Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Fish Shellfish Immunol ; 89: 228-236, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30936046

ABSTRACT

The Runx family is a kind of heteromeric transcription factors, which is defined by the presence of a runt domain. As transcriptional regulator during development and cell fate specification, Runx is best known for its critical roles in hematopoiesis. In the present study, a Runx transcription factor (designed as CgRunx) was identified and characterized from the oyster Crassostrea gigas. The complete coding sequence of CgRunx was of 1638 bp encoding a predicted polypeptide of 545 amino acids with one conserved runt domain, which shared high similarity with other reported Runx proteins. CgRunx was highly expressed in hemocytes, gill and mantle both at the protein and nucleic acid levels. CgRunx protein was localized specifically in the cell nuclei of hemocytes, and distributed at the tubule lumen of gill filament. During the larval developmental stages, the mRNA transcripts of CgRunx gradually increased after fertilization, reached to a relative high level at the 8 cell embryos and the blastula stage of 2-4 hpf (hours post fertilization) (about 40-fold), and peaked at early trochophore larvae (10 hpf) (about 60-fold). Whole-mount immunofluorescence assay further revealed that the abundant immunofluorescence signals of CgRunx distributed through the whole embryo at blastula stage (5 hpf), and progressively reduced with the development to a ring structure around the dorsal region in trochophore larvae (10 hpf). Scattered positive immunoreactivity signals finally appeared in the velum region of D-veliger larvae. After LPS and Vibrio splendidus stimulations, the expression levels of CgRunx mRNA in hemocytes were up-regulated significantly compared with that in the control (0 h), which were 2.98- and 2.46-fold (p < 0.05), 2.67- and 1.5-fold (p < 0.05), 2.36- and 1.38-fold (p < 0.05) at 3 h, 6 h and 12 h, respectively. These results collectively suggested that CgRunx involved in immune response and might participate in larvae hematopoiesis in oyster.


Subject(s)
Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/immunology , Crassostrea/genetics , Crassostrea/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Core Binding Factor alpha Subunits/chemistry , Gene Expression Profiling , Sequence Alignment
2.
Leukemia ; 33(8): 2006-2021, 2019 08.
Article in English | MEDLINE | ID: mdl-30760870

ABSTRACT

Ikaros family zinc finger protein 1 and 3 (IKZF1 and IKZF3) are transcription factors that promote multiple myeloma (MM) proliferation. The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Although IMiDs have been used as first-line drugs for MM, the overall survival of refractory MM patients remains poor and demands the identification of novel agents to potentiate the therapeutic effect of IMiDs. Using an unbiased screen based on mass spectrometry, we identified the Runt-related transcription factor 1 and 3 (RUNX1 and RUNX3) as interactors of IKZF1 and IKZF3. Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Inhibition of RUNXs, via genetic ablation or a small molecule (AI-10-104), results in sensitization of myeloma cell lines and primary tumors to lenalidomide. Thus, RUNX inhibition represents a valuable therapeutic opportunity to potentiate IMiDs therapy for the treatment of multiple myeloma.


Subject(s)
Core Binding Factor alpha Subunits/physiology , Ikaros Transcription Factor/metabolism , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Core Binding Factor alpha Subunits/antagonists & inhibitors , Core Binding Factor alpha Subunits/chemistry , Humans , Peptide Hydrolases/physiology , Ubiquitin-Protein Ligases
3.
EBioMedicine ; 8: 117-131, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27428424

ABSTRACT

Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFß binding partner. CBFß enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFß are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFß and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/metabolism , Neoplasms/metabolism , Allosteric Regulation/drug effects , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , Drug Discovery , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Leukemia , Models, Molecular , Molecular Conformation , Mutation , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction/drug effects , Structure-Activity Relationship
4.
J Exp Med ; 206(11): 2329-37, 2009 Oct 26.
Article in English | MEDLINE | ID: mdl-19841090

ABSTRACT

Runx proteins are essential for hematopoiesis and play an important role in T cell development by regulating key target genes, such as CD4 and CD8 as well as lymphokine genes, during the specialization of naive CD4 T cells into distinct T helper subsets. In regulatory T (T reg) cells, the signature transcription factor Foxp3 interacts with and modulates the function of several other DNA binding proteins, including Runx family members, at the protein level. We show that Runx proteins also regulate the initiation and the maintenance of Foxp3 gene expression in CD4 T cells. Full-length Runx promoted the de novo expression of Foxp3 during inducible T reg cell differentiation, whereas the isolated dominant-negative Runt DNA binding domain antagonized de novo Foxp3 expression. Foxp3 expression in natural T reg cells remained dependent on Runx proteins and correlated with the binding of Runx/core-binding factor beta to regulatory elements within the Foxp3 locus. Our data show that Runx and Foxp3 are components of a feed-forward loop in which Runx proteins contribute to the expression of Foxp3 and cooperate with Foxp3 proteins to regulate the expression of downstream target genes.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Forkhead Transcription Factors/metabolism , Animals , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/metabolism , Feedback, Physiological , Genes, Dominant , Mice , Protein Structure, Tertiary , T-Lymphocytes, Regulatory/metabolism
5.
Blood Cells Mol Dis ; 43(1): 20-6, 2009.
Article in English | MEDLINE | ID: mdl-19349198

ABSTRACT

Runt-related (RUNX) transcription factors are evolutionarily conserved regulators of a number of developmental mechanisms. RUNX proteins often control the balance between proliferation and differentiation and alterations of their functions are associated with different types of cancer and other human pathologies. Moreover, RUNX factors control important steps during the developmental acquisition of mature phenotypes. A number of investigations are beginning to shed light on the involvement of RUNX family members in the development of the nervous system. This review summarizes recent progress in the study of the roles of mammalian RUNX proteins during the differentiation of sensory and motor neurons in the peripheral and central nervous system, respectively. The implications of those findings for RUNX-mediated regulation of hematopoietic development will also be discussed.


Subject(s)
Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Hematopoiesis , Neurogenesis , Animals , Core Binding Factor alpha Subunits/chemistry , Gene Expression Regulation, Developmental , Humans
6.
BMC Evol Biol ; 8: 228, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18681949

ABSTRACT

BACKGROUND: Members of the Runx family of transcriptional regulators, which bind DNA as heterodimers with CBFbeta, are known to play critical roles in embryonic development in many triploblastic animals such as mammals and insects. They are known to regulate basic developmental processes such as cell fate determination and cellular potency in multiple stem-cell types, including the sensory nerve cell progenitors of ganglia in mammals. RESULTS: In this study, we detect and characterize the hitherto unexplored Runx/CBFbeta genes of cnidarians and sponges, two basal animal lineages that are well known for their extensive regenerative capacity. Comparative structural modeling indicates that the Runx-CBFbeta-DNA complex from most cnidarians and sponges is highly similar to that found in humans, with changes in the residues involved in Runx-CBFbeta dimerization in either of the proteins mirrored by compensatory changes in the binding partner. In situ hybridization studies reveal that Nematostella Runx and CBFbeta are expressed predominantly in small isolated foci at the base of the ectoderm of the tentacles in adult animals, possibly representing neurons or their progenitors. CONCLUSION: These results reveal that Runx and CBFbeta likely functioned together to regulate transcription in the common ancestor of all metazoans, and the structure of the Runx-CBFbeta-DNA complex has remained extremely conserved since the human-sponge divergence. The expression data suggest a hypothesis that these genes may have played a role in nerve cell differentiation or maintenance in the common ancestor of cnidarians and bilaterians.


Subject(s)
Cnidaria/genetics , Core Binding Factor alpha Subunits/genetics , Core Binding Factor beta Subunit/genetics , Porifera/genetics , Transcription Factors/genetics , Animals , Cnidaria/classification , Contig Mapping , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/chemistry , Evolution, Molecular , Expressed Sequence Tags , Models, Molecular , Phylogeny , Porifera/classification , Protein Conformation , Transcription Factors/chemistry
7.
J Biochem ; 143(5): 695-709, 2008 May.
Article in English | MEDLINE | ID: mdl-18296717

ABSTRACT

The signal transducers and activators of transcription (STAT) and the Runt-related (Runx) are two of major transcription factor families that play essential roles in lymphocyte development. Although the interaction of Runx2 with STAT1 and STAT3 has been reported before, the interaction between STAT5 and Runx family proteins has not been characterized. In this study, we first showed that STAT5 physically interacts with Runx1, Runx2 and Runx3 by co-immunoprecipitation experiments. The Runt domain of Runx proteins and the DNA-binding domain and alpha-helix loop structure of STAT5 are responsible for the interaction. When expressed in CHO cells, STAT5 inhibits the nuclear localization of Runx proteins and retains them in the cytoplasm. In addition, we showed by reporter assay that the interaction between STAT5 and Runx proteins mutually inhibits their transcriptional activity. Furthermore, Runx proteins inhibit the DNA-binding activity of STAT5. Finally, we found that Runx proteins suppress the transcription of an endogenous STAT5 target gene, cytokine-inducible SH2 protein-1, in an interleukin-3-dependent pro-B cell line, Ba/F3. These results collectively suggested that STAT5 and Runx proteins physically and functionally interact to mutually inhibit their transcriptional activity. Thus, this study implies a potential role of the STAT5-Runx interaction in lymphocyte development.


Subject(s)
Core Binding Factor alpha Subunits/chemistry , Core Binding Factor alpha Subunits/metabolism , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , Animals , CHO Cells , Cell Line , Core Binding Factor alpha Subunits/antagonists & inhibitors , Cricetinae , Cricetulus , Humans , Protein Interaction Domains and Motifs , STAT5 Transcription Factor/antagonists & inhibitors , Transcription, Genetic
8.
Mol Biol Cell ; 19(5): 2113-26, 2008 May.
Article in English | MEDLINE | ID: mdl-18287524

ABSTRACT

Id proteins play important roles in osteogenic differentiation; however, the molecular mechanism remains unknown. In this study, we established that inhibitor of differentiation (Id) proteins, including Id1, Id2, and Id3, associate with core binding factor alpha-1 (Cbfa1) to cause diminished transcription of the alkaline phosphatase (ALP) and osteocalcin (OCL) gene, leading to less ALP activity and osteocalcin (OCL) production. Id acts by inhibiting the sequence-specific binding of Cbfa1 to DNA and by decreasing the expression of Cbfa1 in cells undergoing osteogenic differentiation. p204, an interferon-inducible protein that interacts with both Cbfa1 and Id2, overcame the Id2-mediated inhibition of Cbfa1-induced ALP activity and OCL production. We show that 1) p204 disturbed the binding of Id2 to Cbfa1 and enabled Cbfa1 to bind to the promoters of its target genes and 2) that p204 promoted the translocation from nucleus to the cytoplasm and accelerated the degradation of Id2 by ubiquitin-proteasome pathway during osteogenesis. Nucleus export signal (NES) of p204 is required for the p204-enhanced cytoplasmic translocation and degradation of Id2, because a p204 mutant lacking NES lost these activities. Together, Cbfa1, p204, and Id proteins form a regulatory circuit and act in concert to regulate osteoblast differentiation.


Subject(s)
Cell Differentiation , Core Binding Factor alpha Subunits/metabolism , Helix-Loop-Helix Motifs , Inhibitor of Differentiation Proteins/chemistry , Inhibitor of Differentiation Proteins/metabolism , Nuclear Proteins/metabolism , Osteogenesis , Phosphoproteins/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Core Binding Factor alpha Subunits/chemistry , Female , Humans , Mice , Mice, Inbred BALB C , Nuclear Export Signals , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/drug effects , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Transforming Growth Factor beta/pharmacology , Ubiquitin/metabolism
9.
J Biomol Struct Dyn ; 24(4): 343-58, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17206850

ABSTRACT

The Runt domain proteins are eukaryotic transcription factors that regulate major developmental pathways. All members of this family contain a highly-conserved sequence-specific DNA binding domain: the Runt domain (RD). Structural and biochemical studies have shown that the Runt domain undergoes a conformational transition upon binding to DNA and that this process is regulated by an unrelated partner protein CBFbeta that enhances the DNA binding affinity of RD. Most of the reported studies on the Runt domain transcription factors were performed on proteins from mammals and Drosophila whereas very little has been known about the C. elegans RD protein, RUN, which provides the simplest model system for understanding the function of this class of transcription factors. We performed computational studies on RD domains from various species including C. elegans, Drosophila, and human, using the atom-atom contact surface area scoring method. The scoring analysis indicates that the DNA binding regulation of the C. elegans RD protein (CeRD) occurs via its interaction with a CBFbeta-like partner, as found for the human proteins, whereas a different mode of regulation may occur in the Drosophila system. Sequence, secondary structure and fold analyses of a putative CBFbeta protein identified in the C. elegans genome, CeCBFbeta, sharing a 22% identity with the human protein, predict a similar structure of this protein to that of the human CBFbeta protein. We produced the C. elegans proteins CeRD and CeCBFbeta in bacteria and confirmed their physical interaction as well as cross interactions with the corresponding human proteins. We also confirmed the structural similarity of CBFbeta and CeCBFbeta by circular dichroism analysis. The combined results suggest that a similar mechanism of regulation operates for the human and the C. elegans RD proteins despite the low sequence identity between their CBFbeta proteins and the evolutionary distance between the two systems.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Core Binding Factor alpha Subunits/genetics , Amino Acid Sequence , Animals , Binding Sites , Caenorhabditis elegans Proteins/chemistry , Cloning, Molecular , Core Binding Factor alpha Subunits/chemistry , DNA, Complementary/genetics , Humans , Molecular Sequence Data , Mutagenesis , Protein Biosynthesis , Protein Conformation , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
10.
Crit Rev Eukaryot Gene Expr ; 15(3): 243-54, 2005.
Article in English | MEDLINE | ID: mdl-16390320

ABSTRACT

The mammalian RUNX protein family comprises three transcription factorsRUNX1, RUNX2, and RUNX3. RUNX1 is involved in hematopoiesis, RUNX2 has multiple roles in osteogenesis and RUNX3 is associated with neural and gut development. In addition, all RUNX proteins are expressed during chondrogenesis, the process by which cartilage is formed. This review describes the involvement of Runx proteins in chondrogenesis, delineating their expression pattern and emphasizing their active roles in mesenchymal condensation, chondrocyte proliferation, and chondrocyte maturation. It also highlights how Runx proteins regulate transcription of target genes and how Runx proteins are regulated in the cartilaginous skeleton.


Subject(s)
Chondrocytes/cytology , Chondrogenesis/genetics , Core Binding Factor alpha Subunits/physiology , Gene Expression Regulation, Developmental , Amino Acid Sequence , Animals , Cell Proliferation , Chondrocytes/metabolism , Chondrocytes/pathology , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor alpha Subunits/genetics , Humans , Hypertrophy/metabolism , Mesoderm , Mice , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...