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










Publication year range
1.
Theranostics ; 13(12): 4059-4078, 2023.
Article in English | MEDLINE | ID: mdl-37554277

ABSTRACT

Rationale: CD93, a C-type lectin-like transmembrane glycoprotein, can be shed in a soluble form (sCD93) upon inflammatory stimuli. sCD93 effectively enhances apoptotic cell clearance and has been proposed as an inflammatory disease biomarker. The function of sCD93 involved directly in inflammation remains to be determined. Herein, we attempted to examine the hypothesis that sCD93 might sequester proinflammatory high-mobility group box 1 protein (HMGB1), exerting anti-inflammatory properties. Methods: Different forms of soluble recombinant human CD93 (rCD93) were prepared by a mammalian protein expression system. rCD93-HMGB1 interaction was assessed using co-immunoprecipitation and solid-phase binding assays. Effects of soluble rCD93 were evaluated in HMGB1-induced macrophage and vascular smooth muscle cells (VSMC) activation and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis, CaCl2-induced and angiotensin II-infused abdominal aortic aneurysm (AAA) formation and ovariectomized-induced osteoporosis in mice. Results: Protein binding studies revealed that soluble rCD93, via the lectin-like domain (D1), can bind to HMGB1 and intercept HMGB1-receptor interaction. Soluble rCD93 containing D1 inhibited HMGB1-induced proinflammatory cytokine production and intracellular mitogen-activated protein kinase (MAPK)/nuclear factor (NF)-κB activation in macrophages and VSMCs, thereby attenuating CaCl2-induced and angiotensin II-infused AAA models. During osteoclastogenesis, RANKL stimulated HMGB1 secretion that promoted RANKL-induced osteoclastogenesis in return. Soluble rCD93 containing D1 impeded RANKL-induced osteoclastogenic marker gene expression and intracellular MAPK/NF-κB signaling, thereby mitigating ovariectomized-induced osteoporosis. Conclusion: These findings demonstrate the therapeutic potential of soluble recombinant CD93 containing D1 in inflammatory diseases. Our study highlights a novel anti-inflammatory mechanism, i.e., sequestration of HMGB1, through which sCD93 prevents HMGB1-receptor interaction on effector cells and alleviates inflammation.


Subject(s)
HMGB1 Protein , Humans , Animals , Mice , HMGB1 Protein/metabolism , Lectins , Angiotensin II , Calcium Chloride , Inflammation , Mammals/metabolism
2.
Data Brief ; 30: 105550, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32382597

ABSTRACT

No pharmacotherapy in the clinical setting has been available to alter the natural history of abdominal aortic aneurysm (AAA). Targeting vascular smooth muscle cell (VSMC) dysfunction during the pathogenesis of AAA, including phenotypic switch and apoptosis, could be a potential strategy to limit AAA growth. Here, we provide additional information regarding materials, methods and data related to our recent study published in Atherosclerosis [1]. The therapeutic potential of a self-developed xanthine derivative KMUP-3 was evaluated in VSMC calcification and abdominal aortic aneurysm (AAA). In vitro VSMC calcification was induced using ß-glycerophosphate, and AAA was induced using angiotensin II infusion for 4 weeks in apolipoprotein E-deficient mice. The data contained in this article support the effects of KMUP-3 on VSMC calcification and AAA.

