Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Blood ; 140(14): 1607-1620, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35675516

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) reside in localized microenvironments, or niches, in the bone marrow that provide key signals regulating their activity. A fundamental property of hematopoiesis is the ability to respond to environmental cues such as inflammation. How these cues are transmitted to HSPCs within hematopoietic niches is not well established. Here, we show that perivascular bone marrow dendritic cells (DCs) express a high basal level of Toll-like receptor-1 (TLR1) and TLR2. Systemic treatment with a TLR1/2 agonist induces HSPC expansion and mobilization. It also induces marked alterations in the bone marrow microenvironment, including a decrease in osteoblast activity and sinusoidal endothelial cell numbers. TLR1/2 agonist treatment of mice in which Myd88 is deleted specifically in DCs using Zbtb46-Cre show that the TLR1/2-induced expansion of multipotent HPSCs, but not HSPC mobilization or alterations in the bone marrow microenvironment, is dependent on TLR1/2 signaling in DCs. Interleukin-1ß (IL-1ß) is constitutively expressed in both murine and human DCs and is further induced after TLR1/2 stimulation. Systemic TLR1/2 agonist treatment of Il1r1-/- mice show that TLR1/2-induced HSPC expansion is dependent on IL-1ß signaling. Single-cell RNA-sequencing of low-risk myelodysplastic syndrome bone marrow revealed that IL1B and TLR1 expression is increased in DCs. Collectively, these data suggest a model in which TLR1/2 stimulation of DCs induces secretion of IL-1ß and other inflammatory cytokines into the perivascular niche, which in turn, regulates multipotent HSPCs. Increased DC TLR1/2 signaling may contribute to altered HSPC function in myelodysplastic syndrome by increasing local IL-1ß expression.


Subject(s)
Bone Marrow Cells , Dendritic Cells , Hematopoietic Stem Cells , Interleukin-1beta , Myelodysplastic Syndromes , Animals , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cytokines/metabolism , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Humans , Interleukin-1beta/metabolism , Mice , Myelodysplastic Syndromes/metabolism , Myeloid Differentiation Factor 88/metabolism , RNA/metabolism , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism
2.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35439167

ABSTRACT

Myeloproliferative neoplasms (MPNs) are associated with significant alterations in the bone marrow microenvironment that include decreased expression of key niche factors and myelofibrosis. Here, we explored the contribution of TGF-ß to these alterations by abrogating TGF-ß signaling in bone marrow mesenchymal stromal cells. Loss of TGF-ß signaling in Osx-Cre-targeted MSCs prevented the development of myelofibrosis in both MPLW515L and Jak2V617F models of MPNs. In contrast, despite the absence of myelofibrosis, loss of TGF-ß signaling in mesenchymal stromal cells did not rescue the defective hematopoietic niche induced by MPLW515L, as evidenced by decreased bone marrow cellularity, hematopoietic stem/progenitor cell number, and Cxcl12 and Kitlg expression, and the presence of splenic extramedullary hematopoiesis. Induction of myelofibrosis by MPLW515L was intact in Osx-Cre Smad4fl/fl recipients, demonstrating that SMAD4-independent TGF-ß signaling mediates the myelofibrosis phenotype. Indeed, treatment with a c-Jun N-terminal kinase (JNK) inhibitor prevented the development of myelofibrosis induced by MPLW515L. Together, these data show that JNK-dependent TGF-ß signaling in mesenchymal stromal cells is responsible for the development of myelofibrosis but not hematopoietic niche disruption in MPNs, suggesting that the signals that regulate niche gene expression in bone marrow mesenchymal stromal cells are distinct from those that induce a fibrogenic program.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Primary Myelofibrosis , Bone Marrow/metabolism , Humans , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Neoplasms/metabolism , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
3.
Exp Hematol ; 96: 35-43.e7, 2021 04.
Article in English | MEDLINE | ID: mdl-33556431

ABSTRACT

There is accumulating evidence suggesting that toll-like receptor (TLR) signals play an important role in the regulation of hematopoietic stem/progenitor cells (HSPCs). TLR7/8 stimulation induces the myeloid differentiation of normal HSPCs and acute myeloid leukemia cells. However, the in vivo effect of TLR7/8 agonists on hematopoiesis is largely unknown. Here, we show that, similar to TLR4 and TLR2, treatment with the TLR7/8 agonist R848 induces an expansion of phenotypic hematopoietic stem cells (HSCs) with reduced repopulating potential and HSPC mobilization. In contrast to chronic TLR4 stimulation, treatment with R848 for 5 days did not induce a significant increase in myeloid-biased HSCs. Treatment with R848 results in a significant increase in classic dendritic cells (DCs) in the bone marrow, but a decrease in common dendritic cell progenitors and pre-DCs. Phenotypic analysis of DCs revealed that R848 treatment is associated with altered expression of certain chemokines, activation markers, and migratory receptors. Together, these data indicate that systemic administration of a TLR7/8 agonist has unique effects on hematopoiesis, including the expansion of DCs in the bone marrow, that might have clinical relevance to augment responses to certain immunotherapies, such as cancer vaccines and immune checkpoint blockade.


Subject(s)
Bone Marrow Cells/drug effects , Dendritic Cells/drug effects , Hematopoietic Stem Cells/drug effects , Imidazoles/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , Bone Marrow Cells/cytology , Dendritic Cells/cytology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Imidazoles/administration & dosage , Mice
4.
Cell Rep ; 33(1): 108221, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33027668

ABSTRACT

Synthetic lethality triggered by PARP inhibitor (PARPi) yields promising therapeutic results. Unfortunately, tumor cells acquire PARPi resistance, which is usually associated with the restoration of homologous recombination, loss of PARP1 expression, and/or loss of DNA double-strand break (DSB) end resection regulation. Here, we identify a constitutive mechanism of resistance to PARPi. We report that the bone marrow microenvironment (BMM) facilitates DSB repair activity in leukemia cells to protect them against PARPi-mediated synthetic lethality. This effect depends on the hypoxia-induced overexpression of transforming growth factor beta receptor (TGFßR) kinase on malignant cells, which is activated by bone marrow stromal cells-derived transforming growth factor beta 1 (TGF-ß1). Genetic and/or pharmacological targeting of the TGF-ß1-TGFßR kinase axis results in the restoration of the sensitivity of malignant cells to PARPi in BMM and prolongs the survival of leukemia-bearing mice. Our finding may lead to the therapeutic application of the TGFßR inhibitor in patients receiving PARPis.


Subject(s)
Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Receptors, Transforming Growth Factor beta/metabolism , Smad3 Protein/metabolism , Animals , Humans , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Tumor Microenvironment
5.
PLoS Biol ; 18(8): e3000807, 2020 08.
Article in English | MEDLINE | ID: mdl-32760056

ABSTRACT

Radiotherapy is a commonly used conditioning regimen for bone marrow transplantation (BMT). Cytotoxicity limits the use of this life-saving therapy, but the underlying mechanisms remain poorly defined. Here, we use the syngeneic mouse BMT model to test the hypothesis that lethal radiation damages tissues, thereby unleashing signals that indiscriminately activate the inflammasome pathways in host and transplanted cells. We find that a clinically relevant high dose of radiation causes severe damage to bones and the spleen through mechanisms involving the NLRP3 and AIM2 inflammasomes but not the NLRC4 inflammasome. Downstream, we demonstrate that gasdermin D (GSDMD), the common effector of the inflammasomes, is also activated by radiation. Remarkably, protection against the injury induced by deadly ionizing radiation occurs only when NLRP3, AIM2, or GSDMD is lost simultaneously in both the donor and host cell compartments. Thus, this study reveals a continuum of the actions of lethal radiation relayed by the inflammasome-GSDMD axis, initially affecting recipient cells and ultimately harming transplanted cells as they grow in the severely injured and toxic environment. This study also suggests that therapeutic targeting of inflammasome-GSDMD signaling has the potential to prevent the collateral effects of intense radiation regimens.


Subject(s)
Bone Marrow Cells/radiation effects , Bone Marrow Transplantation , DNA-Binding Proteins/genetics , Inflammasomes/radiation effects , Intracellular Signaling Peptides and Proteins/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phosphate-Binding Proteins/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , DNA-Binding Proteins/deficiency , Female , Femur/cytology , Femur/metabolism , Gene Expression Regulation , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Phosphate-Binding Proteins/deficiency , Pyroptosis/genetics , Pyroptosis/radiation effects , Signal Transduction , Spleen/metabolism , Spleen/pathology , Spleen/radiation effects , Transplantation, Isogeneic , Whole-Body Irradiation , X-Rays
6.
PLoS One ; 15(5): e0233751, 2020.
Article in English | MEDLINE | ID: mdl-32470079

ABSTRACT

Mesenchymal stromal cells are an important component of the bone marrow hematopoietic niche. Prior studies showed that signaling from members of the transforming growth factor (TGF) superfamily in mesenchymal stromal cells is required for normal niche development. Here, we assessed the impact of TGF family signaling on niche maintenance and stress responses by deleting Smad4 in mesenchymal stromal cells at birth, thereby abrogating canonical TGF signaling. No alteration in the number or spatial organization of CXCL12-abundant reticular (CAR) cells, osteoblasts, or adipocytes was observed in Osx-Cre, Smad4fl/fl mice, and expression of key niche factors was normal. Basal hematopoiesis and stress erythropoiesis responses to acute hemolytic anemia were normal. TGF-ß potently inhibits stromal CXCL12 expression in vitro; however, G-CSF induced decreases in bone marrow CXCL12 expression and subsequent hematopoietic stem/progenitor cell mobilization were normal in Osx-Cre, Tgfbr2fl/fl mice, in which all TGF-ß signaling in mesenchymal stromal is lost. Finally, although a prior study showed that TGF-ß enhances recovery from myeloablative therapy, hematopoietic recovery following single or multiple doses of 5-flurauracil were normal in Osx-Cre, Tgfbr2fl/fl mice. Collectively, these data suggest that TGF family member signaling in mesenchymal stromal cells is dispensable for hematopoietic niche maintenance under basal and stress conditions.


Subject(s)
Anemia, Hemolytic/metabolism , Erythropoiesis , Hematopoietic Stem Cells , Mesenchymal Stem Cells , Transforming Growth Factor beta/physiology , Transforming Growth Factors/physiology , Acute Disease , Anemia, Hemolytic/pathology , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Chemokine CXCL12/metabolism , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Stem Cell Niche
7.
PLoS Biol ; 16(11): e3000047, 2018 11.
Article in English | MEDLINE | ID: mdl-30388107

ABSTRACT

Mutated NLRP3 assembles a hyperactive inflammasome, which causes excessive secretion of interleukin (IL)-1ß and IL-18 and, ultimately, a spectrum of autoinflammatory disorders known as cryopyrinopathies of which neonatal-onset multisystem inflammatory disease (NOMID) is the most severe phenotype. NOMID mice phenocopy several features of the human disease as they develop severe systemic inflammation driven by IL-1ß and IL-18 overproduction associated with damage to multiple organs, including spleen, skin, liver, and skeleton. Secretion of IL-1ß and IL-18 requires gasdermin D (GSDMD), which-upon activation by the inflammasomes-translocates to the plasma membrane where it forms pores through which these cytokines are released. However, excessive pore formation resulting from sustained activation of GSDMD compromises membrane integrity and ultimately causes a pro-inflammatory form of cell death, termed pyroptosis. In this study, we first established a strong correlation between NLRP3 inflammasome activation and GSDMD processing and pyroptosis in vitro. Next, we used NOMID mice to determine the extent to which GSDMD-driven pyroptosis influences the pathogenesis of this disorder. Remarkably, all NOMID-associated inflammatory symptoms are prevented upon ablation of GSDMD. Thus, GSDMD-dependent actions are required for the pathogenesis of NOMID in mice.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Cryopyrin-Associated Periodic Syndromes/metabolism , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/metabolism , Cell Membrane/metabolism , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/physiopathology , Inflammasomes/metabolism , Inflammation , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins/genetics , Phosphate-Binding Proteins , Pyroptosis/physiology
8.
Cancer Res ; 78(13): 3510-3521, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29724719

ABSTRACT

Point mutations in the seed sequence of miR-142-3p are present in a subset of acute myelogenous leukemia (AML) and in several subtypes of B-cell lymphoma. Here, we show that mutations associated with AML result both in loss of miR-142-3p function and in decreased miR-142-5p expression. Mir142 loss altered the hematopoietic differentiation of multipotent hematopoietic progenitors, enhancing their myeloid potential while suppressing their lymphoid potential. During hematopoietic maturation, loss of Mir142 increased ASH1L protein expression and consequently resulted in the aberrant maintenance of Hoxa gene expression in myeloid-committed hematopoietic progenitors. Mir142 loss also enhanced the disease-initiating activity of IDH2-mutant hematopoietic cells in mice. Together these data suggest a novel model in which miR-142, through repression of ASH1L activity, plays a key role in suppressing HOXA9/A10 expression during normal myeloid differentiation. AML-associated loss-of-function mutations of MIR142 disrupt this negative signaling pathway, resulting in sustained HOXA9/A10 expression in myeloid progenitors/myeloblasts and ultimately contributing to leukemic transformation.Significance: These findings provide mechanistic insights into the role of miRNAs in leukemogenesis and hematopoietic stem cell function. Cancer Res; 78(13); 3510-21. ©2018 AACR.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors/genetics , Animals , Bone Marrow/pathology , Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , HEK293 Cells , Hematopoietic Stem Cells/pathology , Histone-Lysine N-Methyltransferase/metabolism , Homeobox A10 Proteins , Homeodomain Proteins/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/pathology , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mice, Knockout , Point Mutation , Receptor, EphB2 , Signal Transduction/genetics , Transcription Factors/metabolism
9.
Stem Cells ; 35(1): 3-8, 2017 01.
Article in English | MEDLINE | ID: mdl-27647718

ABSTRACT

Hematopoietic stem cell (HSC) proliferation, self-renewal, and trafficking are dependent, in part, upon signals generated by stromal cells in the bone marrow. Stromal cells are organized into niches that support specific subsets of hematopoietic progenitors. There is emerging evidence that malignant hematopoietic cells may generate signals that alter the number and/or function of specific stromal cell populations in the bone marrow. At least in some cases, the resulting alterations in the bone marrow microenvironment confer a competitive advantage to the malignant HSC and progenitor cells and/or render them less sensitive to chemotherapy. Targeting these signals represents a promising therapeutic strategy for selected hematopoietic malignancies. In this review, we focus on two questions. How do alterations in bone marrow stromal cells arise in hematopoietic malignancies, and how do they contribute to disease pathogenesis? Stem Cells 2017;35:3-8.


Subject(s)
Hematopoietic Stem Cells/cytology , Stem Cell Niche , Animals , Bone Marrow Cells/cytology , Cellular Microenvironment , Disease Progression , Hematologic Neoplasms/pathology , Humans
10.
Cell Death Dis ; 7(10): e2406, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27735949

ABSTRACT

Sarcomas comprise a large heterogeneous group of mesenchymal cancers with limited therapeutic options. When treated with standard cytotoxic chemotherapies, many sarcomas fail to respond completely and rapidly become treatment resistant. A major problem in the investigation and treatment of sarcomas is the fact that no single gene mutation or alteration has been identified among the diverse histologic subtypes. We searched for therapeutically druggable targets that are common to a wide range of histologies and hence could provide alternatives to the conventional chemotherapy. Seven hundred samples comprising 45 separate histologies were examined. We found that almost 90% were arginine auxotrophs, as the expression of argininosuccinate synthetase 1 was lost or significantly reduced. Arginine auxotrophy confers sensitivity to arginine deprivation, leading temporarily to starvation and ultimately to cell survival or death under different circumstances. We showed that, in sarcoma, arginine deprivation therapy with pegylated arginine deiminase (ADI-PEG20) maintains a prolonged state of arginine starvation without causing cell death. However, when starvation was simultaneously prolonged by ADI-PEG20 while inhibited by the clinically available drug chloroquine, sarcoma cells died via necroptosis and apoptosis. These results have revealed a novel metabolic vulnerability in sarcomas and provided the basis for a well-tolerated alternative treatment strategy, potentially applicable to up to 90% of the tumors, regardless of histology.


Subject(s)
Apoptosis/drug effects , Arginine/deficiency , Argininosuccinate Synthase/deficiency , Chloroquine/pharmacology , Sarcoma/enzymology , Sarcoma/pathology , Synthetic Lethal Mutations/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arginine/metabolism , Argininosuccinate Synthase/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Hydrolases/pharmacology , Hydrolases/therapeutic use , Molecular Targeted Therapy , Necrosis , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Prognosis , Sarcoma/drug therapy , Treatment Outcome
11.
Mol Cell ; 58(6): 989-1000, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26051179

ABSTRACT

The regulation of RagA(GTP) is important for amino-acid-induced mTORC1 activation. Although GATOR1 complex has been identified as a negative regulator for mTORC1 by hydrolyzing RagA(GTP), how GATOR1 is recruited to RagA to attenuate mTORC1 signaling remains unclear. Moreover, how mTORC1 signaling is terminated upon amino acid stimulation is also unknown. We show that the recruitment of GATOR1 to RagA is induced by amino acids in an mTORC1-dependent manner. Skp2 E3 ligase drives K63-linked ubiquitination of RagA, which facilitates GATOR1 recruitment and RagA(GTP) hydrolysis, thereby providing a negative feedback loop to attenuate mTORC1 lysosomal recruitment and prevent mTORC1 hyperactivation. We further demonstrate that Skp2 promotes autophagy but inhibits cell size and cilia growth through RagA ubiquitination and mTORC1 inhibition. We thereby propose a negative feedback whereby Skp2-mediated RagA ubiquitination recruits GATOR1 to restrict mTORC1 signaling upon sustained amino acid stimulation, which serves a critical mechanism to maintain proper cellular functions.


Subject(s)
Amino Acids/pharmacology , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , S-Phase Kinase-Associated Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Immunoblotting , Lysine/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Microscopy, Confocal , Models, Biological , NIH 3T3 Cells , Protein Binding/drug effects , RNA Interference , S-Phase Kinase-Associated Proteins/genetics , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...