Neoplasm , Gene Deletion , Integrin alpha6 , Neoplasm Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Pyrimidines/pharmacology , Animals , Antibodies, Neoplasm/pharmacology , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Integrin alpha6/genetics , Integrin alpha6/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy
7.
Pediatr Blood Cancer ; 65(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-28876513

ABSTRACT

While early studies reported superior survival for cancer patients enrolled on clinical trials, recent findings are inconclusive. We investigated the association between enrollment on contemporary trials and event-free survival (EFS) in pediatric B-cell acute lymphoblastic leukemia (B-ALL). In a retrospective cohort of 274 children (1-21 years) treated for B-ALL from 2008 to 2015, 55.5% enrolled with no disparity in enrollment by age, sex, or ethnicity. Three-year EFS was similar for enrolled and not enrolled patients (90.1% [95% CI, 82.5-94.5] versus 86.5% [95% CI, 77.7-92.0]). Clinical trial enrollment did not affect pediatric B-ALL survival, albeit in a limited-size cohort treated at a single academic institution.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Child , Child, Preschool , Clinical Trials as Topic , Disease-Free Survival , Female , Humans , Infant , Male , Retrospective Studies , Survival Rate
9.
Blood ; 121(15): 2891-901, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23412095

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) emerge and develop adjacent to blood vessel walls in the yolk sac, aorta-gonad-mesonephros region, embryonic liver, and fetal bone marrow. In adult mouse bone marrow, perivascular cells shape a "niche" for HSPCs. Mesenchymal stem/stromal cells (MSCs), which support hematopoiesis in culture, are themselves derived in part from perivascular cells. In order to define their direct role in hematopoiesis, we tested the ability of purified human CD146(+) perivascular cells, as compared with unfractionated MSCs and CD146(-) cells, to sustain human HSPCs in coculture. CD146(+) perivascular cells support the long-term persistence, through cell-to-cell contact and at least partly via Notch activation, of human myelolymphoid HSPCs able to engraft primary and secondary immunodeficient mice. Conversely, unfractionated MSCs and CD146(-) cells induce differentiation and compromise ex vivo maintenance of HSPCs. Moreover, CD146(+) perivascular cells express, natively and in culture, molecular markers of the vascular hematopoietic niche. Unexpectedly, this dramatic, previously undocumented ability to support hematopoietic stem cells is present in CD146(+) perivascular cells extracted from the nonhematopoietic adipose tissue.


Subject(s)
Blood Vessels/physiology , CD146 Antigen/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Adult , Animals , Antigens, CD34/metabolism , Blood Vessels/cytology , Blotting, Western , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Communication , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Coculture Techniques , Fetal Blood/cytology , Fetal Blood/metabolism , Hematopoietic Stem Cells/cytology , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Receptors, Notch/genetics , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins
10.
Stem Cells ; 32(9): 2386-96, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24801626

ABSTRACT

A system that allows manipulation of the human thymic microenvironment is needed both to elucidate the extrinsic mechanisms that control human thymopoiesis and to develop potential cell therapies for thymic insufficiency. In this report, we developed an implantable thymic microenvironment composed of two human thymic stroma populations critical for thymopoiesis; thymic epithelial cells (TECs) and thymic mesenchyme (TM). TECs and TM from postnatal human thymi were cultured in specific conditions, allowing cell expansion and manipulation of gene expression, before reaggregation into a functional thymic unit. Human CD34+ hematopoietic stem and progenitor cells (HSPC) differentiated into T cells in the aggregates in vitro and in vivo following inguinal implantation of aggregates in immune deficient mice. Cord blood HSPC previously engrafted into murine bone marrow (BM), migrated to implants, and differentiated into human T cells with a broad T cell receptor repertoire. Furthermore, lentiviral-mediated expression of vascular endothelial growth factor in TM enhanced implant size and function and significantly increased thymocyte production. These results demonstrate an in vivo system for the generation of T cells from human HSPC and represent the first model to allow manipulation of gene expression and cell composition in the microenvironment of the human thymus.


