Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
J Allergy Clin Immunol ; 138(1): 219-228.e9, 2016 07.
Article in English | MEDLINE | ID: mdl-26853280

ABSTRACT

BACKGROUND: Defects in phagocytic nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) function cause chronic granulomatous disease (CGD), a primary immunodeficiency characterized by dysfunctional microbicidal activity and chronic inflammation. OBJECTIVE: We sought to study the effect of chronic inflammation on the hematopoietic compartment in patients and mice with X-linked chronic granulomatous disease (X-CGD). METHODS: We used immunostaining and functional analyses to study the hematopoietic compartment in patients with CGD. RESULTS: An analysis of bone marrow cells from patients and mice with X-CGD revealed a dysregulated hematopoiesis characterized by increased numbers of hematopoietic progenitor cells (HPCs) at the expense of repopulating hematopoietic stem cells (HSCs). In patients with X-CGD, there was a clear reduction in the proportion of HSCs in bone marrow and peripheral blood, and they were also more rapidly exhausted after in vitro culture. In mice with X-CGD, increased cycling of HSCs, expansion of HPCs, and impaired long-term engraftment capacity were found to be associated with high concentrations of proinflammatory cytokines, including IL-1ß. Treatment of wild-type mice with IL-1ß induced enhanced cell-cycle entry of HSCs, expansion of HPCs, and defects in long-term engraftment, mimicking the effects observed in mice with X-CGD. Inhibition of cytokine signaling in mice with X-CGD reduced HPC numbers but had only minor effects on the repopulating ability of HSCs. CONCLUSIONS: Persistent chronic inflammation in patients with CGD is associated with hematopoietic proliferative stress, leading to a decrease in the functional activity of HSCs. Our observations have clinical implications for the development of successful autologous cell therapy approaches.


Subject(s)
Granulomatous Disease, Chronic/metabolism , Hematopoietic Stem Cells/metabolism , Adolescent , Adult , Animals , Biomarkers , Case-Control Studies , Cell Count , Cell Differentiation , Child , Child, Preschool , Colony-Forming Units Assay , Cytokines/metabolism , Cytokines/pharmacology , Disease Models, Animal , Graft Survival , Granulomatous Disease, Chronic/etiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Mice , Mice, Transgenic , Models, Biological , Phenotype , Signal Transduction , Young Adult
2.
Blood Adv ; 6(11): 3513-3528, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35413096

ABSTRACT

Infections are a key source of stress to the hematopoietic system. While infections consume short-lived innate immune cells, their recovery depends on quiescent hematopoietic stem cells (HSCs) with long-term self-renewal capacity. Both chronic inflammatory stress and bacterial infections compromise competitive HSC capacity and cause bone marrow (BM) failure. However, our understanding of how HSCs act during acute and contained infections remains incomplete. Here, we used advanced chimeric and genetic mouse models in combination with pharmacological interventions to dissect the complex nature of the acute systemic response of HSCs to lipopolysaccharide (LPS), a well-established model for inducing inflammatory stress. Acute LPS challenge transiently induced proliferation of quiescent HSCs in vivo. This response was not only mediated via direct LPS-TLR4 conjugation on HSCs but also involved indirect TLR4 signaling in CD115+ monocytic cells, inducing a complex proinflammatory cytokine cascade in BM. Downstream of LPS-TLR4 signaling, the combined action of proinflammatory cytokines such as interferon (IFN)α, IFNγ, tumor necrosis factor-α, interleukin (IL)-1α, IL-1ß, and many others is required to mediate full HSC activation in vivo. Together, our study reveals detailed mechanistic insights into the interplay of proinflammatory cytokine-induced molecular pathways and cell types that jointly orchestrate the complex process of emergency hematopoiesis and HSC activation upon LPS exposure in vivo.


Subject(s)
Lipopolysaccharides , Toll-Like Receptor 4 , Animals , Cytokines/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Lipopolysaccharides/pharmacology , Mice , Toll-Like Receptor 4/metabolism
3.
J Exp Med ; 211(5): 769-79, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24752302

ABSTRACT

The serine protease granzyme B (GzmB) is stored in the granules of cytotoxic T and NK cells and facilitates immune-mediated destruction of virus-infected cells. In this study, we use genetic tools to report novel roles for GzmB as an important regulator of hematopoietic stem cell (HSC) function in response to stress. HSCs lacking the GzmB gene show improved bone marrow (BM) reconstitution associated with increased HSC proliferation and mitochondrial activity. In addition, recipients deficient in GzmB support superior engraftment of wild-type HSCs compared with hosts with normal BM niches. Stimulation of mice with lipopolysaccharide strongly induced GzmB protein expression in HSCs, which was mediated by the TLR4-TRIF-p65 NF-κB pathway. This is associated with increased cell death and GzmB secretion into the BM environment, suggesting an extracellular role of GzmB in modulating HSC niches. Moreover, treatment with the chemotherapeutic agent 5-fluorouracil (5-FU) also induces GzmB production in HSCs. In this situation GzmB is not secreted, but instead causes cell-autonomous apoptosis. Accordingly, GzmB-deficient mice are more resistant to serial 5-FU treatments. Collectively, these results identify GzmB as a negative regulator of HSC function that is induced by stress and chemotherapy in both HSCs and their niches. Blockade of GzmB production may help to improve hematopoiesis in various situations of BM stress.


Subject(s)
Gene Expression Regulation/immunology , Granzymes/metabolism , Hematopoietic Stem Cells/physiology , Signal Transduction/immunology , Stress, Physiological/physiology , Animals , Apoptosis/drug effects , Bone Marrow/physiology , Cell Proliferation/drug effects , Colony-Forming Units Assay , DNA Primers/genetics , Drug Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorouracil/pharmacology , Gene Expression Regulation/drug effects , Granzymes/deficiency , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Mitochondria/metabolism , Mitochondria/physiology , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL