Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Physiol Biochem Zool ; 96(1): 53-61, 2023.
Article in English | MEDLINE | ID: mdl-36626842

ABSTRACT

AbstractIn most systems, the caspase cascade is activated during cellular stress and results in inflammation and apoptosis. Hibernators experience stressors such as extremely low body temperatures, bradycardia, possible ischemia and reperfusion, and acidosis. However, widespread inflammation and apoptosis would represent an energetic expense that is incompatible with hibernation. To better understand global caspase regulation during hibernation, we employed a systems-level approach and analyzed 11 caspases in ground squirrel liver that are involved in inflammatory (caspases 1, 4, 5, 11, and 12) and apoptotic (caspases 2, 6, 7, 8, 9, and 10) pathways. Western blots revealed liberation of active forms for two inflammatory (caspases 11 and 12) and two apoptotic (caspases 6 and 9) caspases during hibernation (e.g., p15, the most active fragment of caspase 6, increased 8.26±0.70-fold in interbout-aroused animals). We used specific peptide substrates to interrogate the four seemingly activated caspases and demonstrated no expected increases in proteolytic activity. Specific targets of these four caspases were similarly not cleaved, demonstrating that initiation of caspase activation may occur without concomitant downstream effects. Similarly, we found no evidence for upstream activation for caspase 9 signaling based on permeabilization of the outer mitochondrial membrane. We contend that these caspases are suppressed after seeming activation during hibernation. Incomplete caspase signaling is effectively mitigating the induction of widespread inflammation and apoptosis during hibernation.


Subject(s)
Hibernation , Rodent Diseases , Animals , Caspases/metabolism , Sciuridae/physiology , Signal Transduction , Apoptosis , Inflammation , Hibernation/physiology
2.
Elife ; 82019 12 23.
Article in English | MEDLINE | ID: mdl-31868589

ABSTRACT

The cullin-RING ligases (CRLs) form the major family of E3 ubiquitin ligases. The prototypic CRLs in yeast, called SCF enzymes, employ a single E2 enzyme, Cdc34, to build poly-ubiquitin chains required for degradation. In contrast, six different human E2 and E3 enzyme activities, including Cdc34 orthologs UBE2R1 and UBE2R2, appear to mediate SCF-catalyzed substrate polyubiquitylation in vitro. The combinatorial interplay of these enzymes raises questions about genetic buffering of SCFs in human cells and challenges the dogma that E3s alone determine substrate specificity. To enable the quantitative comparisons of SCF-dependent ubiquitylation reactions with physiological enzyme concentrations, mass spectrometry was employed to estimate E2 and E3 levels in cells. In combination with UBE2R1/2, the E2 UBE2D3 and the E3 ARIH1 both promoted SCF-mediated polyubiquitylation in a substrate-specific fashion. Unexpectedly, UBE2R2 alone had negligible ubiquitylation activity at physiological concentrations and the ablation of UBE2R1/2 had no effect on the stability of SCF substrates in cells. A genome-wide CRISPR screen revealed that an additional E2 enzyme, UBE2G1, buffers against the loss of UBE2R1/2. UBE2G1 had robust in vitro chain extension activity with SCF, and UBE2G1 knockdown in cells lacking UBE2R1/2 resulted in stabilization of the SCF substrates p27 and CYCLIN E as well as the CUL2-RING ligase substrate HIF1α. The results demonstrate the human SCF enzyme system is diversified by association with multiple catalytic enzyme partners.


Subject(s)
Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Genome, Human/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mass Spectrometry , Polyubiquitin/genetics , Signal Transduction/genetics , Ubiquitination/genetics
3.
J Exp Med ; 214(4): 1029-1047, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28330905

ABSTRACT

Diabetic retinopathy (DR) is a leading cause of vision loss with retinal vascular leakage and/or neovascularization. Current antiangiogenic therapy against vascular endothelial growth factor (VEGF) has limited efficacy. In this study, we applied a new technology of comparative ligandomics to diabetic and control mice for the differential mapping of disease-related endothelial ligands. Secretogranin III (Scg3) was discovered as a novel disease-associated ligand with selective binding and angiogenic activity in diabetic but not healthy vessels. In contrast, VEGF bound to and induced angiogenesis in both diabetic and normal vasculature. Scg3 and VEGF signal through distinct receptor pathways. Importantly, Scg3-neutralizing antibodies alleviated retinal vascular leakage in diabetic mice with high efficacy. Furthermore, anti-Scg3 prevented retinal neovascularization in oxygen-induced retinopathy mice, a surrogate model for retinopathy of prematurity (ROP). ROP is the most common cause of vision impairment in children, with no approved drug therapy. These results suggest that Scg3 is a promising target for novel antiangiogenic therapy of DR and ROP.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Chromogranins/physiology , Diabetic Retinopathy/drug therapy , Animals , Capillary Permeability , Chromogranins/antagonists & inhibitors , High-Throughput Nucleotide Sequencing , Ligands , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology
4.
PLoS One ; 10(5): e0126993, 2015.
Article in English | MEDLINE | ID: mdl-25992960

ABSTRACT

Phagocytosis is critical to the clearance of apoptotic cells, cellular debris and deleterious metabolic products for tissue homeostasis. Phagocytosis ligands directly recognizing deleterious cargos are the key to defining the functional roles of phagocytes, but are traditionally identified on a case-by-case basis with technical challenges. As a result, extrinsic regulation of phagocytosis is poorly defined. Here we demonstrate that microglial phagocytosis ligands can be systematically identified by a new approach of functional screening. One of the identified ligands is reticulocalbin-1 (Rcn1), which was originally reported as a Ca2+-binding protein with a strict expression in the endoplasmic reticulum. Our results showed that Rcn1 can be secreted from healthy cells and that secreted Rcn1 selectively bound to the surface of apoptotic neurons, but not healthy neurons. Independent characterization revealed that Rcn1 stimulated microglial phagocytosis of apoptotic but not healthy neurons. Ingested apoptotic cells were targeted to phagosomes and co-localized with phagosome marker Rab7. These data suggest that Rcn1 is a genuine phagocytosis ligand. The new approach described in this study will enable systematic identification of microglial phagocytosis ligands with broad applicability to many other phagocytes.


Subject(s)
Calcium-Binding Proteins/metabolism , Microglia/physiology , Phagocytosis , Animals , Apoptosis , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/pharmacology , Extracellular Space/metabolism , Humans , Ligands , Macrophages/drug effects , Microglia/drug effects , Neurons/drug effects , Neurons/metabolism , Phagocytosis/drug effects , Phagosomes , Protein Binding , Protein Transport
5.
PLoS One ; 10(5): e0127904, 2015.
Article in English | MEDLINE | ID: mdl-25996149

ABSTRACT

Hepatoma-derived growth factor-related protein-3 (Hdgfrp3 or HRP-3) was recently reported as a neurotrophic factor and is upregulated in hepatocellular carcinoma to promote cancer cell survival. Here we identified HRP-3 as a new endothelial ligand and characterized its in vitro and in vivo functional roles and molecular signaling. We combined open reading frame phage display with multi-round in vivo binding selection to enrich retinal endothelial ligands, which were systematically identified by next generation DNA sequencing. One of the identified endothelial ligands was HRP-3. HRP-3 expression in the retina and brain was characterized by Western blot and immunohistochemistry. Cell proliferation assay showed that HRP-3 stimulated the growth of human umbilical vein endothelial cells (HUVECs). HRP-3 induced tube formation of HUVECs in culture. Wound healing assay indicated that HRP-3 promoted endothelial cell migration. HRP-3 was further confirmed for its in vitro angiogenic activity by spheroid sprouting assay. HRP-3 extrinsically activated the extracellular-signal-regulated kinase ½ (ERK1/2) pathway in endothelial cells. The angiogenic activity of HRP-3 was independently verified by mouse cornea pocket assay. Furthermore, in vivo Matrigel plug assay corroborated HRP-3 activity to promote new blood vessel formation. These results demonstrated that HRP-3 is a novel angiogenic factor.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Nuclear Proteins/pharmacology , Animals , Brain/metabolism , Cell Cycle Proteins , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Neovascularization, Physiologic/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Retina/metabolism
6.
Mol Biol Cell ; 26(12): 2311-20, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25904329