Subject(s)
Thymus Gland/cytology , Tissue Engineering/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Proliferation/physiology , Cellular Microenvironment/physiology , Gene Expression , Humans , Lymphopoiesis/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Thymus Gland/drug effects
11.
Stem Cells ; 32(6): 1503-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24677652

ABSTRACT

Unlimited self renewal capacity and differentiation potential make human pluripotent stem cells (PSC) a promising source for the ex vivo manufacture of red blood cells (RBCs) for safe transfusion. Current methods to induce erythropoiesis from PSC suffer from low yields of RBCs, most of which are immature and contain embryonic and fetal rather than adult hemoglobins. We have previously shown that homodimerization of the intracellular component of MPL (ic-MPL) induces erythropoiesis from human cord blood progenitors. The goal of this study was to investigate the potential of ic-MPL dimerization to induce erythropoiesis from human embryonic stem cells (hESCs) and to identify the signaling pathways activated by this strategy. We present here the evidence that ic-MPL dimerization induces erythropoietin (EPO)-independent erythroid differentiation from hESC by inducing the generation of erythroid progenitors and by promoting more efficient erythroid maturation with increased RBC enucleation as well as increased gamma:epsilon globin ratio and production of beta-globin protein. ic-MPL dimerization is significantly more potent than EPO in inducing erythropoiesis, and its effect is additive to EPO. Signaling studies show that dimerization of ic-MPL, unlike stimulation of the wild type MPL receptor, activates AKT in the absence of JAK2/STAT5 signaling. AKT activation upregulates GATA-1 and FOXO3 transcriptional pathways with resulting inhibition of apoptosis, modulation of cell cycle, and enhanced maturation of erythroid cells. These findings open up potential new targets for the generation of therapeutically relevant RBC products from hPSC.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Erythropoiesis , Erythropoietin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Protein Multimerization , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism
12.
Pediatr Blood Cancer ; 62(11): 2040-3, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25976383

ABSTRACT

The role of end of induction minimal residual disease (MRD) as determined by flow cytometry for treatment assignment in pediatric T-cell acute lymphoblastic leukemia (T-ALL) is not well defined. We studied 33 children with newly diagnosed T-ALL. Thirty-two of 33 patients remain in continuous complete remission at a median of 4 years. Nineteen patients were MRD positive at the end of induction and all remain in remission with augmented Berlin Frankfurt Münster-based therapy. One patient underwent hematopoietic stem cell transplant for rising MRD. Persistent end of induction MRD alone is not an indication to alter therapy in pediatric T-ALL.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Remission Induction , Child, Preschool , Female , Humans , Infant , Male , Neoplasm, Residual , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/blood , Risk Assessment
13.
J Clin Immunol ; 33(4): 711-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23274800

ABSTRACT

During the last five decades, elegant mouse models of hematopoiesis have yielded most of the seminal insights into this complex biological system of self-renewal and lineage commitment. More recent advances in assays to measure human stem and progenitor cells as well as high resolution RNA profiling have revealed that although the basic roadmap of blood development is generally conserved across mammals, evolutionary pressures have generated many differences between the species that have important biological and translational implications. To enhance the utility of the mouse as a model organism, it is more important than ever that research data are presented with regard to how they might be influenced by the species of origin as well as the developmental source of the hematopoietic tissue.


Subject(s)
Hematopoiesis/immunology , Lymphocytes/immunology , Lymphoid Progenitor Cells/immunology , Animals , Cell Differentiation , Cell Lineage , Disease Models, Animal , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Mice , Species Specificity
14.
Stem Cells ; 30(4): 697-708, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22290824

