Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 18(6): 654-664, 2017 06.
Article in English | MEDLINE | ID: mdl-28414311

ABSTRACT

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Subject(s)
Adipocytes, Beige , Adipogenesis/immunology , Adipose Tissue, White/immunology , Cell Differentiation/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion/immunology , Diet, High-Fat , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Integrin alpha4/genetics , Macrophages/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Obesity/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Subcutaneous Fat , T-Lymphocytes/immunology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Young Adult
2.
Immunity ; 43(6): 1125-36, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682985

ABSTRACT

The interleukin-1 receptor I (IL-1RI) is critical for host resistance to Mycobacterium tuberculosis (Mtb), yet the mechanisms of IL-1RI-mediated pathogen control remain unclear. Here, we show that without IL-1RI, Mtb-infected newly recruited Ly6G(hi) myeloid cells failed to upregulate tumor necrosis factor receptor I (TNF-RI) and to produce reactive oxygen species, resulting in compromised pathogen control. Furthermore, simultaneous ablation of IL-1RI and TNF-RI signaling on either stroma or hematopoietic cells led to early lethality, indicating non-redundant and synergistic roles of IL-1 and TNF in mediating macrophage-stroma cross-talk that was critical for optimal control of Mtb infection. Finally, we show that even in the presence of functional Mtb-specific adaptive immunity, the lack of IL-1α and not IL-1ß led to an exuberant intracellular pathogen replication and progressive non-resolving inflammation. Our study reveals functional interdependence between IL-1 and TNF in enabling Mtb control mechanisms that are critical for host survival.


Subject(s)
Interleukin-1alpha/immunology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis , Receptors, Interleukin-1 Type I/immunology
3.
Blood ; 138(17): 1540-1553, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34086867

ABSTRACT

Thalassemia or sickle cell patients with hereditary persistence of fetal hemoglobin (HbF) have an ameliorated clinical phenotype and, in some cases, can achieve transfusion independence. Inactivation via genome editing of γ-globin developmental suppressors, such as BCL11A or LRF/ZBTB7A, or of their binding sites, have been shown to significantly increase expression of endogenous HbF. To broaden the therapeutic window beyond a single-editing approach, we have explored combinations of cis- and trans-editing targets to enhance HbF reactivation. Multiplex mutagenesis in adult CD34+ cells was well tolerated and did not lead to any detectable defect in the cells' proliferation and differentiation, either in vitro or in vivo. The combination of 1 trans and 1 cis mutation resulted in high editing retention in vivo, coupled with almost pancellular HbF expression in NBSGW mice. The greater in vivo performance of this combination was also recapitulated using a novel helper-dependent adenoviral-CRISPR vector (HD-Ad-dualCRISPR) in CD34+ cells from ß-thalassemia patients transplanted to NBSGW mice. A pronounced increase in HbF expression was observed in human red blood cells in mice with established predominant ß0/ß0-thalassemic hemopoiesis after in vivo injection of the HD-Ad-dualCRISPR vector. Collectively, our data suggest that the combination of cis and trans fetal globin reactivation mutations has the potential to significantly increase HbF both totally and on a per cell basis over single editing and could thus provide significant clinical benefit to patients with severe ß-globin phenotype.


Subject(s)
Antigens, CD34/genetics , Fetal Hemoglobin/genetics , Mutagenesis , beta-Thalassemia/genetics , Adult , Animals , CRISPR-Cas Systems , Cells, Cultured , Gene Editing , Genetic Therapy , Humans , Mice , beta-Thalassemia/therapy , gamma-Globins/genetics
4.
Haematologica ; 108(4): 1053-1067, 2023 04 01.
Article in English | MEDLINE | ID: mdl-35861015

ABSTRACT

Although human cell cultures stimulated with dexamethasone suggest that the glucocorticoid receptor (GR) activates stress erythropoiesis, the effects of GR activation on erythropoiesis in vivo remain poorly understood. We characterized the phenotype of a large cohort of patients with Cushing disease, a rare condition associated with elevated cortisol levels. Results from hypercortisolemic patients with active Cushing disease were compared with those obtained from eucortisolemic patients after remission and from volunteers without the disease. Patients with active Cushing disease exhibited erythrocytosis associated with normal hemoglobin F levels. In addition, their blood contained elevated numbers of GR-induced CD163+ monocytes and a unique class of CD34+ cells expressing CD110, CD36, CD133 and the GR-target gene CXCR4. When cultured, these CD34+ cells generated similarly large numbers of immature erythroid cells in the presence and absence of dexamethasone, with raised expression of the GR-target gene GILZ. Of interest, blood from patients with Cushing disease in remission maintained high numbers of CD163+ monocytes and, although their CD34+ cells had a normal phenotype, these cells were unresponsive to added dexamethasone. Collectively, these results indicate that chronic exposure to excess glucocorticoids in vivo leads to erythrocytosis by generating erythroid progenitor cells with a constitutively active GR. Although remission rescues the erythrocytosis and the phenotype of the circulating CD34+ cells, a memory of other prior changes is maintained in remission.


Subject(s)
Pituitary ACTH Hypersecretion , Polycythemia , Humans , Polycythemia/etiology , Hematopoietic Stem Cells/metabolism , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Dexamethasone/pharmacology , Cells, Cultured
5.
J Neuroinflammation ; 19(1): 103, 2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35488271

ABSTRACT

OBJECTIVE: c-Met, a tyrosine kinase receptor, is the unique receptor for hepatocyte growth factor (HGF). The HGF/c-Met axis is reported to modulate cell migration, maturation, cytokine production, and antigen presentation. Here, we report that CD4+c-Met+ T cells are detected at increased levels in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). METHODS: c-Met expression by CD4+ T cells was analyzed mostly by flow cytometry and by immunohistochemistry from mice and human PBMCs. The in vivo role of CD4+c-Met+ T cells was assessed in EAE. RESULTS: CD4+c-Met+ T cells found in the CNS during EAE peak disease are characterized by a pro-inflammatory phenotype skewed towards a Th1 and Th17 polarization, with enhanced adhesion and transmigration capacities correlating with increased expression of integrin α4 (Itgα4). The adoptive transfer of Itgα4-expressing CD4+Vα3.2+c-Met+ T cells induces increased disease severity compared to CD4+Vα3.2+c-Met- T cells. Finally, CD4+c-Met+ T cells are detected in the brain of MS patients, as well as in the blood with a higher level of Itgα4. These results highlight c-Met as an immune marker of highly pathogenic pro-inflammatory and pro-migratory CD4+ T lymphocytes associated with neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Integrin alpha4 , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Neuroinflammatory Diseases , Th17 Cells
6.
Mol Ther ; 27(12): 2195-2212, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31494053

ABSTRACT

Our goal is the development of in vivo hematopoietic stem cell (HSC) transduction technology with targeted integration. To achieve this, we modified helper-dependent HDAd5/35++ vectors to express a CRISPR/Cas9 specific to the "safe harbor" adeno-associated virus integration site 1 (AAVS1) locus and to provide a donor template for targeted integration through homology-dependent repair. We tested the HDAd-CRISPR + HDAd-donor vector system in AAVS1 transgenic mice using a standard ex vivo HSC gene therapy approach as well as a new in vivo HSC transduction approach that involves HSC mobilization and intravenous HDAd5/35++ injections. In both settings, the majority of treated mice had transgenes (GFP or human γ-globin) integrated into the AAVS1 locus. On average, >60% of peripheral blood cells expressed the transgene after in vivo selection with low-dose O6BG/bis-chloroethylnitrosourea (BCNU). Ex vivo and in vivo HSC transduction and selection studies with HDAd-CRISPR + HDAd-globin-donor resulted in stable γ-globin expression at levels that were significantly higher (>20% γ-globin of adult mouse globin) than those achieved in previous studies with a SB100x-transposase-based HDAd5/35++ system that mediates random integration. The ability to achieve therapeutically relevant transgene expression levels after in vivo HSC transduction and selection and targeted integration make our HDAd5/35++-based vector system a new tool in HSC gene therapy.