ABSTRACT

Phagocytosis of shed photoreceptor outer segments (POSs) by retinal pigment epithelial (RPE) cells is critical to retinal homeostasis and shares many conserved signaling pathways with other phagocytes, including extrinsic regulations. Phagocytotic ligands are the key to cargo recognition, engulfment initiation, and activity regulation. In this study, we identified intracellular protein ATP-binding cassette subfamily F member 1 (ABCF1) as a novel RPE phagocytotic ligand by a new approach of functional screening. ABCF1 was independently verified to extrinsically promote phagocytosis of shed POSs by D407 RPE cells. This finding was further corroborated with primary RPE cells and RPE explants. Internalized POS vesicles were colocalized with a phagosome marker, suggesting that ABCF1-mediated engulfment is through a phagocytic pathway. ABCF1 was released from apoptotic cells and selectively bound to shed POS vesicles and apoptotic cells, possibly via externalized phosphatidylserine. ABCF1 is predominantly expressed in POSs and colocalized with the POS marker rhodopsin, providing geographical convenience for regulation of RPE phagocytosis. Collectively these results suggest that ABCF1 is released from and binds to shed POSs in an autocrine manner to facilitate RPE phagocytosis through a conserved pathway. Furthermore, the new approach is broadly applicable to many other phagocytes and will enable systematic elucidation of their ligands to understand extrinsic regulation and cargo recognition.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Phagocytosis , Retinal Photoreceptor Cell Outer Segment/physiology , Retinal Pigment Epithelium/physiology , Animals , Cells, Cultured , Humans , Ligands , Mice , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
7.
J Cell Biochem ; 116(10): 2177-87, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25735755

ABSTRACT

Phagocytosis is critical to tissue homeostasis, as highlighted by phagocytosis defect of retinal pigment epithelial (RPE) cells with debris accumulation, photoreceptor degeneration and blindness. Phagocytosis ligands are the key to delineating molecular mechanisms and functional roles of phagocytes, but are traditionally identified in individual cases with technical challenges. We recently developed open reading frame phage display (OPD) for phagocytosis-based functional cloning (PFC) to identify unknown ligands. One of the identified ligands was Ly-1 antibody reactive clone (Lyar) with functions poorly defined. Herein, we characterized Lyar as a new ligand to stimulate RPE phagocytosis. In contrast to its reported nucleolar expression, immunohistochemistry showed that Lyar was highly expressed in photoreceptor outer segments (POSs) of the retina. Cytoplasmic Lyar was released from apoptotic cells, and selectively bound to shed POSs and apoptotic cells, but not healthy cells. POS vesicles engulfed through Lyar-dependent pathway were targeted to phagosomes and colocalized with phagosome marker Rab7. These results suggest that Lyar is a genuine RPE phagocytosis ligand, which in turn supports the validity of OPD/PFC as the only available approach for unbiased identification of phagocytosis ligands with broad applicability to various phagocytes.


Subject(s)
DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Retina/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Apoptosis/genetics , DNA-Binding Proteins/metabolism , Ligands , Mice , Nuclear Proteins/metabolism , Phagocytes/metabolism , Phagocytosis/genetics , Retina/growth & development , Retinal Pigment Epithelium/metabolism , Retinal Pigments/genetics , Retinal Pigments/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
8.
Adv Exp Med Biol ; 801: 503-9, 2014.
Article in English | MEDLINE | ID: mdl-24664737

ABSTRACT

Mutations in either tubby or tubby-like protein 1 (Tulp1) cause retinal degeneration with undefined mechanisms. We recently identified both proteins with unconventional secretion as novel MerTK-specific phagocytosis ligands for retinal pigment epithelium (RPE) cells. Using our newly-developed open reading frame (ORF) phage display as a technology for protein-protein interactions, we identified Tulp1 as a Tubby-binding protein. The interaction of tubby and Tulp1 was verified by yeast two-hybrid and protein pull-down assays. Tubby and Tulp1 form heterodimer or heterooligomer and their interaction was functionally revealed by their synergistic stimulation of RPE phagocytosis. Tubby and Tulp1 mediated phagocytosis through MerTK-dependent signaling with non-muscle myosin II redistribution leading to colocalization of phagocytosed vesicles with rearranged NMMIIA.


Subject(s)
Eye Proteins/physiology , Phagocytosis/physiology , Proteins/physiology , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/physiology , Adaptor Proteins, Signal Transducing , Cell Line , Dimerization , Eye Proteins/chemistry , Eye Proteins/genetics , Humans , Myosin Type II/metabolism , Protein Interaction Domains and Motifs , Proteins/chemistry , Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Retinal Degeneration/genetics , Retinal Pigment Epithelium/cytology , Two-Hybrid System Techniques , c-Mer Tyrosine Kinase
9.
J Neuroimmunol ; 252(1-2): 40-8, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22884297

ABSTRACT

Immunologically-silent microglial phagocytosis of apoptotic cells and cellular debris is critical for CNS homeostasis and innate immune balance. The beneficial and detrimental effects of microglial phagocytosis on neurons remain controversial. Phagocytosis ligands are the key to selecting extracellular cargos, initiating the engulfment process, defining phagocyte functional roles and regulating phagocyte activities with therapeutic potentials. Here we characterized tubby as a new ligand to regulate microglial phagocytosis through MerTK receptor, which is well known for its immunosuppressive signaling. Tubby at 0.1nM significantly induced microglial phagocytosis of apoptotic cells with a maximal activity at 10nM. Tubby activated MerTK with receptor autophosphorylation in a similar dose range. Excessive soluble MerTK extracellular domain blocked tubby-mediated microglial phagocytosis of plasma membrane vesicles as cellular debris. Immunocytochemistry revealed that the ingested cargos were co-localized with MerTK-dependent non-muscle myosin II, whose rearrangement is necessary for cargo engulfment. Phagosome biomarker Rab7 was colocalized with cargos, suggesting that internalized cargos were targeted to phagocytic pathway. Tubby stimulated phagocytosis by neonatal and aged microglia with similar activities, but not by MerTK(-/-) microglia. These results suggest that tubby is a ligand to facilitate microglial phagocytosis through MerTK for the maintenance of CNS homeostasis.


Subject(s)
Microglia/metabolism , Phagocytosis/physiology , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis/physiology , Flow Cytometry , Humans , Immunohistochemistry , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Proteins/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , c-Mer Tyrosine Kinase
10.
J Cell Physiol ; 227(2): 401-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21792939

ABSTRACT

Phagocytosis of apoptotic cells and cellular debris is a critical process of maintaining tissue and immune homeostasis. Defects in the phagocytosis process cause autoimmunity and degenerative diseases. Phagocytosis ligands or "eat-me" signals control the initiation of the process by linking apoptotic cells to receptors on phagocyte surface and triggering signaling cascades for cargo engulfment. Eat-me signals are traditionally identified on a case-by-case basis with challenges, and the identification of their cognate receptors is equally daunting. Here, we identified galectin-3 (Gal-3) as a new MerTK ligand by an advanced dual functional cloning strategy, in which phagocytosis-based functional cloning is combined with receptor-based affinity cloning to directly identify receptor-specific eat-me signal. Gal-3 interaction with MerTK was independently verified by co-immunoprecipitation. Functional analyses showed that Gal-3 stimulated the phagocytosis of apoptotic cells and cellular debris by macrophages and retinal pigment epithelial cells with MerTK activation and autophosphorylation. The Gal-3-mediated phagocytosis was blocked by excessive soluble MerTK extracellular domain and lactose. These results suggest that Gal-3 is a legitimate MerTK-specific eat-me signal. The strategy of dual functional cloning with applicability to other phagocytic receptors will facilitate unbiased identification of their unknown ligands and improve our capacity for therapeutic modulation of phagocytic activity and innate immune response.


Subject(s)
Galectin 3/metabolism , Phagocytosis/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Line , Cloning, Molecular , Galectin 3/genetics , Gene Expression Regulation/physiology , Humans , Mice , Peptide Library , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , c-Mer Tyrosine Kinase
12.
Proc Natl Acad Sci U S A ; 108(32): E431-9, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21670274

ABSTRACT

The signal transduction networks that initiate multicellular development in bacteria remain largely undefined. Here, we report that Myxococcus xanthus regulates entry into its multicellular developmental program using a novel strategy: a cascade of transcriptional activators known as enhancer binding proteins (EBPs). The EBPs in the cascade function in sequential stages of early development, and several lines of evidence indicate that the cascade is propagated when EBPs that function at one stage of development directly regulate transcription of an EBP gene important for the next developmental stage. We also show that the regulatory cascade is designed in a novel way that extensively expands on the typical use of EBPs: Instead of using only one EBP to regulate a particular gene or group of genes, which is the norm in other bacterial systems, the cascade uses multiple EBPs to regulate EBP genes that are positioned at key transition points in early development. Based on the locations of the putative EBP promoter binding sites, several different mechanisms of EBP coregulation are possible, including the formation of coregulating EBP transcriptional complexes. We propose that M. xanthus uses an EBP coregulation strategy to make expression of EBP genes that modulate stage-stage transitions responsive to multiple signal transduction pathways, which provide information that is important for a coordinated decision to advance the developmental process.


Subject(s)
Bacterial Proteins/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Developmental , Myxococcus xanthus/growth & development , Myxococcus xanthus/genetics , Bacterial Proteins/genetics , Base Sequence , Electrophoretic Mobility Shift Assay , Genes, Bacterial/genetics , Models, Biological , Molecular Sequence Data , Mutation/genetics , Myxococcus xanthus/cytology , Myxococcus xanthus/metabolism , Operon/genetics , Phenotype , Promoter Regions, Genetic , Protein Binding/genetics , RNA Polymerase Sigma 54/genetics , RNA Polymerase Sigma 54/metabolism , Spores, Bacterial/genetics , Transcription, Genetic
13.
Molecules ; 16(2): 1739-48, 2011 Feb 21.
Article in English | MEDLINE | ID: mdl-21339709

ABSTRACT

Substrate identification is the key to defining molecular pathways or cellular processes regulated by proteases. Although phage display with random peptide libraries has been used to analyze substrate specificity of proteases, it is difficult to deduce endogenous substrates from mapped peptide motifs. Phage display with conventional cDNA libraries identifies high percentage of non-open reading frame (non-ORF) clones, which encode short unnatural peptides, owing to uncontrollable reading frames of cellular proteins. We recently developed ORF phage display to identify endogenous proteins with specific binding or functional activity with minimal reading frame problem. Here we used calpain 2 as a protease to demonstrate that ORF phage display is capable of identifying endogenous substrates and showed its advantage to re-verify and characterize the identified substrates without requiring pure substrate proteins. An ORF phage display cDNA library with C-terminal biotin was bound to immobilized streptavidin and released by cleavage with calpain 2. After three rounds of phage selection, eleven substrates were identified, including calpastatin of endogenous calpain inhibitor. These results suggest that ORF phage display is a valuable technology to identify endogenous substrates for proteases.


Subject(s)
Calpain/metabolism , Open Reading Frames , Peptide Library , Peptides/metabolism , Animals , Calcium-Binding Proteins/metabolism , Calpain/antagonists & inhibitors , Calpain/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Peptides/genetics , Rats
14.
Clin Immunol ; 138(1): 60-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20943442

ABSTRACT

Acute anterior uveitis (AAU) is the most common form of autoimmune uveitis in the eye with few known autoantigens. Identification of autoantigens will improve our understanding of the molecular mechanisms and capability for disease diagnosis. Phage display is a powerful technology for autoantigen identification. However, because of uncontrollable reading frames, phage display with conventional cDNA libraries identifies high percentage of non-open reading frames (non-ORFs) with minimal implications for autoantigen identification. We recently developed ORF phage display technology with minimal reading frame problem. Herein we used ORF phage display to identify 18 patient-specific clones, including 16 ORFs encoding endogenous proteins as candidate autoantigens for AAU. One of the identified antigens was heterogeneous nuclear ribonucleoprotein H3 (Hnrph3) that was further characterized for AAU relevance and independently verified by Western blot. These results demonstrate that ORF phage display is a valuable approach for identification of unknown autoantigens.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/immunology , Uveitis, Anterior/immunology , Antigen-Antibody Reactions/immunology , Autoantigens/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Humans , Immunoglobulin G/immunology , Open Reading Frames/genetics , Open Reading Frames/immunology , Peptide Library , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Analysis, DNA
15.
EMBO J ; 29(23): 3898-910, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20978472

ABSTRACT

Tubby and tubby-like protein 1 (Tulp1) are newly identified phagocytosis ligands to facilitate retinal pigment epithelium (RPE) and macrophage phagocytosis. Both proteins without classical signal peptide have been demonstrated with unconventional secretion. Here, we characterized them as novel MerTK ligands to facilitate phagocytosis. Tulp1 interacts with Tyro3, Axl and MerTK of the TAM receptor tyrosine kinase subfamily, whereas tubby binds only to MerTK. Excessive soluble MerTK extracellular domain blocked tubby- or Tulp1-mediated phagocytosis. Both ligands induced MerTK activation with receptor phosphorylation and signalling cascade, including non-muscle myosin II redistribution and co-localization with phagosomes. Tubby and Tulp1 are bridging molecules with their N-terminal region as MerTK-binding domain and C-terminal region as phagocytosis prey-binding domain (PPBD). Five minimal phagocytic determinants (MPDs) of K/R(X)(1-2)KKK in Tulp1 N-terminus were defined as essential motifs for MerTK binding, receptor phosphorylation and phagocytosis. PPBD was mapped to the highly conserved 54 amino acids at the C-terminal end of tubby and Tulp1. These data suggest that tubby and Tulp1 are novel bridging molecules to facilitate phagocytosis through MerTK.


Subject(s)
Eye Proteins/metabolism , Phagocytosis , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Eye Proteins/genetics , Humans , Macrophages/cytology , Mice , Mutation , Myosin Type II/metabolism , Protein Binding , Protein Structure, Tertiary , Proteins/genetics , Swine , c-Mer Tyrosine Kinase
16.
J Mol Recognit ; 23(1): 74-83, 2010.
Article in English | MEDLINE | ID: mdl-19718693

ABSTRACT

Mutation in the tubby gene causes adult-onset obesity, progressive retinal, and cochlear degeneration with unknown mechanism. In contrast, mutations in tubby-like protein 1 (Tulp1), whose C-terminus is highly homologous to tubby, only lead to retinal degeneration. We speculate that their diverse N-terminus may define their distinct disease profile. To elucidate the binding partners of tubby, we used tubby N-terminus (tubby-N) as bait to identify unknown binding proteins with open-reading-frame (ORF) phage display. T7 phage display was engineered with three improvements: high-quality ORF phage display cDNA library, specific phage elution by protease cleavage, and dual phage display for sensitive high throughput screening. The new system is capable of identifying unknown bait-binding proteins in as fast as approximately 4-7 days. While phage display with conventional cDNA libraries identifies high percentage of out-of-frame unnatural short peptides, all 28 tubby-N-binding clones identified by ORF phage display were ORFs. They encode 16 proteins, including 8 nuclear proteins. Fourteen proteins were analyzed by yeast two-hybrid assay and protein pull-down assay with ten of them independently verified. Comparative binding analyses revealed several proteins binding to both tubby and Tulp1 as well as one tubby-specific binding protein. These data suggest that tubby-N is capable of interacting with multiple nuclear and cytoplasmic protein binding partners. These results demonstrated that the newly-engineered ORF phage display is a powerful technology to identify unknown protein-protein interactions.


Subject(s)
Bacteriophage T7/metabolism , Peptide Library , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Base Sequence , Eye Proteins/chemistry , Eye Proteins/metabolism , Gene Library , Genetic Vectors , Humans , Molecular Sequence Data , Open Reading Frames , Protein Structure, Tertiary , Proteins/chemistry , Two-Hybrid System Techniques
17.
Appl Microbiol Biotechnol ; 85(4): 909-19, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19885657

ABSTRACT

Phage display with antibody libraries has been widely used with versatile applications. However, phage display with cDNA libraries is rare and inefficient. Because of uncontrollable reading frames and stop codons in cDNA repertoires, high percentage of phage clones identified from conventional cDNA libraries are non-open reading frames (non-ORFs) encoding unnatural short peptides with minimal implications in protein networks. Consequently, phage display has not been used as a technology of functional proteomics to elucidate protein-protein interactions like yeast two-hybrid system and mass spectrometry-based technologies. Several strategies, including C-terminal display and ORF cDNA libraries, have been explored to circumvent the technical problem. The accumulative endeavors eventually led to the efficient elucidation of a large number of tubby- and phosphatidylserine-binding proteins in recent studies by ORF phage display with minimal reading frame issue. ORF phage display inherits all the versatile applications of antibody phage display, but enables efficient identification of real endogenous proteins with efficiency, sensitivity, and accuracy comparable to other technologies of functional proteomics. Its ELISA-like procedure can be conveniently adapted by individual laboratories or fully automated for high-throughput screening. Thus, ORF phage display is an efficient, sensitive, versatile, and convenient technology of functional proteomics for elucidation of global and pathway-specific protein-protein interactions, disease mechanisms, or therapeutic targets.


Subject(s)
Gene Library , Peptide Library , Proteomics , Capsid Proteins/genetics , DNA, Complementary , Molecular Sequence Data , Open Reading Frames
18.
Exp Cell Res ; 316(2): 245-57, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19837063

ABSTRACT

Phagocytosis is an important process for the removal of apoptotic cells or cellular debris. Eat-me signals control the initiation of phagocytosis and hold the key for in-depth understanding of its molecular mechanisms. However, because of difficulties to identify unknown eat-me signals, only a limited number of them have been identified and characterized. Using a newly developed functional cloning strategy of open reading frame (ORF) phage display, we identified nine putative eat-me signals, including tubby-like protein 1 (Tulp1). This further led to the elucidation of tubby as the second eat-me signal in the same protein family. Both proteins stimulated phagocytosis of retinal pigment epithelium (RPE) cells and macrophages. Tubby-conjugated fluorescent microbeads facilitated RPE phagocytosis. Tubby and Tulp1, but not other family members, enhanced the uptake of membrane vesicles by RPE cells in synergy. Retinal membrane vesicles of Tubby mice and Tulp1(-/-) mice showed reduced activities for RPE phagocytosis, which were compensated by purified tubby and Tulp1, respectively. These data reveal a novel activity of tubby and Tulp1, and demonstrate that unbiased identification of eat-me signals by the broadly applicable strategy of ORF phage display can provide detailed insights into phagocyte biology.


Subject(s)
Eye Proteins/physiology , Peptide Library , Phagocytosis/physiology , Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cell Line, Tumor , Cell Membrane Structures/metabolism , Cloning, Molecular , HeLa Cells , Humans , Jurkat Cells , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microspheres , Mutation, Missense/physiology , Phagocytes/physiology , Protein Binding/physiology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Sequence Deletion/physiology , Swine , Transfection
19.
FEBS Lett ; 583(18): 3057-62, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19695251

ABSTRACT

Tubby-like proteins (Tulps) with no signal peptide have been characterized as cytoplasmic proteins with various intracellular functions, including binding to phosphatidylinositol-4,5-bisphosphate [PI(4,5)P(2)]. PI(4,5)P(2) has been implicated in unconventional secretion of fibroblast growth factor-2 without a signal peptide. Here, we show that all Tulps are expressed intracellularly and extracellularly. Tubby secretion is partially dependent on its PI(4,5)P(2)-binding activity with an essential secretory signal in the N-terminus. Pathogenic mutation in Tubby mice has no impact on tubby extracellular trafficking. Moreover, unconventional secretion of tubby and Tulp1 is independent of endoplasmic reticulum-Golgi pathway. These data implicate that Tulps may function extracellularly as well.


Subject(s)
Eye Proteins/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Compartmentation , Fibroblast Growth Factor 2/metabolism , Mice , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Transport
20.
Biochem Biophys Res Commun ; 386(1): 197-201, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19520055

ABSTRACT

To efficiently elucidate the biological roles of phosphatidylserine (PS), we developed open-reading-frame (ORF) phage display to identify PS-binding proteins. The procedure of phage panning was optimized with a phage clone expressing MFG-E8, a well-known PS-binding protein. Three rounds of phage panning with ORF phage display cDNA library resulted in approximately 300-fold enrichment in PS-binding activity. A total of 17 PS-binding phage clones were identified. Unlike phage display with conventional cDNA libraries, all 17 PS-binding clones were ORFs encoding 13 real proteins. Sequence analysis revealed that all identified PS-specific phage clones had dimeric basic amino acid residues. GST fusion proteins were expressed for 3 PS-binding proteins and verified for their binding activity to PS liposomes, but not phosphatidylcholine liposomes. These results elucidated previously unknown PS-binding proteins and demonstrated that ORF phage display is a versatile technology capable of efficiently identifying binding proteins for non-protein molecules like PS.


Subject(s)
Peptide Library , Phosphatidylserines/metabolism , Proteins/isolation & purification , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, Surface/chemistry , Antigens, Surface/genetics , Antigens, Surface/isolation & purification , Antigens, Surface/metabolism , Gene Library , Mice , Milk Proteins/chemistry , Milk Proteins/genetics , Milk Proteins/isolation & purification , Milk Proteins/metabolism , Molecular Sequence Data , Open Reading Frames , Phosphatidylserines/chemistry , Protein Binding , Proteins/chemistry , Proteins/genetics , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...