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










Publication year range
1.
Adv Sci (Weinh) ; 6(11): 1802115, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31179210

ABSTRACT

Colorectal cancer (CRC) is the second-leading cause of cancer-related mortality worldwide, which may be effectively reduced by early screening. Colon cancer secreted protein-2 (CCSP-2) is a promising blood marker for CRC. An electric-field effect colorectal sensor (E-FECS), an ion-sensitive field-effect transistor under dual gate operation with nanostructure is developed, to quantify CCSP-2 directly from patient blood samples. The sensing performance of the E-FECS is verified in 7 controls and 7 CRC samples, and it is clinically validated on 30 controls, 30 advanced adenomas, and 81 CRC cases. The concentration of CCSP-2 is significantly higher in plasma samples from CRC and advanced adenoma compared with controls (both P < 0.001). Sensitivity and specificity for CRC versus controls are 44.4% and 86.7%, respectively (AUC of 0.67), and 43.3% and 86.7%, respectively, for advanced adenomas (AUC of 0.67). CCSP-2 detects a greater number of CRC cases than carcinoembryonic antigen does (45.6% vs 24.1%), and the combination of the two markers detects an even greater number of cases (53.2%). The E-FECS system successfully detects CCSP-2 in a wide range of samples including early stage cancers and advanced adenoma. CCSP-2 has potential for use as a blood-based biomarker for CRC.

2.
Biomaterials ; 195: 38-50, 2019 03.
Article in English | MEDLINE | ID: mdl-30610992

ABSTRACT

Alveolar macrophages resident in the lung are prominent phagocytic effector cells of the pulmonary innate immune response, and paradoxically, are attractive harbors for pathogens. Consequently, facultative intracellular bacteria, such as Francisella tularensis, can cause severe systemic disease and sepsis, with high morbidity and mortality associated with pulmonary infection. Current clinical treatment, which involves exhaustive oral or intravenous antibiotic therapy, has limitations such as systemic toxicity and off-target effects. Pulmonary administration represents a promising alternative to systemic dosing for delivering antibiotics directly to the lung. Here, we present synthesized mannosylated ciprofloxacin polymeric prodrugs for efficient pulmonary delivery, targeting, and subsequent internalization by alveolar macrophages. We demonstrate significant improvement in efficacy against intracellular infections in an otherwise uniformly lethal airborne Francisella murine model (F. novicida). When administered to the lungs of mice in a prophylactic regimen, the mannosylated ciprofloxacin polymeric prodrugs led to 50% survival. In a treatment regimen that was concurrent with infection, the survival of mice increased to 87.5%. Free ciprofloxacin antibiotic was ineffective in both cases. This significant difference in antibacterial efficacy demonstrates the impact of this delivery platform based on improved physiochemical, pharmacokinetic, and pharmacodynamic properties of ciprofloxacin administered via our glycan polymeric prodrug. This modular platform provides a route for overcoming the limitations of free drug and increasing efficacy in treatment of intracellular infection.


Subject(s)
Macrophages, Alveolar/metabolism , Polysaccharides/chemistry , Prodrugs/chemistry , Francisella tularensis/metabolism , Magnetic Resonance Spectroscopy , Mannose/metabolism , Microbial Sensitivity Tests
3.
Sci Rep ; 8(1): 11314, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054522

ABSTRACT

Owing to the recent progress in regenerative medicine technology, clinical trials that harnessed the regeneration and immune modulation potentiality of stem cells for treating IBD have shown promising results. We investigated the feasibility and utility of intraluminal endoscopic transplantation of rat MSC sheets in murine models of experimental colitis for targeted delivery of stem cells to lesions. We isolated adipose-derived mesenchymal stem cells (AD-MSC) and bone marrow-derived mesenchymal stem cells (BM-MSC) from EGFP-transgenic rats and fabricated the cells in sheet forms using temperature-responsive culture dishes. The MSC sheets were endoscopically transplanted to the inflamed area in electrocoagulation and DNBS colitis model. The effect of the transplantation was verified using endoscopic scoring and histological analysis. In the electrocoagulation model, the AD-MSC group showed significantly decreased ulcer size in the transplanted regions. In the DNBS colitis model, the AD-MSC group showed decreased inflammation and colitis in the transplanted regions. Histologic analysis showed that the MSC sheets had successfully attached to the inflamed mucosa in both the electrocoagulation and DNBS colitis model. Our results show that endoscopic transplantation of MSC sheets could be a new effective mode of stem cell therapy for IBD treatment.