Subject(s)
Adenoviridae/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Transduction, Genetic , Transgenes/physiology , Virus Integration , Animals , CRISPR-Cas Systems , Female , Genes, Reporter , Genetic Therapy , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , gamma-Globins/antagonists & inhibitors , gamma-Globins/genetics
7.
Am J Hum Genet ; 98(6): 1146-1158, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259050

ABSTRACT

Ataxia-pancytopenia (AP) syndrome is characterized by cerebellar ataxia, variable hematologic cytopenias, and predisposition to marrow failure and myeloid leukemia, sometimes associated with monosomy 7. Here, in the four-generation family UW-AP, linkage analysis revealed four regions that provided the maximal LOD scores possible, one of which was in a commonly microdeleted chromosome 7q region. Exome sequencing identified a missense mutation (c.2640C>A, p.His880Gln) in the sterile alpha motif domain containing 9-like gene (SAMD9L) that completely cosegregated with disease. By targeted sequencing of SAMD9L, we subsequently identified a different missense mutation (c.3587G>C, p.Cys1196Ser) in affected members of the first described family with AP syndrome, Li-AP. Neither variant is reported in the public databases, both affect highly conserved amino acid residues, and both are predicted to be damaging. With time in culture, lymphoblastic cell lines (LCLs) from two affected individuals in family UW-AP exhibited copy-neutral loss of heterozygosity for large portions of the long arm of chromosome 7, resulting in retention of only the wild-type SAMD9L allele. Newly established LCLs from both individuals demonstrated the same phenomenon. In addition, targeted capture and sequencing of SAMD9L in uncultured blood DNA from both individuals showed bias toward the wild-type allele. These observations indicate in vivo hematopoietic mosaicism. The hematopoietic cytopenias that characterize AP syndrome and the selective advantage for clones that have lost the mutant allele support the postulated role of SAMD9L in the regulation of cell proliferation. Furthermore, we show that AP syndrome is distinct from the dyskeratoses congenita telomeropathies, with which it shares some clinical characteristics.


Subject(s)
Cerebellar Ataxia/genetics , Chromosome Aberrations , Mutation, Missense/genetics , Pancytopenia/genetics , Proteins/genetics , Adolescent , Adult , Cerebellar Ataxia/pathology , Child , Chromosomes, Human, Pair 7/genetics , Exome/genetics , Female , Genetic Linkage , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , Intracellular Signaling Peptides and Proteins , Loss of Heterozygosity , Male , Middle Aged , Pancytopenia/pathology , Pedigree , Tumor Suppressor Proteins/genetics , Young Adult
8.
Nat Methods ; 12(10): 927-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322838

ABSTRACT

Regulatory regions harbor multiple transcription factor (TF) recognition sites; however, the contribution of individual sites to regulatory function remains challenging to define. We describe an approach that exploits the error-prone nature of genome editing-induced double-strand break repair to map functional elements within regulatory DNA at nucleotide resolution. We demonstrate the approach on a human erythroid enhancer, revealing single TF recognition sites that gate the majority of downstream regulatory function.


Subject(s)
Carrier Proteins/genetics , DNA Footprinting/methods , Genomics/methods , Nuclear Proteins/genetics , Regulatory Sequences, Nucleic Acid , Base Sequence , Binding Sites , DNA Breaks, Double-Stranded , DNA Repair , Enhancer Elements, Genetic , Erythrocytes/physiology , Erythropoiesis , Genome, Human , Humans , Mutation , Repressor Proteins , Transcription Factors/metabolism
9.
Blood ; 128(13): 1756-65, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27543439

