Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Immunol ; 189(4): 1773-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22772446

ABSTRACT

Leukocyte mono-Ig-like receptor 5 (LMIR5, also called CD300b) is an activating receptor expressed in myeloid cells. We have previously demonstrated that T cell Ig mucin 1 works as a ligand for LMIR5 in mouse ischemia/reperfusion injury of the kidneys. In this article, we show that LMIR5 is implicated in LPS-induced sepsis in mice. Notably, neutrophils constitutively released a soluble form of LMIR5 (sLMIR5) through proteolytic cleavage of surface LMIR5. Stimulation with TLR agonists augmented the release of sLMIR5. LPS administration or peritonitis induction increased serum levels of sLMIR5 in mice, which was substantially inhibited by neutrophil depletion. Thus, neutrophils were the main source of LPS-induced sLMIR5 in vivo. On the other hand, i.p. administration of LMIR5-Fc, a surrogate of sLMIR5, bound to resident macrophages (M) and stimulated transient inflammation in mice. Consistently, LMIR5-Fc induced in vitro cytokine production of peritoneal M via its unknown ligand. Interestingly, LMIR5 deficiency profoundly reduced systemic cytokine production and septic mortality in LPS-administered mice, although it did not affect in vitro cytokine production of LPS-stimulated peritoneal M. Importantly, the resistance of LMIR5-deficient mice to LPS- or peritonitis-induced septic death was decreased by LMIR5-Fc administration, implicating sLMIR5 in LPS responses in vivo. Collectively, neutrophil-derived sLMIR5 amplifies LPS-induced lethal inflammation.


Subject(s)
Neutrophils/immunology , Receptors, Immunologic/immunology , Sepsis/immunology , Animals , Blotting, Western , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Sepsis/chemically induced , Solubility , Transfection
2.
FEBS J ; 290(21): 5141-5157, 2023 11.
Article in English | MEDLINE | ID: mdl-37500075

ABSTRACT

Transcription factor RUNX1 plays important roles in hematopoiesis and leukemogenesis. RUNX1 function is tightly controlled through posttranslational modifications, including ubiquitination and acetylation. However, its regulation via ubiquitination, especially proteasome-independent ubiquitination, is poorly understood. We previously identified DTX2 as a RUNX1-interacting E3 ligase using a cell-free AlphaScreen assay. In this study, we examined whether DTX2 is involved in the regulation of RUNX1 using in vitro and ex vivo analyses. DTX2 bound to RUNX1 and other RUNX family members RUNX2 and RUNX3 through their C-terminal region. DTX2-induced RUNX1 ubiquitination did not result in RUNX1 protein degradation. Instead, we found that the acetylation of RUNX1, which is known to enhance the transcriptional activity of RUNX1, was inhibited in the presence of DTX2. Concomitantly, DTX2 reduced the RUNX1-induced activation of an MCSFR luciferase reporter. We also found that DTX2 induced RUNX1 cytoplasmic mislocalization. Moreover, DTX2 overexpression showed a substantial growth-inhibitory effect in RUNX1-dependent leukemia cell lines. Thus, our findings indicate a novel aspect of the ubiquitination and acetylation of RUNX1 that is modulated by DTX2 in a proteosome-independent manner.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Leukemia , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Gene Expression Regulation , Leukemia/genetics
3.
EMBO Mol Med ; 15(1): e15631, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36453131

ABSTRACT

Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme in de novo guanine nucleotide synthesis pathway. Although IMPDH inhibitors are widely used as effective immunosuppressants, their antitumor effects have not been proven in the clinical setting. Here, we found that acute myeloid leukemias (AMLs) with MLL-fusions are susceptible to IMPDH inhibitors in vitro. We also showed that alternate-day administration of IMPDH inhibitors suppressed the development of MLL-AF9-driven AML in vivo without having a devastating effect on immune function. Mechanistically, IMPDH inhibition induced overactivation of Toll-like receptor (TLR)-TRAF6-NF-κB signaling and upregulation of an adhesion molecule VCAM1, which contribute to the antileukemia effect of IMPDH inhibitors. Consequently, combined treatment with IMPDH inhibitors and the TLR1/2 agonist effectively inhibited the development of MLL-fusion AML. These findings provide a rational basis for clinical testing of IMPDH inhibitors against MLL-fusion AMLs and potentially other aggressive tumors with active TLR signaling.


Subject(s)
Leukemia, Myeloid, Acute , Myeloid-Lymphoid Leukemia Protein , Humans , Myeloid-Lymphoid Leukemia Protein/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Enzyme Inhibitors/pharmacology , NF-kappa B , Immunosuppressive Agents/therapeutic use
4.
Exp Hematol ; 112-113: 1-8, 2022.
Article in English | MEDLINE | ID: mdl-35644277

ABSTRACT

Transcription factor RUNX1 plays key roles in the establishment and maintenance of the hematopoietic system. Although RUNX1 has been considered a beneficial tumor suppressor, several recent reports have described the tumor-promoting role of RUNX1 in a variety of hematopoietic neoplasms. In this study, we assessed the effect of RUNX1 depletion in multiple human leukemia cell lines using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system, and confirmed that RUNX1 is in fact required for sustaining their leukemic proliferation. To achieve efficient RUNX1 inhibition in leukemia cells, we then examined the effect of lipid nanoparticle (LNP)-mediated delivery of RUNX1-targeting small interfering (si)RNA using two tumor-tropic LNPs. The LNPs containing RUNX1-targeting siRNA were efficiently incorporated into myeloid and T-cell leukemia cell lines and patient-derived primary human acute myeloid leukemia (AML) cells, downregulated RUNX1 expression, induced cell cycle arrest and apoptosis, and exhibited the growth-inhibitory effect in them. In contrast, the LNPs were not efficiently incorporated into normal cord blood CD34+ cells, indicating their minimum cytotoxicity. Thus, our study highlights RUNX1 as a potential therapeutic target to inhibit leukemogenesis, and provides the LNP-based siRNA delivery as a promising approach to deplete RUNX1 specifically in leukemia cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Liposomes , Nanoparticles , RNA, Small Interfering/genetics
5.
Nat Commun ; 13(1): 4501, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042201

ABSTRACT

KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) represents the most refractory type of childhood leukemia. To uncover the molecular heterogeneity of this disease, we perform RNA sequencing, methylation array analysis, whole exome and targeted deep sequencing on 84 infants with KMT2A-rearranged leukemia. Our multi-omics clustering followed by single-sample and single-cell inference of hematopoietic differentiation establishes five robust integrative clusters (ICs) with different master transcription factors, fusion partners and corresponding stages of B-lymphopoietic and early hemato-endothelial development: IRX-type differentiated (IC1), IRX-type undifferentiated (IC2), HOXA-type MLLT1 (IC3), HOXA-type MLLT3 (IC4), and HOXA-type AFF1 (IC5). Importantly, our deep mutational analysis reveals that the number of RAS pathway mutations predicts prognosis and that the most refractory subgroup of IC2 possesses 100% frequency and the heaviest burden of RAS pathway mutations. Our findings highlight the previously under-appreciated intra- and inter-patient heterogeneity of KMT2A-rearranged infant ALL and provide a rationale for the future development of genomics-guided risk stratification and individualized therapy.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Gene Fusion , Humans , Infant , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics
6.
J Cell Biol ; 175(6): 937-46, 2006 Dec 18.
Article in English | MEDLINE | ID: mdl-17178910

ABSTRACT

STAT transcription factors are tyrosine phosphorylated upon cytokine stimulation and enter the nucleus to activate target genes. We show that Rac1 and a GTPase-activating protein, MgcRacGAP, bind directly to p-STAT5A and are required to promote its nuclear translocation. Using permeabilized cells, we find that nuclear translocation of purified p-STAT5A is dependent on the addition of GTP-bound Rac1, MgcRacGAP, importin alpha, and importin beta. p-STAT3 also enters the nucleus via this transport machinery, and mutant STATs lacking the MgcRacGAP binding site do not enter the nucleus even after phosphorylation. We conclude that GTP-bound Rac1 and MgcRacGAP function as a nuclear transport chaperone for activated STATs.


Subject(s)
Cell Nucleus/metabolism , GTPase-Activating Proteins/metabolism , Protein Transport , STAT5 Transcription Factor/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Blotting, Western , Cell Line , Humans , Immunoprecipitation , Mice , Phosphorylation , Tyrosine/metabolism , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
7.
Sci Rep ; 11(1): 23889, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903756

ABSTRACT

Cell behavior is controlled by complex gene regulatory networks. Although studies have uncovered diverse roles of individual genes, it has been challenging to record or control sequential genetic events in living cells. In this study, we designed two cellular chain reaction systems that enable sequential sgRNA activation in mammalian cells using a nickase Cas9 tethering of a cytosine nucleotide deaminase (nCas9-CDA). In these systems, thymidine (T)-to-cytosine (C) substitutions in the scaffold region of the sgRNA or the TATA box-containing loxP sequence (TATAloxP) are corrected by the nCas9-CDA, leading to activation of the next sgRNA. These reactions can occur multiple times, resulting in cellular chain reactions. As a proof of concept, we established a chain reaction by repairing sgRNA scaffold mutations in 293 T cells. Importantly, the results obtained in yeast or in vitro did not match those obtained in mammalian cells, suggesting that in vivo chain reactions need to be optimized in appropriate cellular contexts. Our system may lay the foundation for building cellular chain reaction systems that have a broad utility in the future biomedical research.


Subject(s)
CRISPR-Cas Systems , DNA Repair , Gene Editing , Mutation , RNA, Guide, Kinetoplastida/genetics , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , HEK293 Cells , Humans , TATA Box/genetics , Thymidine/genetics
8.
Cell Rep ; 36(8): 109576, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433054

ABSTRACT

Paraspeckles are membraneless organelles formed through liquid-liquid phase separation and consist of multiple proteins and RNAs, including NONO, SFPQ, and NEAT1. The role of paraspeckles and the component NONO in hematopoiesis remains unknown. In this study, we show histone modifier ASXL1 is involved in paraspeckle formation. ASXL1 forms phase-separated droplets, upregulates NEAT1 expression, and increases NONO-NEAT1 interactions through the C-terminal intrinsically disordered region (IDR). In contrast, a pathogenic ASXL mutant (ASXL1-MT) lacking IDR does not support the interaction of paraspeckle components. Furthermore, paraspeckles are disrupted and Nono localization is abnormal in the cytoplasm of hematopoietic stem and progenitor cells (HSPCs) derived from ASXL1-MT knockin mice. Nono depletion and the forced expression of cytoplasmic NONO impair the repopulating potential of HSPCs, as does ASXL1-MT. Our study indicates a link between ASXL1 and paraspeckle components in the maintenance of normal hematopoiesis.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Paraspeckles/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Female , HL-60 Cells , HeLa Cells , Hematopoiesis , Humans , Mice , Mice, Transgenic , Paraspeckles/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , THP-1 Cells
9.
Nat Commun ; 12(1): 1826, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758188

ABSTRACT

Somatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.


Subject(s)
Aging/genetics , Clonal Hematopoiesis/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Repressor Proteins/genetics , TOR Serine-Threonine Kinases/metabolism , Aged , Aging/metabolism , Aging/physiology , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Proliferation/genetics , Cells, Cultured , DNA Damage/drug effects , DNA Damage/genetics , Gene Knock-In Techniques , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-akt/metabolism , RNA-Seq , Reactive Oxygen Species/pharmacology , Repressor Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics
10.
Cancer Sci ; 100(9): 1675-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19555392

ABSTRACT

MgcRacGAP plays critical roles in cell division through regulating Rho family small GTPases. As we previously reported, phosphorylation of MgcRacGAP on serine 387 (S387) is induced by Aurora B kinase at the midbody during cytokinesis, which is a critical step of cytokinesis. Phosphorylation of S387-MgcRacGAP converts it from RacGAP to RhoGAP, leading to completion of cytokinesis. Here we show that MgcRacGAP is prominently phosphorylated on S387 even in the interphase of v-Src-transformed NIH3T3 cells in the cytoplasm, but not in the interphase of parental NIH3T3 or H-RasV12-transformed NIH3T3 cells. Interestingly, levels of phosphorylation on S387 (pS387) correlated with soft agar colony-forming abilities of v-Src-transformed NIH3T3 cells. Expression of a phosphorylation-mimic mutant MgcRacGAP-S387D enhanced colony formation of v-Src-transformed NIH3T3 cells. Surprisingly, a Rac1 inhibitor but not kinase inhibitors including Aurora B kinase inhibitor specifically inhibited phosphorylation of S387-MgcRacGAP in v-Src-transformed NIH3T3 cells, suggesting the v-Src-induced pathological positive feedback mechanisms towards Rac1 activation using pS387-MgcRacGAP. These results indicated the difference in the mechanisms between v-Src- and H-RasV12-induced transformation, and should shed some light on pathological roles of disordered phosphorylation of MgcRacGAP at S387 in v-Src-induced cell transformation.


Subject(s)
Cell Transformation, Neoplastic , GTPase-Activating Proteins/metabolism , Oncogene Protein pp60(v-src)/metabolism , Protein Serine-Threonine Kinases/metabolism , Aminoquinolines/pharmacology , Animals , Aurora Kinase B , Aurora Kinases , Blotting, Western , Colony-Forming Units Assay , GTPase-Activating Proteins/antagonists & inhibitors , Mice , NIH 3T3 Cells , Oncogene Protein pp60(v-src)/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Pyrimidines/pharmacology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
11.
Sci Rep ; 8(1): 15873, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367089

ABSTRACT

Mutations in ASXL1 and SETBP1 genes have been frequently detected and often coexist in myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). We previously showed that coexpression of mutant ASXL1 and SETBP1 in hematopoietic progenitor cells induced downregulation of TGFß pathway genes and promoted the development of MDS/AML in a mouse model of bone marrow transplantation. However, whether the repression of TGFß pathway in fact contributes to leukaemogenesis remains unclear. Moreover, mechanisms for the repression of TGFß pathway genes in ASXL1/SETBP1-mutated MDS/AML cells have not been fully understood. In this study, we showed that expression of a constitutively active TGFß type I receptor (ALK5-TD) inhibited leukaemic proliferation of MDS/AML cells expressing mutant ASXL1/SETBP1. We also found aberrantly reduced acetylation of several lysine residues on histone H3 and H4 around the promoter regions of multiple TGFß pathway genes. The histone deacetylase (HDAC) inhibitor vorinostat reversed histone acetylation at these promoter regions, and induced transcriptional derepression of the TGFß pathway genes. Furthermore, vorinostat showed robust growth-inhibitory effect in cells expressing mutant ASXL1, whereas it showed only a marginal effect in normal bone marrow cells. These data indicate that HDAC inhibitors will be promising therapeutic drugs for MDS and AML with ASXL1 and SETBP1 mutations.


Subject(s)
Carrier Proteins/genetics , Histones/metabolism , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transforming Growth Factor beta/metabolism , Acetylation/drug effects , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Histones/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mutation , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Survival Rate , Transcriptional Activation/drug effects , Vorinostat/pharmacology , Vorinostat/therapeutic use
12.
Bone ; 37(3): 323-36, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15993670

ABSTRACT

Levels of HtrA1 protein in cartilage have been reported to elevate in joints of human osteoarthritis patients. To understand roles of HtrA1 in normal osteogenesis as well as in pathogenesis of arthritis, we examine HtrA1 expression pattern during bone and cartilage development and in articular cartilage affected by experimental arthritis. HtrA1 is not expressed in mesenchymal or cartilage condensations before initiation of ossification. When ossification begins in the condensations, the expression of HtrA1 starts in chondrocytes undergoing hypertrophic differentiation near the ossification center. Hypertrophic chondrocytes found in adult articular cartilage and epiphyseal growth plates also express HtrA1. When arthritis is induced by injection of anti-collagen antibodies and lipopolysaccharide, resting chondrocytes proceed to terminal hypertrophic differentiation and start expressing HtrA1. These data suggest that hypertrophic change induces HtrA1 expression in chondrocytes both in normal and pathological conditions. HtrA1 has been reported to inhibit TGF-beta signaling. We show that HtrA1 digests major components of cartilage, such as aggrecan, decorin, fibromodulin, and soluble type II collagen. HtrA1 may, therefore, promote degeneration of cartilage by inducing terminal hypertrophic chondrocyte differentiation and by digesting cartilage matrix though its TGF-beta inhibitory activity and protease activity, respectively. In bone, active cuboidal osteoblasts barely express HtrA1, but osteoblasts which flatten and adhere to the bone matrix and osteocytes embedded in bone are strongly positive for HtrA1 production. The bone matrix shows a high level of HtrA1 protein deposition akin to that of TGF-beta, suggesting a close functional interaction between TGF-beta and HtrA1.