ABSTRACT

The cytokine thrombopoietin (Tpo) plays a critical role in hematopoiesis by binding to the extracellular domain and inducing homodimerization of the intracellular signaling domain of its receptor, c-Mpl. Mpl homodimerization can also be accomplished by binding of a synthetic ligand to a constitutively expressed fusion protein F36VMpl consisting of a ligand binding domain (F36V) and the intracellular signaling domain of Mpl. Unexpectedly, in contrast to Tpo stimulation, robust erythropoiesis is induced after dimerization of F36VMpl in human CD34+ progenitor cells. The goal of this study was to define the hematopoietic progenitor stages at which dimerization of intracellular Mpl induces erythropoiesis and the downstream molecular events that mediate this unanticipated effect. Dimerization (in the absence of erythropoietin and other cytokines) in human common myeloid progenitors and megakaryocytic erythroid progenitors caused a significant increase in CD34+ cells (p < .01) and induced all stages of erythropoiesis including production of enucleated red blood cells. In contrast, erythropoiesis was not seen with Tpo stimulation. CD34+ cell expansion was the result of increased cell cycling and survival (p < .05). Microarray profiling of CD34+ cells demonstrated that a unique transcriptional pattern is activated in progenitors by F36VMpl dimerization. Ligand-inducible dimerization of intracellular Mpl in human myeloerythroid progenitors induces progenitor expansion and erythropoiesis through molecular mechanisms that are not shared by Tpo stimulation of endogenous Mpl.


Subject(s)
Erythropoiesis , Hematopoietic Stem Cells/metabolism , Intracellular Space/metabolism , Protein Multimerization , Receptors, Thrombopoietin/metabolism , Signal Transduction , Animals , Antigens, CD34/metabolism , Cell Count , Cell Cycle/drug effects , Cell Survival/drug effects , Erythropoiesis/drug effects , Gene Expression Regulation/drug effects , Gene Regulatory Networks/genetics , Hematopoietic Stem Cells/drug effects , Humans , Intracellular Space/drug effects , Mice , Protein Multimerization/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Thrombopoietin/pharmacology , Transduction, Genetic
16.
J Neurooncol ; 103(3): 673-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21038110

ABSTRACT

Children with recurrent high grade gliomas (HGG) have a dismal outcome with a median progression free survival (PFS) of 12 weeks. Adults with recurrent HGG treated with irinotecan and bevacizumab reportedly have a 63% response rate and a median PFS of 23 weeks. There is a paucity of corresponding published pediatric data. We retrospectively reviewed the records of patients less than 21 years of age with recurrent or progressive WHO grade 3-4 gliomas who were treated with bevacizumab containing regimens at our institution between January 2006 and September 2008. We identified eight patients. Six out of eight patients received irinotecan, temozolomide and bevacizumab, one patient received irinotecan and bevacizumab, and one patient received CCNU and bevacizumab. Three patients had stable disease for 30-93 weeks. The remaining five patients developed progressive disease within 17 weeks. The median PFS was 15 weeks and the 6-month PFS was 38%. Contrast enhancing disease responded or remained stable in five out of seven patients whereas non-enhancing disease progressed in three out of four patients. New distant non-enhancing lesions developed in three patients. The most common side effects included diarrhea, vomiting, thrombocytopenia and neutropenia. Bevacizumab was well tolerated when used in combination with conventional chemotherapy (irinotecan in most cases). PFS in our cohort was much shorter and the response rate was inferior in this small cohort of patients when compared with published adult data. However, bevacizumab containing regimens might be effective in a subset of pediatric patients, especially those with predominantly contrast-enhancing disease.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Adolescent , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Bevacizumab , Brain Neoplasms/mortality , Child , Child, Preschool , Choline/metabolism , Disease Progression , Drug Therapy, Combination/methods , Female , Follow-Up Studies , Glioma/mortality , Humans , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/mortality , Time Factors , Young Adult
18.
Pediatr Blood Cancer ; 56(3): 460-2, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21225928

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is characterized by fever, cytopenias, splenomegaly, and hemophagocytosis by macrophages activated by high cytokine levels. Chronic granulomatous disease (CGD) is characterized by recurrent infections, hyperinflammation, and excessive cytokine release. This may predispose patients with CGD to developing HLH during an infection. We conducted a retrospective review of patients with CGD, treated at our institution between 1999 and 2008. Three out of 17 patients developed HLH. Patients with CGD may be at increased risk for developing HLH. Remission of HLH was achieved after treatment with antimicrobials, steroids, and intravenous immunoglobulin This approach to treatment appears to be effective.


Subject(s)
Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/therapy , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/therapy , Anti-Bacterial Agents/therapeutic use , Child , Humans , Immunoglobulins, Intravenous/therapeutic use , Lymphohistiocytosis, Hemophagocytic/diagnosis , Male , Prognosis , Retrospective Studies , Steroids/therapeutic use
19.
Front Immunol ; 12: 716661, 2021.
Article in English | MEDLINE | ID: mdl-34394122

ABSTRACT

Although metabolic pathways have been shown to control differentiation and activation in peripheral T cells, metabolic studies on thymic T cell development are still lacking, especially in human tissue. In this study, we use transcriptomics and extracellular flux analyses to investigate the metabolic profiles of primary thymic and in vitro-derived mouse and human thymocytes. Core metabolic pathways, specifically glycolysis and oxidative phosphorylation, undergo dramatic changes between the double-negative (DN), double-positive (DP), and mature single-positive (SP) stages in murine and human thymus. Remarkably, despite the absence of the complex multicellular thymic microenvironment, in vitro murine and human T cell development recapitulated the coordinated decrease in glycolytic and oxidative phosphorylation activity between the DN and DP stages seen in primary thymus. Moreover, by inducing in vitro T cell differentiation from Rag1-/- mouse bone marrow, we show that reduced metabolic activity at the DP stage is independent of TCR rearrangement. Thus, our findings suggest that highly conserved metabolic transitions are critical for thymic T cell development.


Subject(s)
Cell Differentiation , Energy Metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Animals , Biological Evolution , Biomarkers , Cell Line , Computational Biology/methods , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Lymphopoiesis , Metabolome , Metabolomics/methods , Mice , Organoids , Thymocytes/immunology , Tissue Culture Techniques
20.
Front Oncol ; 11: 766888, 2021.
Article in English | MEDLINE | ID: mdl-34926269

ABSTRACT

The PI3K/Akt pathway-and in particular PI3Kδ-is known for its role in drug resistant B-cell acute lymphoblastic leukemia (B-ALL) and it is often upregulated in refractory or relapsed B-ALL. Myc proteins are transcription factors responsible for transcribing pro-proliferative genes and c-Myc is often overexpressed in cancers. The chromatin regulator BRD4 is required for expression of c-Myc in hematologic malignancies including B-ALL. Previously, combination of BRD4 and PI3K inhibition with SF2523 was shown to successfully decrease Myc expression. However, the underlying mechanism and effect of dual inhibition of PI3Kδ/BRD4 in B-ALL remains unknown. To study this, we utilized SF2535, a novel small molecule dual inhibitor which can specifically target the PI3Kδ isoform and BRD4. We treated primary B-ALL cells with various concentrations of SF2535 and studied its effect on specific pharmacological on-target mechanisms such as apoptosis, cell cycle, cell proliferation, and adhesion molecules expression usingin vitro and in vivo models. SF2535 significantly downregulates both c-Myc mRNA and protein expression through inhibition of BRD4 at the c-Myc promoter site and decreases p-AKT expression through inhibition of the PI3Kδ/AKT pathway. SF2535 induced apoptosis in B-ALL by downregulation of BCL-2 and increased cleavage of caspase-3, caspase-7, and PARP. Moreover, SF2535 induced cell cycle arrest and decreased cell counts in B-ALL. Interestingly, SF2535 decreased the mean fluorescence intensity (MFI) of integrin α4, α5, α6, and ß1 while increasing MFI of CXCR4, indicating that SF2535 may work through inside-out signaling of integrins. Taken together, our data provide a rationale for the clinical evaluation of targeting PI3Kδ/BRD4 in refractory or relapsed B-ALL using SF2535.

SELECTION OF CITATIONS
SEARCH DETAIL