ABSTRACT

Although the importance of native bone marrow and spleen macrophages in enhancing baseline and stress erythropoiesis has been emphasized over several decades, their kinetic and phenotypic changes during a variety of stress responses have been unclear. Furthermore, whether monocyte-derived recruited macrophages can functionally substitute for inadequate or functionally impaired native macrophages has been controversial and seem to be not only tissue- but also stress-type dependent. To provide further insight into these issues, we made detailed observations at baseline and post-erythroid stress (E-stress) in 2 mouse models with genetically depressed macrophage numbers and compared them to their controls. We documented that, irrespective of the stress-induced (hemolytic or post-erythropoietin [Epo]) treatment, only native CD11b(lo) splenic macrophages expand dramatically post-stress in normal mice without significant changes in the monocyte-derived CD11b(hi) subset. The latter remained a minority and did not change post-stress in 2 genetic models lacking either Spi-C or VCAM-1 with impaired native macrophage proliferative expansion. Although CD11b(lo) macrophages in these mice were one-fifth of normal at their peak response, surprisingly, their erythroid response was not compromised and was similar to controls. Thus, despite the prior emphasis on numerical macrophage reliance to provide functional rescue from E-stress, our data highlight the importance of previously described non-macrophage-dependent pathways activated under certain stress conditions to compensate for low macrophage numbers.


Subject(s)
Erythropoiesis/physiology , Macrophages/physiology , Animals , Bone Marrow/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Erythropoiesis/drug effects , Erythropoietin/administration & dosage , Hemolysis/physiology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylhydrazines/administration & dosage , Recombinant Proteins/administration & dosage , Spleen/pathology , Stress, Physiological , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
10.
Blood ; 128(18): 2206-2217, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27554082

ABSTRACT

Current protocols for hematopoietic stem/progenitor cell (HSPC) gene therapy, involving the transplantation of ex vivo genetically modified HSPCs are complex and not without risk for the patient. We developed a new approach for in vivo HSPC transduction that does not require myeloablation and transplantation. It involves subcutaneous injections of granulocyte-colony-stimulating factor/AMD3100 to mobilize HSPCs from the bone marrow (BM) into the peripheral blood stream and the IV injection of an integrating, helper-dependent adenovirus (HD-Ad5/35++) vector system. These vectors target CD46, a receptor that is uniformly expressed on HSPCs. We demonstrated in human CD46 transgenic mice and immunodeficient mice with engrafted human CD34+ cells that HSPCs transduced in the periphery home back to the BM where they stably express the transgene. In hCD46 transgenic mice, we showed that our in vivo HSPC transduction approach allows for the stable transduction of primitive HSPCs. Twenty weeks after in vivo transduction, green fluorescent protein (GFP) marking in BM HSPCs (Lin-Sca1+Kit- cells) in most of the mice was in the range of 5% to 10%. The percentage of GFP-expressing primitive HSPCs capable of forming multilineage progenitor colonies (colony-forming units [CFUs]) increased from 4% of all CFUs at week 4 to 16% at week 12, indicating transduction and expansion of long-term surviving HSPCs. Our approach was well tolerated, did not result in significant transduction of nonhematopoietic tissues, and was not associated with genotoxicty. The ability to stably genetically modify HSPCs without the need of myeloablative conditioning is relevant for a broader clinical application of gene therapy.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Mobilization/methods , Membrane Cofactor Protein/biosynthesis , Transduction, Genetic/methods , Adenoviridae , Animals , Genetic Vectors/administration & dosage , Hematopoietic Stem Cells , Heterografts , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL
11.
Immunity ; 31(1): 110-21, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19576795

ABSTRACT

The recognition of viral components by host pattern-recognition receptors triggers the induction of the antiviral innate immune response. Toll-like receptor 9 (TLR9) and NLRP3 inflammasome were shown to be the principal specific sensors of viral double-stranded DNA. Here we present evidence that macrophages in vivo activated an innate immune response to a double-stranded DNA virus, adenovirus (Ad), independently of TLR9 or NLRP3 inflammasome. In response to Ad, macrophage-derived IL-1 alpha triggered IL-1RI-dependent production of a defined set of proinflammatory cytokines and chemokines. The IL-1 alpha-mediated response required a selective interaction of virus arginine-glycine-aspartic acid (RGD) motifs with macrophage beta(3) integrins. Thus, these data identify IL-1 alpha-IL-1RI as a key pathway allowing for the activation of proinflammatory responses to the virus, independently of its genomic nucleic acid recognition.


Subject(s)
Carrier Proteins/immunology , Interleukin-1alpha/immunology , Macrophages/immunology , Receptors, Interleukin-1/immunology , Toll-Like Receptor 9/immunology , Adenoviridae/immunology , Adenoviridae/metabolism , Animals , Carrier Proteins/metabolism , Genetic Vectors/immunology , Genetic Vectors/metabolism , Immunity, Innate , Integrin beta3/immunology , Integrin beta3/metabolism , Interleukin-1alpha/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Toll-Like Receptor 9/metabolism
12.
Blood ; 126(1): 89-93, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26019277

ABSTRACT

Elevated fetal hemoglobin (HbF) ameliorates the clinical severity of hemoglobinopathies such as ß-thalassemia and sickle cell anemia. Currently, the only curative approach for individuals under chronic transfusion/chelation support therapy is allogeneic stem cell transplantation. However, recent analyses of heritable variations in HbF levels have provided a new therapeutic target for HbF reactivation: the transcriptional repressor BCL11A. Erythroid-specific BCL11A abrogation is now actively being sought as a therapeutic avenue, but the specific impact of such disruption in humans remains to be determined. Although single nucleotide polymorphisms in BCL11A erythroid regulatory elements have been reported, coding mutations are scarcer. It is thus of great interest that patients have recently been described with microdeletions encompassing BCL11A. These patients display neurodevelopmental abnormalities, but whether they show increased HbF has not been reported. We have examined the hematological phenotype, HbF levels, and erythroid BCL11A expression in 3 such patients. Haploinsufficiency of BCL11A induces only partial developmental γ-globin silencing. Of greater interest is that a patient with a downstream deletion exhibits reduced BCL11A expression and increased HbF. Novel erythroid-specific regulatory elements in this region may be required for normal erythroid BCL11A expression, whereas loss of separate elements in the developing brain may explain the neurological phenotype.


Subject(s)
Carrier Proteins/genetics , Chromosome Deletion , Chromosomes, Human, Pair 2 , Fetal Hemoglobin/metabolism , Nervous System Diseases/genetics , Nuclear Proteins/genetics , Adolescent , Child , Female , Humans , Male , Nervous System Diseases/blood , Repressor Proteins , Up-Regulation
13.
PLoS Pathog ; 10(3): e1004035, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24651866

ABSTRACT

Inflammation is a highly coordinated host response to infection, injury, or cell stress. In most instances, the inflammatory response is pro-survival and is aimed at restoring physiological tissue homeostasis and eliminating invading pathogens, although exuberant inflammation can lead to tissue damage and death. Intravascular injection of adenovirus (Ad) results in virus accumulation in resident tissue macrophages that trigger activation of CXCL1 and CXCL2 chemokines via the IL-1α-IL-1RI signaling pathway. However, the mechanistic role and functional significance of this pathway in orchestrating cellular inflammatory responses to the virus in vivo remain unclear. Resident metallophilic macrophages expressing macrophage receptor with collagenous structure (MARCO+) in the splenic marginal zone (MZ) play the principal role in trapping Ad from the blood. Here we show that intravascular Ad administration leads to the rapid recruitment of Ly-6G+7/4+ polymorphonuclear leukocytes (PMNs) in the splenic MZ, the anatomical compartment that remains free of PMNs when these cells are purged from the bone marrow via a non-inflammatory stimulus. Furthermore, PMN recruitment in the splenic MZ resulted in elimination of virus-containing cells. IL-1α-IL-1RI signaling is only partially responsible for PMN recruitment in the MZ and requires CXCR2, but not CXCR1 signaling. We further found reduced recruitment of PMNs in the splenic MZ in complement C3-deficient mice, and that pre-treatment of IL-1α-deficient, but not wild-type mice, with complement inhibitor CR2-Crry (inhibits all complement pathways at C3 activation) or CR2-fH (inhibits only the alternative complement activation pathway) prior to Ad infection, abrogates PMN recruitment to the MZ and prevents elimination of MARCO+ macrophages from the spleen. Collectively, our study reveals a non-redundant role of the molecular factors of innate immunity--the chemokine-activating IL-1α-IL-1RI-CXCR2 axis and complement--in orchestrating local inflammation and functional cooperation of PMNs and resident macrophages in the splenic MZ, which collectively contribute to limiting disseminated pathogen spread via elimination of virus-containing cells.


Subject(s)
Adenoviridae Infections/immunology , Complement System Proteins/immunology , Inflammation/immunology , Interleukin-1alpha/immunology , Neutrophils/immunology , Adenoviridae/immunology , Animals , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , Macrophages/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology
14.
Blood ; 121(10): 1814-8, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23319569

ABSTRACT

Bone marrow (BM) provides chemoprotection for acute lymphoblastic leukemia (ALL) cells, contributing to lack of efficacy of current therapies. Integrin alpha4 (alpha4) mediates stromal adhesion of normal and malignant B-cell precursors, and according to gene expression analyses from 207 children with minimal residual disease, is highly associated with poorest outcome. We tested whether interference with alpha4-mediated stromal adhesion might be a new ALL treatment. Two models of leukemia were used, one genetic (conditional alpha4 ablation of BCR-ABL1 [p210(+)] leukemia) and one pharmacological (anti-functional alpha4 antibody treatment of primary ALL). Conditional deletion of alpha4 sensitized leukemia cell to nilotinib. Adhesion of primary pre-B ALL cells was alpha4-dependent; alpha4 blockade sensitized primary ALL cells toward chemotherapy. Chemotherapy combined with Natalizumab prolonged survival of NOD/SCID recipients of primary ALL, suggesting adjuvant alpha4 inhibition as a novel strategy for pre-B ALL.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/physiology , Integrin alpha4/chemistry , Neoplasm, Residual/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion , Child , Flow Cytometry , Humans , Integrases/metabolism , Integrin alpha4/genetics , Integrin alpha4/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Natalizumab , Neoplasm, Residual/metabolism , Neoplasm, Residual/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology
15.
bioRxiv ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38370646

ABSTRACT

Prior evidence indicates that the erythroid cellular response to glucocorticoids (GC) has developmental specificity, namely, that developmentally more advanced cells that are undergoing or have undergone fetal to adult globin switching are more responsive to GC-induced expansion. To investigate the molecular underpinnings of this, we focused on the major developmental globin regulator BCL11A. We compared: a) levels of expression and nuclear content of BCL11A in adult erythroid cells upon GC stimulation; b) response to GC of CD34+ cells from patients with BCL11A microdeletions and reduced BCL11A expression, and; c) response to GC of two cellular models (HUDEP-2 and adult CD34+ cells) before and after reduction of BCL11A expression by shRNA. We observed that: a) GC-expanded erythroid cells from a large cohort of blood donors displayed amplified expression and nuclear accumulation of BCL11A; b) CD34+ cells from BCL11A microdeletion patients generated fewer erythroid cells when cultured with GC compared to their parents, while the erythroid expansion of the patients was similar to that of their parents in cultures without GC, and; c) adult CD34+ cells and HUDEP-2 cells with shRNA-depleted expression of BCL11A exhibit reduced expansion in response to GC. In addition, RNA-seq profiling of shRNA-BCL11A CD34+ cells cultured with and without GC was similar (very few differentially expressed genes), while GC-specific responses (differential expression of GILZ and of numerous additional genes) were observed only in controls cells with unperturbed BCL11A expression. These data indicate that BCL11A is an important participant of certain aspects of the stress pathway sustained by GC.