3.
Atherosclerosis ; 297: 16-24, 2020 03.
Article in English | MEDLINE | ID: mdl-32059119

ABSTRACT

BACKGROUND AND AIMS: Inflammation, oxidative stress, matrix degradation, medial calcification and vascular smooth muscle cell (VSMC) loss are prominent features in abdominal aortic aneurysm (AAA). VSMC phenotypic switch to a proinflammatory state and VSMC apoptosis could be targetable mechanisms implicated in the pathogenesis of AAA formation. Herein, we investigated the hypothesis that a xanthine derivative (KMUP-3) might suppress AAA through inhibition of VSMC phenotypic switch and apoptosis. METHODS: In vitro, VSMC calcification was induced using ß-glycerophosphate. In vivo, AAA was induced using angiotensin II (1000 ng/kg per minute) infusion for 4 weeks in apolipoprotein E-deficient mice. RESULTS: As determined by alizarin red S staining and calcium content measurements, KMUP-3 suppressed VSMC calcification. During VSMC calcification, KMUP-3 inhibited mTOR and ß-catenin upregulation, essential for VSMC phenotypic switch, while it enhanced AMP-activated protein kinase (AMPK) activation that protects against VSMC phenotypic switch. Moreover, KMUP-3 attenuated VSMC apoptosis with an increased Bcl-2/Bax ratio and reduced activated caspase-3 expression. During AAA formation, treatment with KMUP-3 inhibited phosphorylated mTOR expression and increased phosphorylated AMPK expression in the medial layer. In addition, KMUP-3 treatment suppressed aortic dilatation together with reduction in proinflammatory cytokines and infiltrating macrophages, attenuation of medial VSMC apoptosis and mitigation of reactive oxygen species generation, matrix-degrading proteinase activities, elastin breakdown and vascular calcification. CONCLUSIONS: Treatment with KMUP-3 inhibits aneurysm growth possibly through its interference with signaling pathways involved in VSMC phenotypic switch and apoptosis. These findings provide a proof-of-concept validation for VSMC dysfunction as a potential therapeutic target in AAA.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Apoptosis/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Piperidines/pharmacology , Vascular Calcification/prevention & control , Xanthines/pharmacology , Angiotensin II , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Male , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Rats, Sprague-Dawley , Signal Transduction , Vascular Calcification/metabolism , Vascular Calcification/pathology
4.
Eur J Vasc Endovasc Surg ; 59(6): 990-999, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32033870

ABSTRACT

OBJECTIVE: Evidence suggests that cathepsin S (CTSS), a potent mammalian elastase, participates in abdominal aortic aneurysm (AAA) formation. This study examines the hypothesis that pharmacological inhibition of CTSS with an α-ketoamide based compound 6r might suppress AAA in mice. METHODS: Experimental study of the CaCl2 induced AAA model in B6 mice and angiotensin II (AngII) infused AAA model in ApoE-/- mice. The effects of intraperitoneal administration of 6r (25 mg/kg) and vehicle every three days since one day after AAA induction were evaluated at 28 days using CaCl2 induced (n = 12 per group) and AngII infused (n = 8 per group) models. Additionally, the effects of post-treatment with 6r and vehicle from seven days or 14 days after AAA induction were evaluated at 28 days using the CaCl2 induced model (n = 6 per group). Aortic samples were harvested for histological and biochemical analyses, including cathepsin levels, Verhoeff Van Gieson staining, TUNEL assay, and immunostaining for macrophages. RESULTS: In the CaCl2 induced model, treatment with 6r suppressed aortic dilatation observed in vehicle treated controls (median: 0.58 vs. 0.92 mm; p < .001), along with reduced CTSS and cathepsin K (CTSK) levels (both p < .001), preserved elastin integrity (p < .001), fewer medial apoptotic cells (p = .012) and less macrophage infiltration (p = .041). In the AngII infused model, the aortic diameter was smaller in 6r treated mice than in vehicle treated controls (median: 0.95 vs. 1.84 mm; p = .047). The levels of CTSS (p < .001) and CTSK (p = .033) and the numbers of elastin breaks (p < .001), medial apoptotic cells (p < .001) and infiltrating macrophages (p = .030) were attenuated under 6r treatment. Finally, post-treatment with 6r from seven days (p = .046) or 14 days (p = .012) after AAA induction limited CaCl2 induced AAA. CONCLUSION: Pharmacological inhibition of CTSS by 6r suppresses AAA formation in mice. Also, post-treatment with 6r retards mouse AAA progression. These findings provide proof of concept validation for CTSS as a potential therapeutic target in AAA.


Subject(s)
Amides/administration & dosage , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/drug therapy , Cathepsins/antagonists & inhibitors , Angiotensin II/toxicity , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Calcium Chloride/toxicity , Cathepsins/metabolism , Disease Models, Animal , Disease Progression , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Up-Regulation
5.
Atherosclerosis ; 262: 62-70, 2017 07.
Article in English | MEDLINE | ID: mdl-28525804

ABSTRACT

BACKGROUND AND AIMS: Thrombomodulin (TM), through its lectin-like domain (TMD1), sequesters proinflammatory high-mobility group box 1 (HMGB1) to prevent it from engaging the receptor for advanced glycation end product (RAGE) that sustains inflammation and tissue damage. Our previous study demonstrated that short-term treatment with recombinant TM containing all the extracellular domains (i.e., rTMD123) inhibits HMGB1-RAGE signaling and confers protection against CaCl2-induced AAA formation. In this study, we attempted to further optimize TM domains, as a potential therapeutic agent for AAA, using the recombinant adeno-associated virus (AAV) vector. METHODS: The therapeutic effects of recombinant TMD1 (rTMD1) and recombinant AAV vectors carrying the lectin-like domain of TM (rAAV-TMD1) were evaluated in the CaCl2-induced AAA model and angiotensin II-infused AAA model, respectively. RESULTS: In the CaCl2-induced model, treatment with rTMD1 suppressed the tissue levels of HMGB1 and RAGE, macrophage accumulation, elastin destruction and AAA formation, and the effects were comparable to a mole-equivalent dosage of rTMD123. In the angiotensin II-infused model, a single intravenous injection of rAAV-TMD1 (1011 genome copies), which resulted in a persistently high serum level of TMD1 for at least 12 weeks, effectively attenuated AAA formation with suppression of HMGB1 and RAGE levels and inhibition of proinflammatory cytokine production, macrophage accumulation, matrix metalloproteinase activities and oxidative stress in the aortic wall. CONCLUSIONS: These findings corroborate the therapeutic potential of the TM lectin-like domain in AAA. The attenuation of angiotensin II-infused AAA by one-time delivery of rAAV-TMD1 provides a proof-of-concept validation of its application as potential gene therapy for aneurysm development.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Thrombomodulin/genetics , Angiotensin II , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Calcium Chloride , Cytokines/metabolism , Disease Models, Animal , Elastin/metabolism , HMGB1 Protein/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinases/metabolism , Mice, Knockout, ApoE , Oxidative Stress , Protein Domains , Receptor for Advanced Glycation End Products/metabolism , Thrombomodulin/biosynthesis , Thrombomodulin/metabolism , Vascular Remodeling
6.
PLoS One ; 11(1): e0146565, 2016.
Article in English | MEDLINE | ID: mdl-26741694

ABSTRACT

Toll-like receptor (TLR) family plays a key role in innate immunity and various inflammatory responses. TLR4, one of the well-characterized pattern-recognition receptors, can be activated by endogenous damage-associated molecular pattern molecules such as high mobility group box 1 (HMGB1) to sustain sterile inflammation. Evidence suggested that blockade of TLR4 signaling may confer protection against abdominal aortic aneurysm (AAA). Herein we aimed to obtain further insight into the mechanism by which TLR4 might promote aneurysm formation. Characterization of the CaCl2-induced AAA model in mice revealed that upregulation of TLR4 expression, localized predominantly to vascular smooth muscle cells (VSMCs), was followed by a late decline during a 28-day period of AAA development. In vitro, TLR4 expression was increased in VSMCs treated with HMGB1. Knockdown of TLR4 by siRNA attenuated HMGB1-enhanced production of proinflammatory cytokines, specifically interleukin-6 and monocyte chemoattractant protein-1 (MCP-1), and matrix-degrading matrix metalloproteinase (MMP)-2 from VSMCs. In vivo, two different strains of TLR4-deficient (C57BL/10ScNJ and C3H/HeJ) mice were resistant to CaCl2-induced AAA formation compared to their respective controls (C57BL/10ScSnJ and C3H/HeN). Knockout of TLR4 reduced interleukin-6 and MCP-1 levels and HMGB1 expression, attenuated macrophage accumulation, and eventually suppressed MMP production, elastin destruction and VSMC loss. Finally, human AAA exhibited higher TLR4 expression that was localized to VSMCs. These data suggest that TLR4 signaling contributes to AAA formation by promoting a proinflammatory status of VSMCs and by inducing proteinase release from VSMCs during aneurysm initiation and development.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Toll-Like Receptor 4/physiology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Calcium Chloride , Case-Control Studies , Cells, Cultured , Cytokines/biosynthesis , HMGB1 Protein/metabolism , Humans , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred C3H , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Up-Regulation
7.
Hum Gene Ther ; 26(9): 603-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25950911

ABSTRACT

Angiostatin and other plasminogen derivatives exhibit antitumor activities directly or indirectly, have demonstrated promising anticancer effects in preclinical studies, but have mostly failed in clinical trials partly due to their short serum half-lives. Our previous studies demonstrated that recombinant human plasminogen kringle 1-5 (K1-5) has superior antitumor activity compared with angiostatin. In addition, optimization of recombinant K1-5 with three amino acid substitutions enhances its antitumor effect. The current study was thus undertaken to evaluate prolonged expression of optimized K1-5 as cancer gene therapy. The recombinant adeno-associated virus (AAV) vector was used to express a secreted form of the optimized K1-5 (AAV-sK15tm) to improve its pharmacokinetic profile, which was considered to be the hurdle in angiostatin treatment of cancer. We successfully generated high-titer recombinant AAV vectors and observed sustained transgene expression for 567 days after a single injection of virus. The treated animals did not display any visible signs of abnormalities and showed normal serum biochemistry. The therapeutic potential of this treatment modality was demonstrated by both a strong inhibition of lung metastasis in the mouse B16F10 melanoma model and significant growth retardation of Lewis lung carcinoma xenografts in C57BL/6N mice as well as human A2058 melanoma xenografts in NOD/SCID (nonobese diabetic/severe combined immunodeficient) mice. Taken together, our results suggested that AAV-sK15tm produced long-term suppressive effects on cancer growth in vivo and should warrant serious consideration for clinical development.


Subject(s)
Carcinoma, Lewis Lung/therapy , Dependovirus/genetics , Genetic Therapy , Lung Neoplasms/prevention & control , Melanoma/therapy , Plasminogen/genetics , Animals , Apoptosis , Gene Expression , HEK293 Cells , Humans , Lung Neoplasms/secondary , Male , Melanoma/pathology , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic , Protein Structure, Tertiary
8.
PLoS One ; 10(3): e0122491, 2015.
Article in English | MEDLINE | ID: mdl-25816372

ABSTRACT

PURPOSE: To determine the role of thrombomodulin (TM) in corneal epithelial wound healing, and to investigate whether recombinant TM epidermal growth factor-like domain plus serine/threonine-rich domain (rTMD23) has therapeutic potential in corneal epithelial wound healing. METHODS: TM localization and expression in the murine cornea were examined by immunofluorescence staining. TM expression after injury was also studied. The effect of rTMD23 on corneal wound healing was evaluated by in vitro and in vivo assays. RESULTS: TM was expressed in the cornea in normal adult mice. TM expression increased in the early phase of wound healing and decreased after wound recovery. In the in vitro study, platelet-derived growth factor-BB (PDGF-BB) induced TM expression in murine corneal epithelial cells by mediating E26 transformation-specific sequence-1 (Ets-1) via the mammalian target of rapamycin (mTOR) signaling pathway. The administration of rTMD23 increased the rate of corneal epithelial wound healing. CONCLUSIONS: TM expression in corneal epithelium was modulated during the corneal wound healing process, and may be regulated by PDGF-BB. In addition, rTMD23 has therapeutic potential in corneal injury.


Subject(s)
Corneal Injuries/genetics , Proto-Oncogene Proteins c-sis/genetics , Thrombomodulin/genetics , Wound Healing/genetics , Animals , Becaplermin , Corneal Injuries/pathology , Corneal Injuries/therapy , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Mice , Proto-Oncogene Proteins c-sis/metabolism , Recombinant Proteins/administration & dosage , Signal Transduction/drug effects , Thrombomodulin/administration & dosage , Thrombomodulin/biosynthesis , Wound Healing/drug effects
9.
Cardiovasc Res ; 105(1): 107-17, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25388665

ABSTRACT

AIMS: The recombinant epidermal growth factor-like domain plus the serine/threonine-rich domain of thrombomodulin (rTMD23) promotes angiogenesis and accelerates the generation of activated protein C (APC), which facilitates angiogenesis. The aim of this study was to elucidate the molecular mechanisms underlying the angiogenic activity of rTMD23. METHODS AND RESULTS: We prepared rTMD23 and its mutants that did not possess the ability to promote APC generation and investigated their angiogenic activities in vitro and in vivo. rTMD23 mutants promoted proliferation, migration, and tube formation of human umbilical vein endothelial cells in vitro and induced neovascularization in vivo; these effects were similar to those exerted by wild-type rTMD23. To investigate its interaction with rTMD23, Type I fibroblast growth factor receptor (FGFR1) was precipitated along with syndecan-4 by rTMD23-conjugated Sepharose in human umbilical vein endothelial cells and FGFR1-expressing human embryonic kidney 293 cells. Additionally, the kinetics of the interaction between rTMD23 and FGFR1 were analysed using surface plasmon resonance. rTMD23-induced FGFR1 activation and tube formation were inhibited by an FGFR1-specific tyrosine kinase inhibitor, PD173074, or by knockdown of FGFR1 using siRNA technology. We observed an improvement in rat hindlimb recovery in an ischaemic model following rTMD23 treatment, and this was associated with increased neovascularization and FGFR1 phosphorylation. CONCLUSION: rTMD23 induced angiogenesis via FGFR1, a process that is independent of the APC pathway.


Subject(s)
Neovascularization, Physiologic/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Thrombomodulin/physiology , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacology , Animals , Corneal Neovascularization/etiology , Gene Knockdown Techniques , HEK293 Cells , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/drug therapy , Ischemia/pathology , Ischemia/physiopathology , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Syndecan-4/metabolism , Thrombomodulin/chemistry , Thrombomodulin/genetics
10.
Ann Surg ; 258(6): 1103-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23295319

ABSTRACT

OBJECTIVE: To investigate whether recombinant thrombomodulin containing all the extracellular domains (rTMD123) has therapeutic potential against aneurysm development. SUMMARY BACKGROUND DATA: The pathogenesis of abdominal aortic aneurysm (AAA) is characterized by chronic inflammation and proteolytic degradation of extracellular matrix. Thrombomodulin, a transmembrane glycoprotein, exerts anti-inflammatory activities such as inhibition of cytokine production and sequestration of proinflammatory high-mobility group box 1 (HMGB1) to prevent it from engaging the receptor for advanced glycation end product (RAGE) that may sustain inflammation and tissue damage. METHODS: The in vivo effects of treatment and posttreatment with rTMD123 on aortic dilatation were measured using the CaCl2-induced AAA model in mice. RESULTS: Characterization of the CaCl2-induced model revealed that HMGB1 and RAGE, both localized mainly to macrophages, were persistently upregulated during a 28-day period of AAA development. In vitro, rTMD123-HMGB1 interaction prevented HMGB1 binding to macrophages, thereby prohibiting activation of HMGB1-RAGE signaling in macrophages. In vivo, short-term treatment with rTMD123 upon AAA induction suppressed the levels of proinflammatory cytokines, HMGB1, and RAGE in the aortic tissue; reduced the infiltrating macrophage number; and finally attenuated matrix metalloproteinase production, extracellular matrix destruction, and AAA formation without disturbing vascular calcification. Consistently, posttreatment with rTMD123 seven days after AAA induction alleviated vascular inflammation and retarded AAA progression. CONCLUSIONS: These data suggest that rTMD123 confers protection against AAA development. The mechanism of action may be associated with reduction of proinflammatory mediators, blockade of macrophage recruitment, and suppression of HMGB1-RAGE signaling involved in aneurysm formation and downstream macrophage activation.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Thrombomodulin/therapeutic use , Animals , Aortic Aneurysm, Abdominal/chemically induced , Calcium Chloride/administration & dosage , Mice , Mice, Inbred C57BL , Receptor for Advanced Glycation End Products , Receptors, Immunologic/physiology , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...