Subject(s)
Colitis/therapy , Inflammation/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dinitrofluorobenzene/analogs & derivatives , Dinitrofluorobenzene/toxicity , Disease Models, Animal , Endoscopes , Green Fluorescent Proteins/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Mice , Rats , Rats, Transgenic/genetics
4.
Biomater Sci ; 6(7): 1976-1985, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29850694

ABSTRACT

Pulmonary intracellular infections, such as tuberculosis, anthrax, and tularemia, have remained a significant challenge to conventional antibiotic therapy. Ineffective antibiotic treatment of these infections can lead not only to undesired side effects, but also to the emergence of antibiotic resistance. Aminoglycosides (e.g., streptomycin) have long been part of the therapeutic regiment for many pulmonary intracellular infections. Their bioavailability for intracellular bacterial pools, however, is limited by poor membrane permeability and rapid elimination. To address this challenge, polymer-augmented liposomes (PALs) were developed to provide improved cytosolic delivery of streptomycin to alveolar macrophages, an important host cell for intracellular pathogens. A multifunctional diblock copolymer was engineered to functionalize PALs with carbohydrate-mediated targeting, pH-responsive drug release, and endosomal release activity with a single functional polymer that replaces the pegylated lipid component to simplify the liposome formulation. The pH-sensing functionality enabled PALs to provide enhanced release of streptomycin under endosomal pH conditions (70% release in 6 hours) with limited release at physiological pH 7.4 (16%). The membrane-destabilizing activity connected to endosomal release was characterized in a hemolysis assay and PALs displayed a sharp pH profile across the endosomal pH development target range. The direct connection of this membrane-destabilizing pH profile to model drug release was demonstrated in an established pyranine/p-xylene bispyridinium dibromide (DPX) fluorescence dequenching assay. PALs displayed similar sharp pH-responsive release, whereas PEGylated control liposomes did not, and similar profiles were then shown for streptomycin release. The mannose-targeting capability of the PALs was also demonstrated with 2.5 times higher internalization compared to non-targeted PEGylated liposomes. Finally, the streptomycin-loaded PALs were shown to have a significantly improved intracellular antibacterial activity in a Francisella-macrophage co-culture model, compared with free streptomycin or streptomycin delivered by control PEGylated liposomes (13× and 16×, respectively). This study suggests the potential of PALs as a useful platform to deliver antibiotics for the treatment of intracellular macrophage infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Delivery Systems/methods , Francisella tularensis/drug effects , Liposomes/pharmacology , Streptomycin/pharmacology , Animals , Anti-Bacterial Agents/metabolism , Arylsulfonates/chemistry , Drug Compounding/methods , Drug Liberation , Endosomes/drug effects , Endosomes/metabolism , Endosomes/microbiology , Fluorescent Dyes/chemistry , Francisella tularensis/growth & development , Francisella tularensis/metabolism , Hydrogen-Ion Concentration , Kinetics , Liposomes/chemical synthesis , Liposomes/metabolism , Mannose/metabolism , Methacrylates/chemistry , Mice , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Pyridinium Compounds/chemistry , RAW 264.7 Cells , Streptomycin/metabolism
5.
Mol Pharm ; 14(6): 1988-1997, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28394614

ABSTRACT

Lung-based intracellular bacterial infections remain one of the most challenging infectious disease settings. For example, the current standard for treating Franciscella tularensis pneumonia (tularemia) relies on administration of oral or intravenous antibiotics that poorly achieve and sustain pulmonary drug bioavailability. Inhalable antibiotic formulations are approved and in clinical development for upper respiratory infections, but sustained drug dosing from inhaled antibiotics against alveolar intracellular infections remains a current unmet need. To provide an extended therapy against alveolar intracellular infections, we have developed a macromolecular therapeutic platform that provides sustained local delivery of ciprofloxacin with controlled dosing profiles. Synthesized using RAFT polymerization, these macromolecular prodrugs characteristically have high drug loading (16-17 wt % drug), tunable hydrolysis kinetics mediated by drug linkage chemistry (slow-releasing alkyllic vs fast-releasing phenolic esters), and, in general, represent new fully synthetic nanotherapeutics with streamlined manufacturing profiles. In aerosolized and completely lethal F.t. novicida mouse challenge models, the fast-releasing ciprofloxacin macromolecular prodrug provided high cure efficiencies (75% survival rate under therapeutic treatment), and the importance of release kinetics was demonstrated by the inactivity of the similar but slow-releasing prodrug system. Pharmacokinetics and biodistribution studies further demonstrated that the efficacious fast-releasing prodrug retained drug dosing in the lung above the MIC over a 48 h period with corresponding Cmax/MIC and AUC0-24h/MIC ratios being greater than 10 and 125, respectively; the thresholds for optimal bactericidal efficacy. These findings identify the macromolecular prodrug platform as a potential therapeutic system to better treat alveolar intracellular infections such as F. tularensis, where positive patient outcomes require tailored antibiotic pharmacokinetic and treatment profiles.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/therapeutic use , Administration, Intranasal , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Ciprofloxacin/administration & dosage , Ciprofloxacin/pharmacokinetics , Disease Models, Animal , Female , Francisella tularensis/drug effects , Francisella tularensis/pathogenicity , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Tissue Distribution
6.
Sci Rep ; 6: 36818, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830760

ABSTRACT

The PDZ domain-containing scaffold protein, syntenin-1, binds to the transmembrane proteoglycan, syndecan-4, but the molecular mechanism/function of this interaction are unknown. Crystal structure analysis of syntenin-1/syndecan-4 cytoplasmic domains revealed that syntenin-1 forms a symmetrical pair of dimers anchored by a syndecan-4 dimer. The syndecan-4 cytoplasmic domain is a compact intertwined dimer with a symmetrical clamp shape and two antiparallel strands forming a cavity within the dimeric twist. The PDZ2 domain of syntenin-1 forms a direct antiparallel interaction with the syndecan-4 cytoplasmic domain, inhibiting the functions of syndecan-4 such as focal adhesion formation. Moreover, C-terminal region of syntenin-1 reveals an essential role for enhancing the molecular homodimerization. Mutation of key syntenin-1 residues involved in the syndecan-4 interaction or homodimer formation abolishes the inhibitory function of syntenin-1, as does deletion of the homodimerization-related syntenin-1 C-terminal domain. Syntenin-1, but not dimer-formation-incompetent mutants, rescued the syndecan-4-mediated inhibition of migration and pulmonary metastasis by B16F10 cells. Therefore, we conclude that syntenin-1 negatively regulates syndecan-4 function via oligomerization and/or syndecan-4 interaction, impacting cytoskeletal organization and cell migration.


Subject(s)
Syndecan-4/chemistry , Syntenins/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement , Crystallography, X-Ray , Humans , Lymphatic Metastasis , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Rats , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Syndecan-4/physiology , Syntenins/physiology
7.
Nanomedicine ; 12(7): 2031-2041, 2016 10.
Article in English | MEDLINE | ID: mdl-27184097

ABSTRACT

Carbohydrate receptors on alveolar macrophages are attractive targets for receptor-mediated delivery of nanostructured therapeutics. In this study, we employed reversible addition fragmentation chain transfer polymerization to synthesize neoglycopolymers, consisting of mannose- and galactose methacrylate-based monomers copolymerized with cholesterol methacrylate for use in functional liposome studies. Glycopolymer-functional liposomes were employed to elucidate macrophage mannose receptor (CD206) and macrophage galactose-type lectin (CD301) targeting in both primary macrophage and immortal macrophage cell lines. Expression of CD206 and CD301 was observed to vary significantly between cell lines (murine alveolar macrophage, murine bone marrow-derived macrophage, RAW264.7, and MH-S), which has significant implications in in vitro targeting and uptake studies. Synthetic glycopolymers and glycopolymer augmented liposomes demonstrated specific receptor-mediated uptake in a manner dependent on carbohydrate receptor expression. These results establish a platform capable of probing endogenous carbohydrate receptor-mediated targeting via glycofunctional nanomaterials.


Subject(s)
Carbohydrate Metabolism , Liposomes , Macrophages, Alveolar , Animals , Cell Line , Drug Carriers , Humans , Lectins , Macrophages , Mannose , Mice
8.
J Med Food ; 18(10): 1121-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26336081

ABSTRACT

Decursin, a bioactive phytochemical isolated from Angelica gigas Nakai (danggwi), has shown preclinical anticancer efficacy in various cancer models. However, the antitumor effect of decursin in melanoma models remains undefined. The antitumor activities of decursin were investigated in B16F10 cells in vitro and in vivo. In this study, we show that treatment with decursin inhibited cell proliferation in a dose-dependent manner in B16F10 cells, but not in normal cells. Decursin also induced apoptosis in B16F10 cells, as determined by annexin V-staining assay and transferase-mediated nick-end labeling (TUNEL) staining assay. Decursin increased the phosphorylation of p38 as well as the expression of Bax while decreasing the phosphorylation of extracellular signaling-regulated kinase (ERK) and the expression of Bcl-2 in B16F10 cells. Moreover, decursin activated caspase-3 in B16F10 cells and xenograft tumor tissue. Together, these findings support further investigations into the potential use of decursin in the treatment of melanoma cells.


Subject(s)
Angelica/chemistry , Antineoplastic Agents, Phytogenic , Apoptosis/drug effects , Benzopyrans/pharmacology , Butyrates/pharmacology , Melanoma, Experimental/pathology , 3T3 Cells , Animals , Benzopyrans/therapeutic use , Butyrates/therapeutic use , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , In Situ Nick-End Labeling , Male , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/analysis , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/analysis , p38 Mitogen-Activated Protein Kinases/metabolism
9.
PLoS One ; 10(3): e0121171, 2015.
Article in English | MEDLINE | ID: mdl-25803297

ABSTRACT

A number of peptide-based indicators have been identified and reported as potential apoptosis probes, offering great promise for early assessment of therapeutic efficacy in several types of cancer. Direct comparison of the newly developed probes with previously used ones would be an important step in assessing possible applications. Here, we compared the newly identified peptide-based phosphatidylserine (PS) indicator PSP1 (CLSYYPSYC) with annexin V, a common probe for molecular imaging of apoptotic cells, with respect to PS binding kinetics, apoptotic cell-targeting ability, and the efficacy of homing to apoptotic tumor cells in a mouse model after treatment with the anticancer agent camptothecin. Our results indicate that PSP1 efficiently targeted apoptotic cells and generated apoptosis/tumor-specific signals after cancer treatment in the animal model, whereas a similar dose of annexin V showed weak signals. The formation of a stable complex of PSP1 with PS might be one reason for the efficient in vivo targeting. We suggest that PSP1 has potential advantages for in vivo apoptotic cell imaging and could serve as a platform for the development of de novo peptide-based probes for apoptosis.


Subject(s)
Annexin A5/chemistry , Apoptosis/physiology , Lung Neoplasms/pathology , Molecular Imaging/methods , Oligopeptides/chemistry , Phosphatidylserines/analysis , Animals , Annexin A5/metabolism , Cell Line, Tumor , Female , Humans , Indicators and Reagents/chemistry , Lung Neoplasms/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/metabolism , Phosphatidylserines/metabolism
10.
PLoS One ; 10(2): e0118310, 2015.
Article in English | MEDLINE | ID: mdl-25705892

ABSTRACT

We developed and tested a multicomponent peptide-woven siRNA nanocomplex (PwSN) comprising different peptides designed for efficient cellular targeting, endosomal escape, and release of siRNA. To enhance tumor-specific cellular uptake, we connected an interleukin-4 receptor-targeting peptide (I4R) to a nine-arginine peptide (9r), yielding I4R-9r. To facilitate endosomal escape, we blended endosomolytic peptides into the I4R-9r to form a multicomponent nanocomplex. Lastly, we modified 9r peptides by varying the number and positions of positive charges to obtain efficient release of siRNA from the nanocomplex in the cytosol. Using this step-wise approach for overcoming the biological challenges of siRNA delivery, we obtained an optimized PwSN with significant biological activity in vitro and in vivo. Interestingly, surface plasmon resonance analyses and three-dimensional peptide models demonstrated that our designed peptide adopted a unique structure that was correlated with faster complex disassembly and a better gene-silencing effect. These studies further elucidate the siRNA nanocomplex delivery pathway and demonstrate the applicability of our stepwise strategy to the design of siRNA carriers capable of overcoming multiple challenges and achieving efficient delivery.


Subject(s)
Drug Delivery Systems/methods , Nanoparticles/chemistry , Peptides/chemistry , RNA, Small Interfering/administration & dosage , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Nanoparticles/administration & dosage , Peptides/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/genetics , Reproducibility of Results , Surface Plasmon Resonance , Transplantation, Heterologous
11.
Biochim Biophys Acta ; 1833(10): 2378-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23792174

ABSTRACT

TGFBI, a transforming growth factor ß-induced extracellular matrix protein, circulates at a level of ~300ng/ml in humans and modulates several integrin-mediated cellular functions. The protein contains an N-terminal EMI domain, four consecutive FAS1 domains, and the RGD motif. Each FAS1 domain and the RGD motif have been known to interact with avb3 integrin. Here, we found that the binding affinity (Kd) of TGFBI for αvß3 integrin was approximately 3.8×10(-8)M, a value ~2300-fold higher than that of a single FAS1 domain, and demonstrated that this greater affinity was due to the cooperative action of the four FAS1 domains and the RGD motif. Moreover, TGFBI exhibited more potent anti-angiogenic and anti-tumorigenic activities, even at a 100-fold lower molar dose than the reported effective dose of the FAS1 domain. Finally, our data showed that TGFBI specifically targeted the tumor vasculature and accumulated at the tumor site. Collectively, our results support the theory that TGFBI acts as a potent endogenous anti-tumor and anti-angiogenic molecule by targeting αvß3 integrin, and highlights the importance of physiological circulating TGFBI levels in inhibiting tumor growth.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Cell Proliferation , Extracellular Matrix Proteins/metabolism , Integrin alphaVbeta3/metabolism , Melanoma, Experimental/prevention & control , Neovascularization, Pathologic/prevention & control , Oligopeptides/chemistry , Transforming Growth Factor beta/metabolism , fas Receptor/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibody Formation , Blotting, Western , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Extracellular Matrix Proteins/immunology , Humans , Immunoenzyme Techniques , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/immunology , Tumor Cells, Cultured , fas Receptor/metabolism
12.
Mol Cancer Res ; 10(8): 1010-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22710795

ABSTRACT

It is known that VEGF receptors (VEGFR) and integrins interact with each other to regulate angiogenesis. We reported previously that the fasciclin 1 (FAS1) domain-containing protein, TGFBIp/ßig-h3 (TGF-ß-induced protein) is an angiogenesis regulator that inhibits both endothelial cell migration and growth via αvß3 integrin. In an attempt to target the interaction between VEGFR-2 and αvß3 integrin, we determined whether the FAS1 domain region of TGFBIp/ßig-h3 (FAS1 domain protein) can block the interaction between the two receptors, leading to the suppression of angiogenesis. In this study, we showed that FAS1 domain protein inhibits VEGF165-induced endothelial cell proliferation and migration via αvß3 integrin, resulting in the inhibition of VEGF165-induced angiogenesis. We also defined a molecular mechanism by which FAS1 domain protein blocks the association between αvß3 integrin and VEGFR-2, showing that it binds to αvß3 integrin but not to VEGFR-2. Blocking the association of these major angiogenic receptors with FAS1 domain protein inhibits signaling pathways downstream of VEGFR-2. Collectively, our results indicate that FAS1 domain protein, in addition to its inhibitory effect on αvß3 integrin-mediated angiogenesis, also inhibits VEGF165-induced angiogenesis. Thus, FAS1 domain protein can be further developed into a potent anticancer drug that targets two principal angiogenic pathways.


Subject(s)
Extracellular Matrix Proteins , Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic , Transforming Growth Factor beta , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Movement , Cell Proliferation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , fas Receptor/chemistry , fas Receptor/metabolism
13.
Blood ; 114(25): 5206-15, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19738031

ABSTRACT

Transforming growth factor-beta-induced protein (TGFBIp)/beta ig-h3 is a 68-kDa extracellular matrix protein that is functionally associated with the adhesion, migration, proliferation, and differentiation of various cells. The presence of TGFBIp in platelets led us to study the role of this protein in the regulation of platelet functions. Upon activation, platelet TGFBIp was released and associated with the platelets. TGFBIp mediates not only the adhesion and spread of platelets but also activates them, resulting in phosphatidylserine exposure, alpha-granule secretion, and increased integrin affinity. The fasciclin 1 domains of TGFBIp are mainly responsible for the activation of platelets. TGFBIp promotes thrombus formation on type I fibrillar collagen under flow conditions in vitro and induces pulmonary embolism in mice. Moreover, transgenic mice, which have approximately a 1.7-fold greater blood TGFBIp concentration, are significantly more susceptible to collagen- and epinephrine-induced pulmonary embolism than wild-type mice. These results suggest that TGFBIp, a human platelet protein, plays important roles in platelet activation and thrombus formation. Our findings will increase our understanding of the novel mechanism of platelet activation, contributing to a better understanding of thrombotic pathways and the development of new antithrombotic therapies.


Subject(s)
Blood Platelets/metabolism , Extracellular Matrix Proteins/physiology , Platelet Activation/physiology , Thrombosis/physiopathology , Transforming Growth Factor beta/physiology , Animals , Blood Cell Count , Blood Platelets/cytology , Blotting, Western , Cell Adhesion , Cell Movement , Collagen , Epinephrine , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Embolism/blood , Pulmonary Embolism/chemically induced , Pulmonary Embolism/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thrombosis/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...