16.
Hum Mutat ; 34(10): 1361-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878091

ABSTRACT

We describe the molecular etiology of ß(+)-thalassemia that is caused by the insertion of the full-length transposable element LINE-1 (L1) into the intron-2 of the ß-globin gene (HBB). The transcript level of the affected ß-globin gene was severely reduced. The remaining transcripts consisted of full-length, correctly processed ß-globin mRNA and a minute amount of three aberrantly spliced transcripts with a decreased half-life due to activation of the nonsense-mediated decay pathway. The lower steady-state amount of mRNA produced by the ß-globin(L1) allele also resulted from a reduced rate of transcription and decreased production of full-length ß-globin primary transcripts. The promoter and enhancer sequences of the ß-globin(L1) allele were hypermethylated; however, treatment with a demethylating agent did not restore the impaired transcription. A histone deacetylase inhibitor partially reactivated the ß-globin(L1) transcription despite permanent ß-globin(L1) promoter CpG methylation. This result indicates that the decreased rate of transcription from the ß-globin(L1) allele is associated with an altered chromatin structure. Therefore, the molecular defect caused by intronic L1 insertion in the ß-globin gene represents a novel etiology of ß-thalassemia.


Subject(s)
Introns , Long Interspersed Nucleotide Elements , Mutagenesis, Insertional , beta-Globins/genetics , beta-Thalassemia/genetics , Adult , Alleles , Alternative Splicing , CpG Islands , DNA Methylation , Female , Gene Expression Regulation , Gene Order , Gene Silencing , Humans , Promoter Regions, Genetic , RNA Stability , Transcription, Genetic
17.
Blood ; 117(3): 975-85, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20956802

ABSTRACT

To delineate the role of specific members of ß1 integrins in stress erythropoiesis in the adult, we compared the response to phenylhydrazine stress in 3 genetically deficient models. The survival of ß1-conditionally deficient mice after phenylhydrazine is severely compromised because of their inability to mount a successful life saving splenic erythroid response, a phenotype reproduced in ß1(Δ/Δ) reconstituted animals. The response of bone marrow to phenylhydrazine-induced stress was, unlike that of spleen, appropriate in terms of progenitor cell expansion and mobilization to peripheral blood although late differentiation defects qualitatively similar to those in spleen were present in bone marrow. In contrast to ß1-deficient mice, α4(Δ/Δ) mice showed only a kinetic delay in recovery and similar to ß1(Δ/Δ), terminal maturation defects in both bone marrow and spleen, which were not present in VCAM-1(Δ/Δ) mice. Convergence of information from these comparative studies lends new insight to the distinct in vivo roles of α4 and α5 integrins in erythroid stress, suggesting that the presence of mainly α5ß1 integrin in all hematopoietic progenitor cells interacting with splenic microenvironmental ligands/cells is instrumental for their survival and accumulation during hemolytic stress, whereas presence of α4 or of both α5 and α4, is important for completion of terminal maturation steps.


Subject(s)
Anemia/physiopathology , Erythropoiesis/physiology , Integrin alpha4/physiology , Integrin alpha5/physiology , Acute Disease , Anemia/chemically induced , Animals , Bone Marrow/metabolism , Bone Marrow Transplantation , Cell Differentiation , Cell Survival , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Flow Cytometry , Immunohistochemistry , Integrin alpha4/genetics , Integrin alpha4/metabolism , Integrin alpha5/genetics , Integrin alpha5/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Integrin beta1/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenylhydrazines , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/physiology
18.
Haematologica ; 98(11): 1769-77, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23812936

ABSTRACT

We have previously reported that ß1(Δ/Δ) mice have a markedly impaired response to hemolytic stress, but the mechanisms of this were unclear. In the present study we explored in detail quantitative, phenotypic and functional aspects of erythropoiesis at homeostasis in a large number of animals for each of 3 murine models with specific ß1 heterodimer integrin deficiencies. We found that, at homeostasis, ß1-deficient mice have a modest uncompensated anemia with ineffective erythropoiesis and decreased red blood cell survival. Mice lacking only α4 integrins (α4ß1/α4ß7) do not share this phenotype. There is an increased tendency for reactive oxygen species accumulation in ß1(Δ/Δ) erythroid cells with decreased anti-oxidant defenses at homeostasis which are exaggerated after stress. Furthermore, expansion of erythroid cells in spleen post-stress is dependent on α5ß1, likely through mechanisms activating focal adhesion kinase complexes that are distinct from α4ß1-mediated responses. In vivo inhibition of focal adhesion kinase activation partially recapitulates the ß1(Δ/Δ) stress response. Mice lacking all α4 and ß1 integrins (double knockouts) had, at homeostasis, the most severe phenotype with selective impairment of erythroid responses. The fact that integrins participate in mitigating stress in erythroid cells through redox activation of distinct signaling pathways by specific integrin heterodimers is a link that has not been appreciated until now.


Subject(s)
Antioxidants/metabolism , Erythroid Cells/metabolism , Homeostasis/physiology , Integrin beta1/metabolism , Protein Multimerization/physiology , Reactive Oxygen Species/metabolism , Animals , Mice , Mice, Knockout , Mice, Transgenic
19.
Haematologica ; 98(3): 414-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22983574

ABSTRACT

The transcription factor TWIST-1 is up-regulated in CD34(+) cells in myelodysplastic syndrome and is involved in resistance to apoptosis. There is evidence that TWIST-1 affects apoptosis via microRNAs (miRs). Expression of miRs was determined in myeloid cell lines and primary CD34(+) marrow cells from patients with myelodysplastic syndrome and healthy donors using NanoString/array and validated by real-time-polymerase chain reaction. Expression levels of miR10a and miR10b were significantly higher in CD34(+) marrow cells from 28 patients with myelodysplastic syndrome than in CD34(+) cells from healthy donors (P=0.05 and P=0.012, respectively). Levels of miR10a/b correlated with TWIST-1 miR levels in CD34(+) myelodysplastic marrow cells (miR10a, R=+0.69, P<0.0001; miR10b, R=+0.56, P=0.0008). Inhibition of miR10a/10b in clonal cells interfered with proliferation and enhanced sensitivity to apoptosis, which involved NF-κB-dependent p53 activation. These data support a role for miR10a/10b in the regulation of apoptosis in myelodysplastic syndrome and suggest the TWIST-1/miR10a/b-axis as a therapeutic target in myelodysplastic syndrome.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Nuclear Proteins/metabolism , Transcription, Genetic , Twist-Related Protein 1/metabolism , Adult , Aged , Apoptosis/genetics , Bone Marrow/pathology , Cell Proliferation , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , NF-kappa B/metabolism , Nuclear Proteins/genetics , Protein Binding , Tumor Suppressor Protein p53/metabolism , Twist-Related Protein 1/genetics , Young Adult
20.
Mol Ther ; 20(1): 230-8, 2012 01.
Article in English | MEDLINE | ID: mdl-21952171

ABSTRACT

The safety and efficacy of hematopoietic stem cell (HSC) mobilization was investigated in adult splenectomized (SPL) and non-SPL patients with thalassemia major, in two clinical trials, using different mobilization modes: granulocyte-colony-stimulating factor (G-CSF)-alone, G-CSF following pretreatment with hydroxyurea (HU), plerixafor-alone. G-CSF-mobilization was both safe and effective in non-SPL patients. However, in SPL patients the procedure resulted in excessive response to G-CSF, expressed as early hyperleukocytosis necessitating significant dose reduction, and suboptimal CD34(+) cells yields. One-month HU-pretreatment prevented hyperleukocytosis and allowed successful CD34(+) cell collections when an optimal washout period was maintained, but it significantly prolonged the mobilization procedure. Plerixafor resulted in rapid and effective mobilization in both SPL and non-SPL patients and was well-tolerated. For gene therapy of thalassemia, G-CSF or Plerixafor could be used as mobilization agents in non-SPL patients whereas Plerixafor appears to be the mobilization agent of choice in SPL adult thalassemics in terms of safety and efficacy.


Subject(s)
Genetic Therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds/therapeutic use , Splenectomy , beta-Thalassemia/therapy , Adult , Antigens, CD34/metabolism , Benzylamines , Cyclams , Female , Granulocyte Colony-Stimulating Factor/adverse effects , Humans , Hydroxyurea/therapeutic use , Immunophenotyping , Leukocyte Count , Leukocytosis/etiology , Male , Splenectomy/adverse effects , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL