Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Blood ; 140(16): 1748-1750, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36264590
3.
Cancer Discov ; 12(7): 1760-1781, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35405016

ABSTRACT

Leukemic blasts are immune cells gone awry. We hypothesized that dysregulation of inflammatory pathways contributes to the maintenance of their leukemic state and can be exploited as cell-intrinsic, self-directed immunotherapy. To this end, we applied genome-wide screens to discover genetic vulnerabilities in acute myeloid leukemia (AML) cells implicated in inflammatory pathways. We identified the immune modulator IRF2BP2 as a selective AML dependency. We validated AML cell dependency on IRF2BP2 with genetic and protein degradation approaches in vitro and genetically in vivo. Chromatin and global gene-expression studies demonstrated that IRF2BP2 represses IL1ß/TNFα signaling via NFκB, and IRF2BP2 perturbation results in an acute inflammatory state leading to AML cell death. These findings elucidate a hitherto unexplored AML dependency, reveal cell-intrinsic inflammatory signaling as a mechanism priming leukemic blasts for regulated cell death, and establish IRF2BP2-mediated transcriptional repression as a mechanism for blast survival. SIGNIFICANCE: This study exploits inflammatory programs inherent to AML blasts to identify genetic vulnerabilities in this disease. In doing so, we determined that AML cells are dependent on the transcriptional repressive activity of IRF2BP2 for their survival, revealing cell-intrinsic inflammation as a mechanism priming leukemic blasts for regulated cell death. See related commentary by Puissant and Medyouf, p. 1617. This article is highlighted in the In This Issue feature, p. 1599.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Inflammation/genetics , Leukemia, Myeloid, Acute/genetics , NF-kappa B/metabolism , Signal Transduction
4.
Blood ; 128(17): 2130-2134, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27581357

ABSTRACT

Favorable-risk human acute myeloid leukemia (AML) engrafts poorly in currently used immunodeficient mice, possibly because of insufficient environmental support of these leukemic entities. To address this limitation, we here transplanted primary human AML with isolated nucleophosmin (NPM1) mutation and AML with inv(16) in mice in which human versions of genes encoding cytokines important for myelopoiesis (macrophage colony-stimulating factor [M-CSF], interleukin-3, granulocyte-macrophage colony-stimulating factor, and thrombopoietin) were knocked into their respective mouse loci. NPM1mut AML engrafted with higher efficacy in cytokine knock-in (KI) mice and showed a trend toward higher bone marrow engraftment levels in comparison with NSG mice. inv(16) AML engrafted with high efficacy and was serially transplantable in cytokine KI mice but, in contrast, exhibited virtually no engraftment in NSG mice. Selected use of cytokine KI mice revealed that human M-CSF was required for inv(16) AML engraftment. Subsequent transcriptome profiling in an independent AML patient study cohort demonstrated high expression of M-CSF receptor and enrichment of M-CSF inducible genes in inv(16) AML cases. This study thus provides a first xenotransplantation mouse model for and informs on the disease biology of inv(16) AML.


Subject(s)
Disease Models, Animal , Leukemia, Myeloid, Acute , Neoplasm Transplantation/methods , Transplantation, Heterologous/methods , Animals , Chromosome Aberrations , Chromosomes, Human, Pair 16/genetics , Cytokines , Gene Knock-In Techniques , Heterografts , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mutation , Nuclear Proteins/genetics , Nucleophosmin
5.
Blood ; 128(18): 2253-2257, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27574191

ABSTRACT

Although the molecular pathways that cause acute myeloid leukemia (AML) are increasingly well understood, the pathogenesis of peripheral blood cytopenia, a major cause of AML mortality, remains obscure. A prevailing assumption states that AML spatially displaces nonleukemic hematopoiesis from the bone marrow. However, examining an initial cohort of 223 AML patients, we found no correlation between bone marrow blast content and cytopenia, questioning the displacement theory. Measuring serum concentration of thrombopoietin (TPO), a key regulator of hematopoietic stem cells and megakaryocytes, revealed loss of physiologic negative correlation with platelet count in AML cases with blasts expressing MPL, the thrombopoietin (scavenging) receptor. Mechanistic studies demonstrated that MPLhi blasts could indeed clear TPO, likely therefore leading to insufficient cytokine levels for nonleukemic hematopoiesis. Microarray analysis in an independent multicenter study cohort of 437 AML cases validated MPL expression as a central predictor of thrombocytopenia and neutropenia in AML. Moreover, t(8;21) AML cases demonstrated the highest average MPL expression and lowest average platelet and absolute neutrophil counts among subgroups. Our work thus explains the pathophysiology of peripheral blood cytopenia in a relevant number of AML cases.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Neutropenia/physiopathology , Receptors, Thrombopoietin/biosynthesis , Thrombocytopenia/physiopathology , Cohort Studies , Gene Knock-In Techniques , Hematopoiesis/physiology , Heterografts , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/metabolism , Neutropenia/etiology , Thrombocytopenia/etiology , Thrombopoietin/blood , Transcriptome
6.
J Neurooncol ; 117(2): 303-10, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24504499

ABSTRACT

Brain metastases treated with stereotactic radiosurgery may show delayed enlargement on post-treatment imaging that is of ambiguous etiology. Histopathologic interpretation of brain specimens is often challenging due to the presence of significant radiation effects admixed with irradiated residual tumor of indeterminate viability. The purpose of this study was to assess the impact of histologic findings on clinical outcomes following resection of these lesions. Between 2004 and 2010, 690 patients with brain metastases were enrolled in a prospective gamma knife data repository, and lesions requiring excision were identified. Tissue specimens were divided into four groups based on the ratio of treatment related inflammatory changes (TRIC) to tumor cells, and subsequently patient outcomes were assessed. Of 2,583 metastases treated, 36 were excised due to symptomatic enlargement. Only TRIC, without residual evidence of tumor, was seen in 36 % (13/36) of specimens. Resection of these lesions resulted in 100 % local control in follow-up. Of the remaining 23 lesions that contained any viable-appearing tumor within the resected specimen, 8 recurred after resection. Lesions that enlarged in the first 6 months were more likely to contain higher amounts of residual tumor cells. Patients with even <2 % tumors cells on excision had significantly worse local control (75 vs. 100 %, p = 0.024) and survival (HR 0.27, p = 0.029) compared with those patients with exclusively TRIC. In summary, our findings underscore the importance of surgically obtaining tissue in a method that facilitates complete lesional interpretive histology in order to accurately guide ongoing patient management.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/surgery , Neoplasm Recurrence, Local/pathology , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Radiosurgery
7.
J Neurooncol ; 117(1): 33-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24442402

ABSTRACT

Progressively enlarging encephalopathic changes are now well-documented effects of gamma knife radiosurgery (GKRS) occurring ~3-30 months after treatment of both benign and malignant brain lesions. These changes can be variably associated with inflammatory demyelination and necrosis and/or recurrent tumor. While radiographic differentiation between encephalopathic changes and recurrent tumor is of high clinical relevance, confident interpretation of post-radiosurgery imaging changes can be challenging or even impossible in some cases. Gadolinium-enhanced MRI of these lesions reveals variable amounts of enhancing and non-enhancing components within these lesions that have not been clearly correlated with structural-pathologic change. The goal of this study is to characterize the histopathological changes associated with enhancing versus non-enhancing regions of GKRS-treated lesions. MRI images of patients with progressive, etiologically ambiguous brain lesions following GKRS were reviewed prior to explorative neurosurgery. Chosen for this study were lesions in which distinct areas of enhancement and non-enhancement of at least 5 mm in size could be identified (n = 16). Distinctly enhancing and non-enhancing areas were separately biopsied and histologically evaluated. Only cases with uniform histological results are presented in this study. Enhancing and non-enhancing areas in post GKRS lesions represent separate pathological changes. Radiographically enhancing areas correlate either with recurrent tumor growth or inflammatory demyelinating changes. Lack of radiographic enhancement correlates with coagulative necrosis if the sample is taken from the center of the lesion, or with reactive astrocytosis if the sample is taken from the periphery. Separate biopsy of enhancing and non-enhancing regions of post-GKRS encephalopathy was able to confirm that the pathologies in these areas are distinct. These findings allow for better-informed correlation of histological and radiological changes and a better understanding of post-treatment tissue pathology.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/surgery , Brain/pathology , Radiosurgery/adverse effects , Adult , Aged , Brain/surgery , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Female , Gliosis/etiology , Gliosis/pathology , Humans , Magnetic Resonance Imaging , Male , Meningioma/etiology , Meningioma/pathology , Middle Aged , Necrosis/etiology , Necrosis/pathology , Neoplasm Recurrence, Local
8.
Immunity ; 38(2): 296-308, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333075

ABSTRACT

Macrophages frequently infiltrate tumors and can enhance cancer growth, yet the origins of the macrophage response are not well understood. Here we address molecular mechanisms of macrophage production in a conditional mouse model of lung adenocarcinoma. We report that overproduction of the peptide hormone Angiotensin II (AngII) in tumor-bearing mice amplifies self-renewing hematopoietic stem cells (HSCs) and macrophage progenitors. The process occurred in the spleen but not the bone marrow, and was independent of hemodynamic changes. The effects of AngII required direct hormone ligation on HSCs, depended on S1P(1) signaling, and allowed the extramedullary tissue to supply new tumor-associated macrophages throughout cancer progression. Conversely, blocking AngII production prevented cancer-induced HSC and macrophage progenitor amplification and thus restrained the macrophage response at its source. These findings indicate that AngII acts upstream of a potent macrophage amplification program and that tumors can remotely exploit the hormone's pathway to stimulate cancer-promoting immunity.


Subject(s)
Adenocarcinoma/metabolism , Angiotensin II/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Spleen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Angiotensin II/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Communication , Cell Movement , Cell Proliferation , Gene Expression , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lysophospholipids/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spleen/pathology , Tumor Burden
9.
Proc Natl Acad Sci U S A ; 109(7): 2491-6, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308361

ABSTRACT

Tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) can control cancer growth and exist in almost all solid neoplasms. The cells are known to descend from immature monocytic and granulocytic cells, respectively, which are produced in the bone marrow. However, the spleen is also a recently identified reservoir of monocytes, which can play a significant role in the inflammatory response that follows acute injury. Here, we evaluated the role of the splenic reservoir in a genetic mouse model of lung adenocarcinoma driven by activation of oncogenic Kras and inactivation of p53. We found that high numbers of TAM and TAN precursors physically relocated from the spleen to the tumor stroma, and that recruitment of tumor-promoting spleen-derived TAMs required signaling of the chemokine receptor CCR2. Also, removal of the spleen, either before or after tumor initiation, reduced TAM and TAN responses significantly and delayed tumor growth. The mechanism by which the spleen was able to maintain its reservoir capacity throughout tumor progression involved, in part, local accumulation in the splenic red pulp of typically rare extramedullary hematopoietic stem and progenitor cells, notably granulocyte and macrophage progenitors, which produced CD11b(+) Ly-6C(hi) monocytic and CD11b(+) Ly-6G(hi) granulocytic cells locally. Splenic granulocyte and macrophage progenitors and their descendants were likewise identified in clinical specimens. The present study sheds light on the origins of TAMs and TANs, and positions the spleen as an important extramedullary site, which can continuously supply growing tumors with these cells.


Subject(s)
Macrophages/immunology , Neoplasms/pathology , Neutrophils/immunology , Animals , Humans , Mice , Neoplasms/immunology , Spleen/immunology , Spleen/pathology
10.
Science ; 335(6068): 597-601, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22245738

ABSTRACT

Recognition and clearance of a bacterial infection are a fundamental properties of innate immunity. Here, we describe an effector B cell population that protects against microbial sepsis. Innate response activator (IRA) B cells are phenotypically and functionally distinct, develop and diverge from B1a B cells, depend on pattern-recognition receptors, and produce granulocyte-macrophage colony-stimulating factor. Specific deletion of IRA B cell activity impairs bacterial clearance, elicits a cytokine storm, and precipitates septic shock. These observations enrich our understanding of innate immunity, position IRA B cells as gatekeepers of bacterial infection, and identify new treatment avenues for infectious diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , Escherichia coli Infections/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Peritonitis/immunology , Sepsis/immunology , Animals , B-Lymphocyte Subsets/metabolism , Cell Lineage , Cell Separation , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin M/metabolism , Immunophenotyping , Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Lipopolysaccharides , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Parabiosis , Shock, Septic/immunology , Spleen/immunology , Toll-Like Receptor 4/immunology
11.
J Exp Med ; 209(1): 123-37, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22213805

ABSTRACT

Monocytes (Mo) and macrophages (MΦ) are emerging therapeutic targets in malignant, cardiovascular, and autoimmune disorders. Targeting of Mo/MΦ and their effector functions without compromising innate immunity's critical defense mechanisms first requires addressing gaps in knowledge about the life cycle of these cells. Here we studied the source, tissue kinetics, and clearance of Mo/MΦ in murine myocardial infarction, a model of acute inflammation after ischemic injury. We found that a) Mo tissue residence time was surprisingly short (20 h); b) Mo recruitment rates were consistently high even days after initiation of inflammation; c) the sustained need of newly made Mo was fostered by extramedullary monocytopoiesis in the spleen; d) splenic monocytopoiesis was regulated by IL-1ß; and e) the balance of cell recruitment and local death shifted during resolution of inflammation. Depending on the experimental approach, we measured a 24 h Mo/MΦ exit rate from infarct tissue between 5 and 13% of the tissue cell population. Exited cells were most numerous in the blood, liver, and spleen. Abrogation of extramedullary monocytopoiesis proved deleterious for infarct healing and accelerated the evolution of heart failure. We also detected rapid Mo kinetics in mice with stroke. These findings expand our knowledge of Mo/MΦ flux in acute inflammation and provide the groundwork for novel anti-inflammatory strategies for treating heart failure.


Subject(s)
Hematopoiesis, Extramedullary , Macrophages/physiology , Monocytes/cytology , Monocytes/physiology , Myocardial Infarction/physiopathology , Adoptive Transfer , Animals , Biomarkers/metabolism , Cell Death/genetics , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kinetics , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myeloid Cells/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Signal Transduction , Spleen/physiology , Stroke/immunology , Stroke/metabolism , Wound Healing/physiology
12.
Int J Radiat Oncol Biol Phys ; 83(1): 369-75, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22024206

ABSTRACT

PURPOSE: Recently, single-fraction, high-dosed focused radiation therapy such as that administered by Gamma Knife radiosurgery has been used increasingly for the treatment of metastatic brain cancer. Radiation therapy to the brain can cause delayed leukoencephalopathy, which carries its own significant morbidity and mortality. While radiosurgery-induced leukoencephalopathy is known to be clinically different from that following fractionated radiation, pathological differences are not well characterized. In this study, we aimed to integrate novel radiographic and histopathologic observations to gain a conceptual understanding of radiosurgery-induced leukoencephalopathy. METHODS AND MATERIALS: We examined resected tissues of 10 patients treated at Yale New Haven Hospital between January 1, 2009, and June 30, 2010, for brain metastases that had been previously treated with Gamma Knife radiosurgery, who subsequently required surgical management of a symptomatic regrowing lesion. None of the patients showed pathological evidence of tumor recurrence. Clinical and magnetic resonance imaging data for each of the 10 patients were then studied retrospectively. RESULTS: We provide evidence to show that radiosurgery-induced leukoencephalopathy may present as an advancing process that extends beyond the original high-dose radiation field. Neuropathologic examination of the resected tissue revealed traditionally known leukoencephalopathic changes including demyelination, coagulation necrosis, and vascular sclerosis. Unexpectedly, small and medium-sized vessels revealed transmural T-cell infiltration indicative of active vasculitis. CONCLUSIONS: We propose that the presence of a vasculitic component in association with radiation-induced leukoencephalopathy may facilitate the progressive nature of the condition. It may also explain the resemblance of delayed leukoencephalopathy with recurring tumor on virtually all imaging modalities used for posttreatment follow-up.


Subject(s)
Brain Neoplasms/surgery , Brain/radiation effects , Leukoencephalopathies/pathology , Radiosurgery/adverse effects , Vasculitis, Central Nervous System/etiology , Vasculitis, Central Nervous System/pathology , Adult , Aged , Brain/blood supply , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Female , Humans , Leukoencephalopathies/etiology , Magnetic Resonance Imaging/methods , Male , Middle Aged , Radiosurgery/instrumentation , Retrospective Studies
13.
Circulation ; 125(2): 364-74, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22144566

ABSTRACT

BACKGROUND: Atherosclerotic lesions are believed to grow via the recruitment of bone marrow-derived monocytes. Among the known murine monocyte subsets, Ly-6C(high) monocytes are inflammatory, accumulate in lesions preferentially, and differentiate. Here, we hypothesized that the bone marrow outsources the production of Ly-6C(high) monocytes during atherosclerosis. METHODS AND RESULTS: Using murine models of atherosclerosis and fate-mapping approaches, we show that hematopoietic stem and progenitor cells progressively relocate from the bone marrow to the splenic red pulp, where they encounter granulocyte macrophage colony-stimulating factor and interleukin-3, clonally expand, and differentiate to Ly-6C(high) monocytes. Monocytes born in such extramedullary niches intravasate, circulate, and accumulate abundantly in atheromata. On lesional infiltration, Ly-6C(high) monocytes secrete inflammatory cytokines, reactive oxygen species, and proteases. Eventually, they ingest lipids and become foam cells. CONCLUSIONS: Our findings indicate that extramedullary sites supplement the hematopoietic function of the bone marrow by producing circulating inflammatory cells that infiltrate atherosclerotic lesions.


Subject(s)
Antigens, Ly , Atherosclerosis/pathology , Cell Movement/immunology , Hematopoiesis, Extramedullary/immunology , Monocytes/pathology , Animals , Antigens, Ly/biosynthesis , Atherosclerosis/immunology , Bone Marrow , Cell Differentiation , Granulocyte-Macrophage Colony-Stimulating Factor , Hematopoietic Stem Cells/pathology , Inflammation , Interleukin-3 , Mice , Monocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...