Subject(s)
Arthritis, Experimental/enzymology , Bone and Bones/enzymology , Cartilage/enzymology , Joints/enzymology , Serine Endopeptidases/metabolism , Aging/physiology , Animals , Animals, Newborn , Apoptosis , Arthritis, Experimental/chemically induced , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Collagen/pharmacology , Disease Progression , Enzyme Induction , Female , High-Temperature Requirement A Serine Peptidase 1 , Mice , Mice, Inbred BALB C , Serine Endopeptidases/genetics
13.
J Biochem ; 131(1): 87-96, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11754739

ABSTRACT

Chitosanase is the glycolytic enzyme that hydrolyzes the glucosamine GlcN-GlcN bonds of chitosan. To determine the catalytically important residues of chitosanase A (ChoA) from Matsuebacter chitosanotabidus 3001, we performed both site-directed and random mutagenesis of choA, obtaining 31 mutants. These mutations indicated that Glu-121 and Glu-141 were catalytically important residues, as mutation at these sites to Ala or Asp drastically decreased the enzymatic activity to 0.1-0.3% of that of the wild type enzyme. Glu-141 mutations remarkably decreased kinetic constant k(cat) for hydrolysis of chitosan, meanwhile Glu-121 mutations decreased the activities to undeterminable levels, precluding parameter analysis. No hydrolysis of (GlcN)(6) was observed with the purified Glu-121 mutant and extremely slow hydrolysis with the Glu-141 mutant. We also found that Asp-139, Asp-148, Arg-150, Gly-151, Asp-164, and Gly-280 were important residues for enzymatic activities, although they are not directly involved in catalysis. In addition, mutation of any of the six cysteine residues of ChoA abrogated the enzymatic activity, and Cys-136 and Cys-231 were found to form a disulfide bond. In support of the significance of the disulfide bond of ChoA, chitosanase activity was impaired on incubation with a reducing agent. Thus, ChoA from M. chitosanotabidus 3001 uses two glutamic acid residues as putative catalytic residues and has at least one disulfide bond.


Subject(s)
Bacterial Proteins , Catalytic Domain/physiology , Disulfides/metabolism , Glutamic Acid/metabolism , Glycoside Hydrolases/metabolism , Proteobacteria/enzymology , Catalytic Domain/genetics , Circular Dichroism , Cysteine/chemistry , Cysteine/genetics , Disulfides/chemistry , Glutamic Acid/chemistry , Glutamic Acid/genetics , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Kinetics , Mutagenesis , Oligosaccharides/metabolism , Point Mutation , Sequence Alignment
14.
J Clin Invest ; 123(11): 4627-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24216483

ABSTRACT

Recurrent mutations in the gene encoding additional sex combs-like 1 (ASXL1) are found in various hematologic malignancies and associated with poor prognosis. In particular, ASXL1 mutations are common in patients with hematologic malignancies associated with myelodysplasia, including myelodysplastic syndromes (MDSs), and chronic myelomonocytic leukemia. Although loss-of-function ASXL1 mutations promote myeloid transformation, a large subset of ASXL1 mutations is thought to result in stable truncation of ASXL1. Here we demonstrate that C-terminal­truncating Asxl1 mutations (ASXL1-MTs) inhibited myeloid differentiation and induced MDS-like disease in mice. ASXL1-MT mice displayed features of human-associated MDS, including multi-lineage myelodysplasia, pancytopenia, and occasional progression to overt leukemia. ASXL1-MT resulted in derepression of homeobox A9 (Hoxa9) and microRNA-125a (miR-125a) expression through inhibition of polycomb repressive complex 2­mediated (PRC2-mediated) methylation of histone H3K27. miR-125a reduced expression of C-type lectin domain family 5, member a (Clec5a), which is involved in myeloid differentiation. In addition, HOXA9 expression was high in MDS patients with ASXL1-MT, while CLEC5A expression was generally low. Thus, ASXL1-MT­induced MDS-like disease in mice is associated with derepression of Hoxa9 and miR-125a and with Clec5a dysregulation. Our data provide evidence for an axis of MDS pathogenesis that implicates both ASXL1 mutations and miR-125a as therapeutic targets in MDS.


Subject(s)
Mutant Proteins/genetics , Mutation , Myelodysplastic Syndromes/genetics , Repressor Proteins/genetics , Animals , Cell Line , Disease Models, Animal , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Lectins, C-Type/genetics , Methylation , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myelopoiesis/genetics , Peptide Fragments/genetics , Receptors, Cell Surface/genetics
15.
Dev Growth Differ ; 46(3): 257-74, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15206957

ABSTRACT

The expression of mouse HtrA1 is developmentally regulated and restricted in embryo tissues which depend largely on TGF-beta signaling for their differentiation. We examined whether mouse HtrA3, another HtrA family member very close to HtrA1, shows similar expression patterns. HtrA3 and -1 were expressed mostly in the same embryonic organs but exhibited complementary patterns in various tissues; the lens epithelial cells in day 12.5 embryo expressed HtrA3 whereas the ciliary body and pigment retina expressed HtrA1. In the vertebrae of day 14.5 embryo, HtrA3 was expressed in the tail region, but HtrA1 was predominantly expressed in the thoracic and lumbar regions. Similar to HtrA1, HtrA3 bound to various TGF-beta proteins and inhibited the signaling of BMP-4, -2 and TGF-beta 1. HtrA3 did not inhibit signaling originated from a constitutively active BMP receptor, indicating that the inhibition occurred upstream of the cell surface receptor. HtrA3 also showed proteolytic activities indistinguishable from those of HtrA1 toward beta-casein and some extracellular matrix (ECM) proteoglycans. The protease activity was absolutely required for the TGF-beta signal inhibition activity. All these data suggest that HtrA3 and -1 have the overlapping biological activities but can function in complementary fashion in certain types of tissues.


Subject(s)
Gene Expression Regulation, Developmental , Serine Endopeptidases/genetics , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Bone and Bones/embryology , Bone and Bones/metabolism , Caseins/metabolism , Gene Expression Profiling , High-Temperature Requirement A Serine Peptidase 1 , Humans , Mice , Protein Binding , Serine Endopeptidases/metabolism , Serine Endopeptidases/physiology , Transforming Growth Factor beta/metabolism
16.
Development ; 131(5): 1041-53, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14973287

ABSTRACT

HtrA1, a member of the mammalian HtrA serine protease family, has a highly conserved protease domain followed by a PDZ domain. Because HtrA1 is a secretory protein and has another functional domain with homology to follistatin, we examined whether HtrA1 functions as an antagonist of Tgfbeta family proteins. During embryo development, mouse HtrA1 was expressed in specific areas where signaling by Tgfbeta family proteins plays important regulatory roles. The GST-pulldown assay showed that HtrA1 binds to a broad range of Tgfbeta family proteins, including Bmp4, Gdf5, Tgfbetas and activin. HtrA1 inhibited signaling by Bmp4, Bmp2, and Tgfbeta1 in C2C12 cells, presumably by preventing receptor activation. Experiments using a series of deletion mutants indicated that the binding activity of HtrA1 required the protease domain and a small linker region preceding it, and that inhibition of Tgfbeta signaling is dependent on the proteolytic activity of HtrA1. Misexpression of HtrA1 near the developing chick eye led to suppression of eye development that was indistinguishable from the effects of noggin. Taken together, these data indicate that HtrA1 protease is a novel inhibitor of Tgfbeta family members.


Subject(s)
Heat-Shock Proteins/metabolism , Periplasmic Proteins/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Avian Proteins/genetics , Base Sequence , Bone Development/genetics , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Cell Line , Chick Embryo , DNA, Complementary/genetics , Eye/embryology , Gene Expression Regulation, Developmental , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Homeodomain Proteins/genetics , Mice , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL