1
|
Kaplan AP, Ghebrehiwet B, Joseph K. Alzheimer's Disease: In Vitro and In Vivo Evidence of Activation of the Plasma Bradykinin-Forming Cascade and Implications for Therapy. Cells 2024; 13:2039. [PMID: 39768131 PMCID: PMC11674638 DOI: 10.3390/cells13242039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
The plaques associated with Alzheimer's disease are formed as a result of the aggregation of Aβ peptides, which vary in length from 38 to 43 amino acids. The 1-40 peptide is the most abundant, while the 1-42 peptide appears to be the most destructive to neurons and/or glial cells in a variety of assays. We have demonstrated that aggregated Aβ, a state prior to plaque formation, will activate the plasma bradykinin-forming pathway when tested in vitro. Aggregation is zinc-dependent, optimal at 25-50 µM, and the rate of aggregation is paralleled by the rate of activation of the bradykinin-forming pathway as assessed by plasma kallikrein formation. The aggregation of Aβ 1-38, 1-40, and 1-42 is optimal after incubation for 3 days, 3 h, and under 1 min, respectively. The cascade is initiated by the autoactivation of factor XII upon binding to aggregated Aβ; then, prekallikrein is converted to kallikrein, which cleaves high-molecular-weight kininogen (HK) to release bradykinin. Studies by a variety of other researchers have demonstrated the presence of each "activation-step" in either the plasma or spinal fluid of patients with Alzheimer's disease, including activated factor XII, kallikrein, and bradykinin itself. There is also evidence that activation is more prominent as dementia worsens. We now have medications that can block each step of the bradykinin-forming pathway as currently employed for the therapy of hereditary angioedema. Given the current state of therapy for Alzheimer's disease, which includes monoclonal antibodies that retard the rate of progression by 30% at most and have significant side effects, it seems imperative to explore prophylaxis using one of the long-acting agents that target plasma kallikrein or factor XIIa. There is a long-acting bradykinin antagonist in development, and techniques to target kallikrein mRNA to lower levels or knock out the prekallikrein gene are being developed.
Collapse
Affiliation(s)
- Allen P. Kaplan
- Department of Medicine, The Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
2
|
Komar AA. Molecular Peptide Grafting as a Tool to Create Novel Protein Therapeutics. Molecules 2023; 28:2383. [PMID: 36903628 PMCID: PMC10005171 DOI: 10.3390/molecules28052383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The study of peptides (synthetic or corresponding to discrete regions of proteins) has facilitated the understanding of protein structure-activity relationships. Short peptides can also be used as powerful therapeutic agents. However, the functional activity of many short peptides is usually substantially lower than that of their parental proteins. This is (as a rule) due to their diminished structural organization, stability, and solubility often leading to an enhanced propensity for aggregation. Several approaches have emerged to overcome these limitations, which are aimed at imposing structural constraints into the backbone and/or sidechains of the therapeutic peptides (such as molecular stapling, peptide backbone circularization and molecular grafting), therefore enforcing their biologically active conformation and thus improving their solubility, stability, and functional activity. This review provides a short summary of approaches aimed at enhancing the biological activity of short functional peptides with a particular focus on the peptide grafting approach, whereby a functional peptide is inserted into a scaffold molecule. Intra-backbone insertions of short therapeutic peptides into scaffold proteins have been shown to enhance their activity and render them a more stable and biologically active conformation.
Collapse
Affiliation(s)
- Anton A. Komar
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological and Environmental Sciences, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA; ; Tel.: +1-216-687-2516
- Department of Biochemistry and Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
3
|
Kaplan AP, Joseph K, Ghebrehiwet B. The complex role of kininogens in hereditary angioedema. FRONTIERS IN ALLERGY 2022; 3:952753. [PMID: 35991308 PMCID: PMC9382879 DOI: 10.3389/falgy.2022.952753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Human high molecular weight kininogen (HK) is the substrate from which bradykinin is released as a result of activation of the plasma "contact" system, a cascade that includes the intrinsic coagulation pathway, and a fibrinolytic pathway leading to the conversion of plasminogen to plasmin. Its distinction from low molecular weight kininogen (LK) was first made clear in studies of bovine plasma. While early studies did suggest two kininogens in human plasma also, their distinction became clear when plasma deficient in HK or both HK and LK were discovered. The light chain of HK is distinct and has the site of interaction with negatively charged surfaces (domain 5) plus a 6th domain that binds either prekallikrein or factor XI. HK is a cofactor for multiple enzymatic reactions that relate to the light chain binding properties. It augments the rate of conversion of prekallikrein to kallikrein and is essential for the activation of factor XI. It indirectly augments the "feedback" activation of factor XII by plasma kallikrein. Thus, HK deficiency has abnormalities of intrinsic coagulation and fibrinolysis akin to that of factor XII deficiency in addition to the inability to produce bradykinin by factor XII-dependent reactions. The contact cascade binds to vascular endothelial cells and HK is a critical binding factor with binding sites within domains 3 and 5. Prekallikrein (or factor XI) is attached to HK and is brought to the surface. The endothelial cell also secretes proteins that interact with the HK-prekallikrein complex resulting in kallikrein formation. These have been identified to be heat shock protein 90 (HSP 90) and prolylcarboxypeptidase. Cell release of urokinase plasminogen activator stimulates fibrinolysis. There are now 6 types of HAE with normal C1 inhibitors. One of them has a mutated kininogen but the mechanism for overproduction (presumed) of bradykinin has not yet been determined. A second has a mutation involving sulfation of proteoglycans which may lead to augmented bradykinin formation employing the cell surface reactions noted above.
Collapse
Affiliation(s)
- Allen P. Kaplan
- Medicine/Pulmonary and Critical Care, Medical University of South Carolina, Charleston, SC, United States
| | | | - Berhane Ghebrehiwet
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
4
|
Bioinformatics Study Identified EGF as a Crucial Gene in Papillary Renal Cell Cancer. DISEASE MARKERS 2022; 2022:4761803. [PMID: 35655917 PMCID: PMC9155928 DOI: 10.1155/2022/4761803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022]
Abstract
Background Due to a lack of knowledge of the disease process, papillary renal cell carcinoma (PRCC) has a dismal outlook. This research was aimed at uncovering the possible biomarkers and the underlying principles in PRCC using a bioinformatics method. Methods We searched the Gene Expression Omnibus (GEO) datasets to obtain the GSE11151 and GSE15641 gene expression profiles of PRCC. We used the R package limma to identify the differentially expressed genes (DEGs). The online tool DAVID and ClusterProfiler package in R software were used to analyze Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway dominance, respectively. The STRING database was utilized to construct the PPI network of DEGs. Using the Cytoscape technology, a protein-protein interaction (PPI) network that associated with DEGs was created, and the hub genes were identified using the Cytoscape plug-in CytoHubba. The hub genes were subjected to a Kaplan-Meier analysis to identify their correlations with survival rates. Results From the selected datasets, a total of 240 common DEGs were identified in the PRCC, including 50 upregulated genes and 190 downregulated regulated genes. Renal growth, external exosome, binding of heparin, and metabolic processes were all substantially associated with DEGs. The CytoHubba plug-in-based analysis identified the 10 hub genes (ALB, KNG1, C3, CXCL12, EGF, TIMP1, VCAN, PLG, LAMC1, and CASR) from the original PPI network. The higher expression group of EGF was associated with poor outcome in patients with PRCC. Conclusions We revealed important genes and proposed biological pathways that may be implicated in the formation of PRCC. EGF might be a predictive biomarker for PRCC and therefore should be investigated as a novel treatment strategy.
Collapse
|
5
|
Droll SH, Sheng Hsu YM, Drake SK, Kim A, Wang W, Calvo KR, Cao Z, Hu TY, Zhao Z. Differential processing of high-molecular-weight kininogen during normal pregnancy. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 1:e8552. [PMID: 31412146 PMCID: PMC7018535 DOI: 10.1002/rcm.8552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/18/2019] [Accepted: 08/07/2019] [Indexed: 06/10/2023]
Abstract
RATIONALE Studies identified kininogen as a potential biomarker of preeclampsia, a major cause of adverse maternal outcomes. High-molecular-weight kininogen (HK) and its activated form participate in numerous pathways associated with establishing and maintaining pregnancy. However, dynamic changes in HK and naturally occurring HK-derived peptides during the natural course of pregnancy are largely unknown. METHODS Longitudinal serum samples during the course of normal pregnancy (trimesters T1, T2, T3) from 60 pregnant women were analyzed by western blot with an anti-HK antibody. Circulating peptides in longitudinal serum specimens derived from 50 participants were enriched using nanoporous silica thin films. Peptides were identified by liquid chromatography/tandem mass spectrometry (LC/MS/MS) and database searching. Relative quantification was performed using MaxQuant and in-house scripts. Normality was evaluated by either ANOVA or Friedman tests with p < 0.05 for statistical significance. RESULTS Western blotting revealed that HK significantly decreased during normal pregnancy (T1 vs T2, p < 0.05; T1 vs T3, p < 0.0001). A 100 kDa intermediate increased during pregnancy (T1 vs T2, p < 0.005; T1 vs T3, p < 0.01). Moreover, the heavy chain (T1 vs T2, p < 0.0001; T1 vs T3, p < 0.0001; T2 vs T3, p < 0.01) and light chain (T1 vs T2, p < 0.0001; T1 vs T3, p < 0.0001; T2 vs T3, p < 0.05) significantly increased during pregnancy. LC/MS/MS analysis identified 180 kininogen-1 peptides, of which 167 mapped to domain 5 (D5). Seventy-three peptides with ten or more complete data sets were included for further analysis. Seventy peptides mapped to D5, and 3, 24, and 43 peptides showed significant decrease, no trend, and significant increase, respectively, during pregnancy. CONCLUSIONS This study demonstrates dynamic changes in HK and naturally occurring HK-derived peptides during pregnancy. Our study sheds light on the gestational changes of HK and its peptides for further validation of them as potential biomarkers for pregnancy-related complications.
Collapse
Affiliation(s)
- Stephenie H. Droll
- Chemistry Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, 20892 MD, USA
- IBiS - Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208-3500
| | - Yen-Michael Sheng Hsu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Steven K. Drake
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ashley Kim
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
| | - Weixin Wang
- Hematology Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Katherine R. Calvo
- Hematology Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Zheng Cao
- Department of Laboratory Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Tony Y Hu
- Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281
| | - Zhen Zhao
- Chemistry Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, 20892 MD, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
6
|
Identification and Analysis of Novel Biomarkers Involved in Chromophobe Renal Cell Carcinoma by Integrated Bioinformatics Analyses. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2671281. [PMID: 32090070 PMCID: PMC7029304 DOI: 10.1155/2020/2671281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/06/2020] [Indexed: 12/26/2022]
Abstract
In renal cell carcinoma, chromophobe renal cell carcinoma (ChRCC) is a distinct subtype, whose clinical manifestations often lack specificity, and the molecular mechanisms of ChRCC tumorigenesis remain generally vague. The target of this study was to discover novel biomarkers involved in ChRCC by integrated bioinformatics analyses. We found 2608 differentially expressed genes (DEGs), of which 1518 were upregulated and 1090 were downregulated. Gene ontology (GO) analysis of DEGs uncovered significant functional enrichment in three aspects: biological process (BP), molecular function (MF), and cellular component (CC). The results of Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated DEGs were largely enriched in retinol metabolism, arachidonic acid metabolism, and pentose and glucuronate interconversions. Then, the protein–protein interactions (PPI) network was constructed and top three hub genes were identified by the Cytoscape plugin cytoHubba. Through calculating the degree, betweenness centrality, and Stress of mRNAs, CENPA was upregulated and KNG1 and AGT were downregulated. A survival assay performed according to Oncomine data showed only CENPA high expression exhibited a worse prognosis. This study identified crucial genes and pathways for the progress of ChRCC, and CENPA might be a novel biomarker for diagnosis, treatment, and prognosis of ChRCC.
Collapse
|
7
|
Xu J, Fang J, Cheng Z, Fan L, Hu W, Zhou F, Shen H. Overexpression of the Kininogen-1 inhibits proliferation and induces apoptosis of glioma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:180. [PMID: 30068373 PMCID: PMC6090912 DOI: 10.1186/s13046-018-0833-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
Background Glioma is the most common primary central nervous system tumor derived from glial cells. Kininogen-1 (KNG1) can exert antiangiogenic properties and inhibit proliferation of endothelial cells. The effect of KNG1 on the glioma is rarely reported, so our purpose in to explore its mechanism in glioma cells. Methods The differentially expressed genes (DEGs) were identified based on The Cancer Genome Atlas (TCGA) database. The KNG1-vector was transfected into the two glioma cells. The viability, apoptosis and cell cycle of glioma cells and microvessel density (MVD) were detected by cell counting kit-8 assay, flow cytometry and immunohistochemistry, respectively. The expression were measured by quantitative real-time PCR and Western blot, respectively. A tumor mouse model was established to determine apoptosis rate of brain tissue by terminal deoxynucleotidyl transfer-mediated dUTP nick end labeling (TUNEL) analysis. Results KNG1 was identified as the core gene and lowly expressed in the glioma cells. Overexpression of KNG1 inhibited cell viability and angiogenesis of glioma cells. Overexpression of KNG1 promoted the apoptosis and G1 phase cell cycle arrest of glioma cells. Moreover, the expressions of VEGF, cyclinD1, ki67, caspase-3/9 and XIAP were regulated by overexpression of KNG1. In addition, overexpression of KNG1 inhibited the activity of PI3K/Akt. Furthermore, overexpression of KNG1 decreased the tumor growth and promoted the apoptosis of decreased by overexpression of KNG1 in vivo. . Conclusions Overexpression of KNG1 suppresses glioma progression by inhibiting the proliferation and promoting apoptosis of glioma cells, providing a therapeutic strategy for the malignant glioma.
Collapse
Affiliation(s)
- Jinfang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Jun Fang
- Department of Radiotherapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang Province, 310022, China
| | - Zhonghao Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Longlong Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Weiwei Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Feng Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China.
| | - Hong Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| |
Collapse
|
8
|
Zhao K, Yuan Y, Lin B, Miao Z, Li Z, Guo Q, Lu N. LW-215, a newly synthesized flavonoid, exhibits potent anti-angiogenic activity in vitro and in vivo. Gene 2017; 642:533-541. [PMID: 29196258 DOI: 10.1016/j.gene.2017.11.065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/25/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
LW-215 is a newly synthesized flavonoid, which is the derivative of wogonin. Our group has previously confirmed that wogonin has an anti-angiogenic activity, while the anti-angiogenic effect of LW-215 is unclear. In this study, we explored whether LW-215 can inhibit angiogenesis and further probed the potential molecular mechanisms. We found that LW-215 inhibited migration and tube formation in human umbilical vein endothelial cells (HUVECs) and immortalized endothelial EA.hy926 cells without a significant decrease in cell viability. Microvessels sprouting from rat aortic ring and chicken chorioallantoic membrane (CAM) model also revealed that LW-215 could suppress angiogenesis in vivo. Western blot and ELISA analysis indicated that LW-215 could prevent VEGFR2 activation though reducing VEGF autocrine other than VEGFR1. Thus, its downstream kinases, such as Akt, ERK and p38 signaling, were inhibited. Taken together, these results fully showed that LW-215 might be a promising anti-angiogenesis agent.
Collapse
Affiliation(s)
- Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Yang Yuan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Binyan Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Zhaorui Miao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Zhiyu Li
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China.
| |
Collapse
|
9
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
10
|
Betapudi V, Shukla M, Alluri R, Merkulov S, McCrae KR. Novel role for p56/Lck in regulation of endothelial cell survival and angiogenesis. FASEB J 2016; 30:3515-3526. [PMID: 27402674 DOI: 10.1096/fj.201500040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 06/28/2016] [Indexed: 01/08/2023]
Abstract
Previous studies have demonstrated that cleaved high-molecular-weight kininogen (HKa) induces endothelial apoptosis and inhibits angiogenesis and have suggested that this occurs through inhibition of Src family kinases. This study assessed the role of tyrosine-protein kinase Lck (p56/Lck) in this pathway. We analyzed early events leading to apoptosis of human endothelial cells exposed to HKa. The role of p56/Lck was investigated using short interfering (si) RNA knockdown and lentivirus expression in assays of endothelial tube formation, sprouting of neovessels from murine aorta, and angiogenesis in Matrigel plugs. HKa stimulated expression and phosphorylation of p56/Lck. siRNA knockdown of p56/Lck promoted endothelial proliferation and blocked HKa-induced apoptosis and activation of p53, Bax, and Bak. Lentivirus expression of p56/Lck in endothelial cells induced apoptosis and blocked tube formation. Expression of p56/Lck in murine aortic rings blocked sprouting angiogenesis. Lentivirus expressing p56/Lck blocked angiogenesis in Matrigel plugs, while p56/Lck short hairpin RNA inhibited the antiangiogenic effect of HKa. Scrambled siRNAs and empty lentiviral vectors were used in all experiments. Apoptosis of proliferating endothelial cells and inhibition of angiogenesis by HKa requires p56/Lck. This suggests a novel role for p56/Lck in regulation of endothelial cell survival and angiogenesis.-Betapudi, V., Shukla, M., Alluri, R., Merkulov, S., McCrae, K. R. Novel role for p56/Lck in regulation of endothelial cell survival and angiogenesis.
Collapse
Affiliation(s)
- Venkaiah Betapudi
- Department of Cellular and Molecular Medicine Cleveland Clinic, Cleveland, Ohio, USA; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA; and
| | - Meenal Shukla
- Department of Cellular and Molecular Medicine Cleveland Clinic, Cleveland, Ohio, USA
| | - Ravi Alluri
- Department of Cellular and Molecular Medicine Cleveland Clinic, Cleveland, Ohio, USA
| | - Sergei Merkulov
- Department of Cellular and Molecular Medicine Cleveland Clinic, Cleveland, Ohio, USA
| | - Keith R McCrae
- Department of Cellular and Molecular Medicine Cleveland Clinic, Cleveland, Ohio, USA; Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
11
|
Fan J, Tea MKM, Yang C, Ma L, Meng QH, Hu TY, Singer CF, Ferrari M. Profiling of Cross-Functional Peptidases Regulated Circulating Peptides in BRCA1 Mutant Breast Cancer. J Proteome Res 2016; 15:1534-45. [PMID: 27058005 PMCID: PMC5124559 DOI: 10.1021/acs.jproteome.6b00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Women with inherited BRCA1 mutations are more likely to develop breast cancer (BC); however, not every carrier will progress to BC. The aim of this study was to identify and characterize circulating peptides that correlate with BC patients carrying BRCA1 mutations. Circulating peptides were enriched using our well-designed nanoporous silica thin films (NanoTraps) and profiled by mass spectrometry to identify among four clinical groups. To determine the corresponding proteolytic processes and their sites of activity, purified candidate peptidases and synthesized substrates were assayed to verify the processes predicted by the MERPOS database. Proteolytic processes were validated using patient serum samples. The peptides, KNG1K438-R457 and C 3fS1304-R1320, were identified as putative peptide candidates to differentiate BRCA1 mutant BC from sporadic BC and cancer-free BRCA1 mutant carriers. Kallikrein-2 (KLK2) is the major peptidase that cleaves KNG1K438-R457 from kininogen-1, and its expressions and activities were also found to be dependent on BRCA1 status. We further determined that KNG1K438-R457 is cleaved at its C-terminal arginine by carboxypeptidase N1 (CPN1). Increased KLK2 activity, with decreased CPN1 activity, results in the accumulation of KNG1K438-R457 in BRCA1-associated BC. Our work outlined a useful strategy for determining the peptide-petidase relationship and thus establishing a biological mechanism for changes in the peptidome in BRCA1-associated BC.
Collapse
Affiliation(s)
- Jia Fan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Muy-Kheng M. Tea
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Division of Senology, Medical University of Vienna, Vienna 1090, Austria
| | - Chuan Yang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Qing H. Meng
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Tony Y. Hu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, New York 10021, United States
| | - Christian F. Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Division of Senology, Medical University of Vienna, Vienna 1090, Austria
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Internal Medicine, Weill Cornell Medical College of Cornell University, New York, New York 10021, United States
| |
Collapse
|
12
|
Ponraj SB, Chen Z, Li LH, Shankaranarayanan JS, Rajmohan GD, du Plessis J, Sinclair AJ, Chen Y, Wang X, Kanwar JR, Dai XJ. Fabrication of boron nitride nanotube-gold nanoparticle hybrids using pulsed plasma in liquid. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:10712-10720. [PMID: 25127006 DOI: 10.1021/la502960h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Plasma, generated in liquid at atmospheric pressure by a nanosecond pulsed voltage, was used to fabricate hybrid structures from boron nitride nanotubes and gold nanoparticles in deionized water. The pH was greatly reduced, conductivity was significantly increased, and concentrations of reactive oxygen and nitrogen species in the water were increased by the plasma treatment. The treatment reduced the length of the nanotubes, giving more individual cuplike structures, and introduced functional groups onto the surface. Gold nanoparticles were successively assembled onto the functionalized surfaces. The reactive species from the liquid plasma along with the nanosecond pulsed electric field seem to play a role in the shortening and functionalization of the nanotubes and the assembly of gold nanoparticles. The potential for targeted drug delivery was tested in a preliminary investigation using doxorubicin-loaded plasma-treated nanotubes which were effective at killing ∼99% of prostate cancer cells.
Collapse
Affiliation(s)
- Sri balaji Ponraj
- Institute for Frontier Materials and ‡School of Medicine, Deakin University , Waurn Ponds Campus, Geelong, VIC 3216, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Huhn AJ, Parsonage D, Horita DA, Torti FM, Torti SV, Hollis T. The high-molecular-weight kininogen domain 5 is an intrinsically unstructured protein and its interaction with ferritin is metal mediated. Protein Sci 2014; 23:1013-22. [PMID: 24810540 DOI: 10.1002/pro.2486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/06/2014] [Accepted: 05/06/2014] [Indexed: 12/30/2022]
Abstract
High-molecular-weight kininogen domain 5 (HK5) is an angiogenic modulator that is capable of inhibiting endothelial cell proliferation, migration, adhesion, and tube formation. Ferritin can bind to a histidine-glycine-lysine-rich region within HK5 and block its antiangiogenic effects. However, the molecular intricacies of this interaction are not well understood. Analysis of the structure of HK5 using circular dichroism and nuclear magnetic resonance [(1) H, (15) N]-heteronuclear single quantum coherence determined that HK5 is an intrinsically unstructured protein, consistent with secondary structure predictions. Equilibrium binding studies using fluorescence anisotropy were used to study the interaction between ferritin and HK5. The interaction between the two proteins is mediated by metal ions such as Co(2+) , Cd(2+) , and Fe(2+) . This metal-mediated interaction works independently of the loaded ferrihydrite core of ferritin and is demonstrated to be a surface interaction. Ferritin H and L bind to HK5 with similar affinity in the presence of metals. The ferritin interaction with HK5 is the first biological function shown to occur on the surface of ferritin using its surface-bound metals.
Collapse
Affiliation(s)
- Annissa J Huhn
- Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | |
Collapse
|
14
|
Larusch GA, Merkulova A, Mahdi F, Shariat-Madar Z, Sitrin RG, Cines DB, Schmaier AH. Domain 2 of uPAR regulates single-chain urokinase-mediated angiogenesis through β1-integrin and VEGFR2. Am J Physiol Heart Circ Physiol 2013; 305:H305-20. [PMID: 23709605 DOI: 10.1152/ajpheart.00110.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How single-chain urokinase (ScuPA) mediates angiogenesis is incompletely understood. ScuPA (≥4 nM) induces phosphorylated (p)ERK1/2 (MAPK44 and MAPK42) and pAkt (Ser(473)) in umbilical vein and dermal microvascular endothelial cells. Activation of pERK1/2 by ScuPA is blocked by PD-98059 or U-0126, and pAkt (Ser(473)) activation is inhibited by wortmannin or LY-294002. ScuPA (32 nM) or protease-inhibited two-chain urokinase stimulates pERK1/2 to the same extent, indicating that signaling is not dependent on enzymatic activity. ScuPA induces pERK1/2, but not pAkt (Ser(473)), in SIN1(-/-) cells, indicating that the two pathways are not identical. Peptides from domain 2 of the urokinase plasminogen activator receptor (uPAR) or domain 5 of high-molecular-weight kininogen compete with ScuPA for the induction of pERK1/2 and pAkt (Ser(473)). A peptide of the integrin-binding site on uPAR, a β1-integrin peptide that binds uPAR, antibody 6S6 to β1-integrin, tyrosine kinase inhibitors AG-1478 or PP3, and small interfering RNA knockdown of VEFG receptor 2, but not HER1-HER4, blocked ScuPA-induced pERK1/2 and pAkt (Ser(473)). ScuPA-induced endothelial cell proliferation was blocked by inhibitors of pERK1/2 and pAkt (Ser(473)), antibody 6S6, and uPAR or kininogen peptides. ScuPA initiated aortic sprouts and Matrigel plug angiogenesis in normal, but not uPAR-deficient, mouse aortae or mice, respectively, but these were blocked by PD-98059, LY-294002, AG-1478, or cleaved high-molecular-weight kininogen. In summary, this investigation indicates a novel, a nonproteolytic signaling pathway initiated by zymogen ScuPA and mediated by domain 2 of uPAR, β1-integrins, and VEGF receptor 2 leading to angiogenesis. Kininogens or peptides from it downregulate this pathway.
Collapse
Affiliation(s)
- Gretchen A Larusch
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Kashuba E, Bailey J, Allsup D, Cawkwell L. The kinin-kallikrein system: physiological roles, pathophysiology and its relationship to cancer biomarkers. Biomarkers 2013; 18:279-96. [PMID: 23672534 DOI: 10.3109/1354750x.2013.787544] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The kinin-kallikrein system (KKS) is an endogenous multiprotein cascade, the activation of which leads to triggering of the intrinsic coagulation pathway and enzymatic hydrolysis of kininogens with the consequent release of bradykinin-related peptides. This system plays a crucial role in inflammation, vasodilation, smooth muscle contraction, cardioprotection, vascular permeability, blood pressure control, coagulation and pain. In this review, we will outline the physiology and pathophysiology of the KKS and also highlight the association of this system with carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Elena Kashuba
- Postgraduate Medical Institute, University of Hull, Hull, UK
| | | | | | | |
Collapse
|
16
|
Hatoh T, Maeda T, Takeuchi K, Ogikubo O, Uchiyama S, Otsuka T, Ohkubo I, Ogita H. Domain 5 of high molecular weight kininogen inhibits collagen-mediated cancer cell adhesion and invasion in association with α-actinin-4. Biochem Biophys Res Commun 2012; 427:497-502. [PMID: 23000411 DOI: 10.1016/j.bbrc.2012.09.079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 09/13/2012] [Indexed: 11/19/2022]
Abstract
High molecular weight kininogen (HK) is a plasma glycoprotein with multiple functions, including the regulation of coagulation. We previously demonstrated that domain 5 (D5(H)), a functional domain of HK, and its derived peptides played an important role in the vitronectin-mediated suppression of cancer cell adhesion and invasion. However, the underlying mechanisms of the D5(H)-mediated suppressive effects remain to be elucidated. Here, we showed that D5(H) and its derivatives inhibited the collagen-mediated cell adhesion and invasion of human osteosarcoma MG63 cells. Using purified D5(H) fused to glutathione-S-transferase (GST) and D5(H)-derived peptides for column chromatography, an actin-binding protein, α-actinin-4, was identified as a binding protein of D5(H) with high-affinity for P-5m, a core octapeptide of D5(H). Immunofluorescence microscopy demonstrated that D5(H) co-localized with α-actinin-4 inside MG63 cells. In addition, exogenous GST-D5(H) added to the culture media was transported into MG63 cells, although GST alone as a control was not. As α-actinin-4 regulates actin polymerization necessary for cell adhesion and is related to the integrin-dependent attachment of cells to the extracellular matrix, our results suggest that D5(H) may modulate cell adhesion and invasion together with actinin-4.
Collapse
Affiliation(s)
- Tsunetoshi Hatoh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Tesfay L, Huhn AJ, Hatcher H, Torti FM, Torti SV. Ferritin blocks inhibitory effects of two-chain high molecular weight kininogen (HKa) on adhesion and survival signaling in endothelial cells. PLoS One 2012; 7:e40030. [PMID: 22768328 PMCID: PMC3388046 DOI: 10.1371/journal.pone.0040030] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 06/03/2012] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is tightly regulated through complex crosstalk between pro- and anti-angiogenic signals. High molecular weight kininogen (HK) is an endogenous protein that is proteolytically cleaved in plasma and on endothelial cell surfaces to HKa, an anti-angiogenic protein. Ferritin binds to HKa and blocks its anti-angiogenic activity. Here, we explore mechanisms underlying the cytoprotective effect of ferritin in endothelial cells exposed to HKa. We observe that ferritin promotes adhesion and survival of HKa-treated cells and restores key survival and adhesion signaling pathways mediated by Erk, Akt, FAK and paxillin. We further elucidate structural motifs of both HKa and ferritin that are required for effects on endothelial cells. We identify an histidine-glycine-lysine (HGK) -rich antiproliferative region within domain 5 of HK as the target of ferritin, and demonstrate that both ferritin subunits of the H and L type regulate HKa activity. We further demonstrate that ferritin reduces binding of HKa to endothelial cells and restores the association of uPAR with α5β1 integrin. We propose that ferritin blocks the anti-angiogenic activity of HKa by reducing binding of HKa to UPAR and interfering with anti-adhesive and anti-proliferative signaling of HKa.
Collapse
Affiliation(s)
- Lia Tesfay
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Annissa J. Huhn
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Heather Hatcher
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Frank M. Torti
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Suzy V. Torti
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
18
|
Bentley AA, Merkulov SM, Peng Y, Rozmarynowycz R, Qi X, Pusztai-Carey M, Merrick WC, Yee VC, McCrae KR, Komar AA. Chimeric glutathione S-transferases containing inserts of kininogen peptides: potential novel protein therapeutics. J Biol Chem 2012; 287:22142-50. [PMID: 22577144 DOI: 10.1074/jbc.m112.372854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The study of synthetic peptides corresponding to discrete regions of proteins has facilitated the understanding of protein structure-activity relationships. Short peptides can also be used as powerful therapeutic agents. However, in many instances, small peptides are prone to rapid degradation or aggregation and may lack the conformation required to mimic the functional motifs of the protein. For peptides to function as pharmacologically active agents, efficient production or expression, high solubility, and retention of biological activity through purification and storage steps are required. We report here the design, expression, and functional analysis of eight engineered GST proteins (denoted GSHKTs) in which peptides ranging in size from 8 to 16 amino acids and derived from human high molecular weight kininogen (HK) domain 5 were inserted into GST (between Gly-49 and Leu-50). Peptides derived from HK are known to inhibit cell proliferation, angiogenesis, and tumor metastasis, and the biological activity of the HK peptides was dramatically (>50-fold) enhanced following insertion into GST. GSHKTs are soluble and easily purified from Escherichia coli by affinity chromatography. Functionally, these hybrid proteins cause inhibition of endothelial cell proliferation. Crystallographic analysis of GSHKT10 and GSHKT13 (harboring 10- and 13-residue HK peptides, respectively) showed that the overall GST structure was not perturbed. These results suggest that the therapeutic efficacy of short peptides can be enhanced by insertion into larger proteins that are easily expressed and purified and that GST may potentially be used as such a carrier.
Collapse
Affiliation(s)
- Amber A Bentley
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio 44115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
McMichael M. New Models of Hemostasis. Top Companion Anim Med 2012; 27:40-5. [DOI: 10.1053/j.tcam.2012.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 07/23/2012] [Indexed: 11/11/2022]
|
20
|
Polyphosphate: a link between platelets, coagulation and inflammation. Int J Hematol 2012; 95:346-52. [PMID: 22477540 DOI: 10.1007/s12185-012-1054-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 03/08/2012] [Accepted: 03/08/2012] [Indexed: 10/28/2022]
Abstract
Inorganic polyphosphate (polyP) is abundant in biological organisms. PolyP is a major component of dense granules of human platelets and is secreted upon platelet activation. Studies from our lab and others have shown that polyP is a potent modulator of the blood clotting cascade, acting as a pro-hemostatic, prothrombotic and proinflammatory agent depending on its polymer size and location. PolyP may represent at least one of the long-sought (patho)physiologic activators of the contact pathway of blood clotting, and its actions may also help to explain previously unexplained abilities of activated platelets to enhance plasma clotting reactions. PolyP may have utility as a hemostatic agent to control bleeding, and conversely, polyP antagonists might have utility as antithrombotic/anti-inflammatory agents with reduced bleeding side effects. The detailed molecular mechanisms by which polyP modulates blood clotting reactions still remain to be elucidated.
Collapse
|
21
|
Mousa SA. Comparative Pharmacodynamic Assessment of the Antiangiogenesis Activity of Heparin and Low-Molecular-Weight Heparin Fractions. Clin Appl Thromb Hemost 2012; 19:48-54. [DOI: 10.1177/1076029611436194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shaker A. Mousa
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Pasupuleti M, Schmidtchen A, Malmsten M. Antimicrobial peptides: key components of the innate immune system. Crit Rev Biotechnol 2011; 32:143-71. [PMID: 22074402 DOI: 10.3109/07388551.2011.594423] [Citation(s) in RCA: 545] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Life-threatening infectious diseases are on their way to cause a worldwide crisis, as treating them effectively is becoming increasingly difficult due to the emergence of antibiotic resistant strains. Antimicrobial peptides (AMPs) form an ancient type of innate immunity found universally in all living organisms, providing a principal first-line of defense against the invading pathogens. The unique diverse function and architecture of AMPs has attracted considerable attention by scientists, both in terms of understanding the basic biology of the innate immune system, and as a tool in the design of molecular templates for new anti-infective drugs. AMPs are gene-encoded short (<100 amino acids), amphipathic molecules with hydrophobic and cationic amino acids arranged spatially, which exhibit broad spectrum antimicrobial activity. AMPs have been the subject of natural evolution, as have the microbes, for hundreds of millions of years. Despite this long history of co-evolution, AMPs have not lost their ability to kill or inhibit the microbes totally, nor have the microbes learnt to avoid the lethal punch of AMPs. AMPs therefore have potential to provide an important breakthrough and form the basis for a new class of antibiotics. In this review, we would like to give an overview of cationic antimicrobial peptides, origin, structure, functions, and mode of action of AMPs, which are highly expressed and found in humans, as well as a brief discussion about widely abundant, well characterized AMPs in mammals, in addition to pharmaceutical aspects and the additional functions of AMPs.
Collapse
Affiliation(s)
- Mukesh Pasupuleti
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada.
| | | | | |
Collapse
|
23
|
Pixley RA, Espinola RG, Ghebrehiwet B, Joseph K, Kao A, Bdeir K, Cines DB, Colman RW. Interaction of high-molecular-weight kininogen with endothelial cell binding proteins suPAR, gC1qR and cytokeratin 1 determined by surface plasmon resonance (BiaCore). Thromb Haemost 2011; 105:1053-9. [PMID: 21544310 DOI: 10.1160/th10-09-0591] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 03/02/2011] [Indexed: 01/09/2023]
Abstract
The physiologic activation of the plasma kallikrein-kinin system requires the assembly of its constituents on a cell membrane. High- molecular-weight kininogen (HK) and cleaved HK (HKa) both interact with at least three endothelial cell binding proteins: urokinase plasminogen activator receptor (uPAR), globular C1q receptor (gC1qR,) and cytokeratin 1 (CK1). The affinity of HK and HKa for endothelial cells are KD=7-52 nM. The contribution of each protein is unknown. We examined the direct binding of HK and HKa to the soluble extracellular form of uPAR (suPAR), gC1qR and CK1 using surface plasmon resonance. Each binding protein linked to a CM-5 chip and the association, dissociation and KD (equilibrium constant) were measured. The interaction of HK and HKa with each binding protein was zinc-dependent. The affinity for HK and HKa was gC1qR>CK1>suPAR, indicating that gC1qR is dominant for binding. The affinity for HKa compared to HK was the same for gC1qR, 2.6-fold tighter for CK1 but 53-fold tighter for suPAR. Complex between binding proteins was only observed between gC1qR and CK1 indicating that a binary CK1-gC1qR complex can form independently of kininogen. Although suPAR has the weakest affinity of the three binding proteins, it is the only one that distinguished between HK and HKa. This finding indicates that uPAR may be a key membrane binding protein for differential binding and signalling between the cleaved and uncleaved forms of kininogen. The role of CK1 and gC1qR may be to initially bind HK to the membrane surface before productive cleavage to HKa.
Collapse
Affiliation(s)
- R A Pixley
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Umemura H, Togawa A, Sogawa K, Satoh M, Mogushi K, Nishimura M, Matsushita K, Tanaka H, Takizawa H, Kodera Y, Nomura F. Identification of a high molecular weight kininogen fragment as a marker for early gastric cancer by serum proteome analysis. J Gastroenterol 2011; 46:577-85. [PMID: 21298293 DOI: 10.1007/s00535-010-0369-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 12/23/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND Serum biomarkers currently available for gastric cancers are not sufficiently sensitive and specific. METHODS We used matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MS) to generate comparative peptide profiles of serum samples obtained from gastric cancer patients (n = 81) and age- and sex-matched healthy controls (n = 66). RESULTS Because of initial screening and further validation, we found that the intensities of a 2209 m/z MS peak were increased in the preoperative sera obtained from gastric cancer patients, and we identified this peak, a 2209 Da peptide, as a high molecular weight (HMW) kininogen fragment. Receiver operating characteristic analyses showed that the area under the curve (AUC) for the 2209 Da peptide (AUC = 0.715) was greater than those for conventional tumor markers (carcinoembryonic antigen AUC = 0.593, carbohydrate antigen 19-9 AUC = 0.527) used for the detection of stage I gastric cancers. Inverse correlations were observed between the levels of intact HMW kininogen and the 2209 Da peptide, suggesting that the upregulation of some protease activities is responsible for the overproduction of a kininogen fragment in gastric cancer patients. CONCLUSIONS Serum levels of the 2209 Da peptide identified in this study have a greater diagnostic ability than those of conventional tumor markers used for the early detection of gastric cancer.
Collapse
Affiliation(s)
- Hiroshi Umemura
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhang X, Song Y, Wu Y, Dong Y, Lai L, Zhang J, Lu B, Dai F, He L, Liu M, Yi Z. Indirubin inhibits tumor growth by antitumor angiogenesis via blocking VEGFR2-mediated JAK/STAT3 signaling in endothelial cell. Int J Cancer 2011; 129:2502-11. [DOI: 10.1002/ijc.25909] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Accepted: 12/08/2010] [Indexed: 11/08/2022]
|
26
|
Abstract
Tissue kallikrein cleaves kininogens to release kinins. Kinins mediate inflammation by activating constitutive bradykinin receptor-2 (BR2), which are rapidly desensitized, and induced by inflammatory cytokines bradykinin receptor-1 (BR1), resistant to desensitization. Intestinal tissue kallikrein (ITK) may hydrolyze growth factors and peptides, whereas kinins are responsible for capillary permeability, pain, synthesis of cytokines, and adhesion molecule-neutrophil cascade. Our and others results have demonstrated ITK in intestinal goblet cells and its release into interstitial space during inflammation. Kallistatin, an inhibitor of ITK, has been shown in epithelial and goblet cells, and was decreased in inflamed intestine as well as in plasma compared with noninflammatory controls. BR1 was upregulated in patients with inflammatory bowel disease (IBD), and it has expressed in an apical part of enterocytes in inflamed intestine, but in the basal part in normal intestine. ITK and BR1 were visualized in macrophages forming granuloma in Crohn's disease. In animal studies BR2 blockade decreased intestinal contraction, but had limited effect on inflammatory lesions. BR1 was found to be upregulated in animal inflamed intestine, in part dependent on tumor necrosis factor alpha (TNF-α). A selective BR1 receptor antagonist decreased morphological and biochemical features of experimental intestinal inflammation. Both BR1 and BR2 mediate epithelial ion transport that leads to secretory diarrhea. The upregulation of BR1 in inflamed intestine provides a structural basis for the kinins function, suggesting that a selective BR1 antagonist may have potential in therapeutic trial of IBD patients.
Collapse
Affiliation(s)
- Antoni Stadnicki
- Department of Basis Biomedical Sciences, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
27
|
Dai J, Zhu X, Yoder MC, Wu Y, Colman RW. Cleaved high-molecular-weight kininogen accelerates the onset of endothelial progenitor cell senescence by induction of reactive oxygen species. Arterioscler Thromb Vasc Biol 2011; 31:883-9. [PMID: 21252071 DOI: 10.1161/atvbaha.110.222430] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Cleaved high-molecular-weight kininogen (HKa), an activation product of the plasma kallikrein-kinin system, inhibits endothelial cell functions. We questioned whether HKa affects the function of endothelial progenitor cells (EPCs) and accelerates their senescence. METHODS AND RESULTS Treatment with HKa for 2 weeks markedly inhibited the formation of large colonies and proliferation of EPCs on collagen surfaces, whereas HKa did not affect collagen-mediated EPC adhesion and survival. Concomitantly, treated EPCs displayed flattened and giant cell morphological changes and formation of intracellular vacuoles. As determined by acidic β-galactosidase staining, HKa increased senescent EPCs by 2- and >3-fold after culture for 1 and 2 weeks, respectively. In addition, HKa suppressed the telomerase activity of EPCs. HKa concentration-dependently increased the generation of intracellular reactive oxygen species (ROS) and markedly upregulated p38 kinase phosphorylation and prosenescence molecule p16(INK4a) expression. SB203580, a p38 inhibitor, attenuated the level of HKa-enhanced p16(INK4a) expression. Either quenching of ROS or inhibition of p38 kinase prevented HKa-induced EPC senescence. CONCLUSIONS HKa accelerates the onset of EPC senescence by activating the ROS-p38 kinase-p16(INK4a) signaling cascade. This novel activity of HKa points out the likelihood of HKa serving as an endogenous inducer of EPC senescence.
Collapse
Affiliation(s)
- Jihong Dai
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, Suzhou, China
| | | | | | | | | |
Collapse
|
28
|
de Sousa M. An outsider's perspective—ecotaxis revisited: an integrative review of cancer environment, iron and immune system cells. Integr Biol (Camb) 2010; 3:343-9. [DOI: 10.1039/c0ib00116c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Maria de Sousa
- Iron Genes and the Immune System (IRIS) Lab, IBMC-Porto and ICBAS, University of Porto, 823 Campo Alegre, 4150 Porto, Portugal
| |
Collapse
|
29
|
Khan ST, Pixley RA, Liu Y, Bakdash N, Gordon B, Agelan A, Huang Y, Achary MP, Colman RW. Inhibition of metastasis of syngeneic murine melanoma in vivo and vasculogenesis in vitro by monoclonal antibody C11C1 targeted to domain 5 of high molecular weight kininogen. Cancer Immunol Immunother 2010; 59:1885-93. [PMID: 20811885 PMCID: PMC11030835 DOI: 10.1007/s00262-010-0915-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/23/2010] [Indexed: 01/24/2023]
Abstract
Metastasis of malignant tumors is a major cause of morbidity and mortality. Inhibition of tumor growth in distant organs is of clinical importance. We have demonstrated that C11C1, a murine monoclonal antibody to the light chain region of high molecular weight kininogen (HK), reduces growth of murine multiple myeloma in normal mice and human colon cancer in nude mice. C11C1 inhibits angiogenesis by reducing tumor microvascular density by blocking binding of HK to endothelial cells. We now evaluate the anti-metastatic effect of C11C1 on C57BL/6 mouse lung metastatic model using B16F10 melanoma cells. The tail veins of mice were injected with 0.5 × 10(6) cells of melanoma B16F10. One group received C11C1 and the other received saline (control) intraperitoneally. When mice were killed at 28 days, 6 of 10 control mice had detectable metastatic pulmonary nodules which stained positive with an antibody against S-100 protein, a tumor antigen present in malignant melanoma cells. In the C11C1 groups, none of the mice showed metastatic foci in their lungs. We showed that C11C1 inhibits endothelial cell tube formation in a 3-D collagen fibrinogen gel model by inhibiting the rate of cleavage of HK by plasma kallikrein without changing the binding affinity for HK. These studies demonstrate that a monoclonal antibody to HK has the potential to prevent metastasis with minimal side effects.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Cell Line, Tumor
- Female
- Humans
- Kininogen, High-Molecular-Weight/immunology
- Kininogen, High-Molecular-Weight/metabolism
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/prevention & control
- Protein Structure, Tertiary
Collapse
Affiliation(s)
- Sabina T. Khan
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Robin A. Pixley
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Yuchuan Liu
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Nadia Bakdash
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Brigitte Gordon
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Alexis Agelan
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| | - Yajue Huang
- Department of Pathology, Temple University School of Medicine, Philadelphia, PA 19140 USA
| | - Mohan P. Achary
- Department of Radiation Oncology, Temple University School of Medicine, Philadelphia, PA 19140 USA
| | - Robert W. Colman
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, OMS 418, Philadelphia, PA 19140 USA
| |
Collapse
|
30
|
Wang WJ. Acurhagin-C, an ECD disintegrin, inhibits integrin alphavbeta3-mediated human endothelial cell functions by inducing apoptosis via caspase-3 activation. Br J Pharmacol 2010; 160:1338-51. [PMID: 20590625 DOI: 10.1111/j.1476-5381.2010.00781.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Acurhagin, a member of versatile metalloproteinase disintegrins from Agkistrodon acutus venom, has been identified as a platelet aggregation inhibitor, previously. Here, acurhagin-C, the C-terminal Glu-Cys-Asp (ECD)-containing fragment of acurhagin, was evaluated for its biological activities and potential applications in anti-angiogenic therapy. EXPERIMENTAL APPROACH Human umbilical vein endothelial cells (HUVECs) were treated with acurhagin-C to assay effects on viability, apoptosis, adhesion, migration, invasion, proliferation and angiogenesis. The recognition site and signalling involved for the interactions of acurhagin-C with HUVEC were determined using flow cytometric, electrophoresis and immunoblotting analyses. KEY RESULTS Acurhagin-C decreased viability and induced apoptosis in HUVEC. It also dose-dependently inhibited HUVEC adhesion to immobilized extracellular matrices fibronectin, collagen I and vitronectin with respective IC(50) values of approximately 0.6, 0.3 and 0.1 microM. Acurhagin-C prevented migration and invasion of HUVEC through vitronectin- and Matrigel-coated barriers respectively. Furthermore, acurhagin-C attenuated fibroblast growth factor-2-primed angiogenesis both in vitro and in vivo, and specifically blocked the binding of anti-alphavbeta3 monoclonal antibody 23C6 to HUVEC in an ECD-dependent manner. However, purified alphavbeta3 also dose-dependently bound to immobilized acurhagin and acurhagin-C with a saturable pattern. Interference with integrin alphavbeta3-mediated functions and promotion of caspase-3 activation by acurhagin-C affected morphology of HUVEC and induced apoptosis. CONCLUSIONS AND IMPLICATIONS Acurhagin-C elicited endothelial anoikis via disruption of alphavbeta3/focal adhesion kinase/phosphatidylinositol 3-kinase/Akt survival cascade and subsequent initiation of the procaspase-3 apoptotic signalling pathway.
Collapse
Affiliation(s)
- Wen-Jeng Wang
- Department of Nutrition and Health Sciences, Chang-Gung Institute of Technology, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
31
|
Abstract
Hemostasis is a sensitive and tightly regulated process, involving vascular endothelium and blood cells, as well as factors of the coagulation and fibrinolytic cascades. In severe and invasive infectious diseases, the equilibrium between the procoagulant and anticoagulant status of the host may change dramatically and can induce life-threatening complications. A growing body of evidence suggests that the contact system, also known as the intrinsic pathway of coagulation or kallikrein/kinin system, participate in these processes. Contact activation leads to the release of the highly potent proinflammatory peptide bradykinin and initiates the intrinsic pathway of coagulation. Several studies have shown a systemic activation of the contact system in animal models of severe bacterial infections, and similar findings were also reported when monitoring patients suffering from sepsis, severe sepsis, or septic shock. Complications resulting from a systemic activation of the contact system are pathologically high levels of bradykinin, consumption of contact factors, and a subsequent induction of inflammatory reactions. These conditions may contribute to serious complications such as hypotension and vascular leakage. Here, we summarize the state of the art in this field of research with a focus on the contact system, and we also discuss a potential role for the contact system as a target for the development of novel antimicrobial strategies.
Collapse
Affiliation(s)
- Sonja Oehmcke
- Department of Clinical Sciences, Division of Infection Medicine, BMC, B14, Lund University, Tornavägen 10, SE-221 84 Lund, Sweden.
| | | |
Collapse
|
32
|
Guérout N, Derambure C, Drouot L, Bon-Mardion N, Duclos C, Boyer O, Marie JP. Comparative gene expression profiling of olfactory ensheathing cells from olfactory bulb and olfactory mucosa. Glia 2010; 58:1570-80. [DOI: 10.1002/glia.21030] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
33
|
Kininogens: More than cysteine protease inhibitors and kinin precursors. Biochimie 2010; 92:1568-79. [PMID: 20346387 DOI: 10.1016/j.biochi.2010.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 03/10/2010] [Indexed: 12/13/2022]
Abstract
Two kininogens are found in mammalian sera: HK (high molecular weight kininogen) and LK (low molecular weight kininogen) with the exception of the rat which encompasses a third kininogen, T-Kininogen (TK). Kininogens are multifunctional glycosylated molecules related to cystatins (clan IH, family I25). They harbor three cystatin domains but only two of them are tight-binding inhibitors of cysteine cathepsins. HK and LK, but not TK, are precursors of potent peptide hormones, the kinins, which are released proteolytically by tissue and plasma kallikreins. Besides these classical features novel functions of kininogens have been recently discovered; they are described in the second part of this review. HKa, which corresponds to the kinin-free two-chain HK and its isolated domain D5 (kininostatin), possesses angiostatic and pro-apoptotic properties, inhibits the proliferation of endothelial cells and participates in the regulation of angiogenesis. Moreover, some HK-derived peptides display potent and broad-spectrum microbicidal properties against both Gram-positive and Gram-negative bacteria, and thus may offer a promising alternative to conventional antibiotic therapy. Of seminal interest, a kininogen-derived peptide inhibits activation of the contact phase system of coagulation and protects mice with invasive Streptococcus pyogenes infection from pulmonary lesions. On the other hand, TK is a biomarker of aging at the end of lifespan of elderly rats. However, although TK has been initially identified as an acute phase reactant, and earlier known as alpha-l-acute phase globulin, the increase of TK in liver and plasma is not known to relate to any inflammatory event during the senescence process.
Collapse
|
34
|
The Chick Embryo Chorioallantoic Membrane as an In Vivo Assay to Study Antiangiogenesis. Pharmaceuticals (Basel) 2010; 3:482-513. [PMID: 27713265 PMCID: PMC4033966 DOI: 10.3390/ph3030482] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 01/29/2010] [Accepted: 03/02/2010] [Indexed: 12/15/2022] Open
Abstract
Antiangiogenesis, e.g., inhibition of blood vessel growth, is being investigated as a way to prevent the growth of tumors and other angiogenesis-dependent diseases. Pharmacological inhibition interferes with the angiogenic cascade or the immature neovasculature with synthetic or semi-synthetic substances, endogenous inhibitors or biological antagonists.The chick embryo chorioallantoic membrane (CAM) is an extraembryonic membrane, which serves as a gas exchange surface and its function is supported by a dense capillary network. Because its extensive vascularization and easy accessibility, CAM has been used to study morphofunctional aspects of the angiogenesis process in vivo and to study the efficacy and mechanism of action of pro- and anti-angiogenic molecules. The fields of application of CAM in the study of antiangiogenesis, including our personal experience, are illustrated in this review article.
Collapse
|
35
|
uPA/PAI-1 : un outil d’individualisation de la prise en charge des cancers du sein. Biologie, implications cliniques et méthodes de dosage. Bull Cancer 2010; 97:341-8. [DOI: 10.1684/bdc.2010.1047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
36
|
Wu Y, Dai J, Schmuckler NG, Bakdash N, Yoder MC, Overall CM, Colman RW. Cleaved high molecular weight kininogen inhibits tube formation of endothelial progenitor cells via suppression of matrix metalloproteinase 2. J Thromb Haemost 2010; 8:185-93. [PMID: 19874467 PMCID: PMC3142619 DOI: 10.1111/j.1538-7836.2009.03662.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Endothelial progenitor cells (EPCs) contribute to postnatal neovascularization, thus promoting wide interest in their therapeutic potential in vascular injury and prevention of their dysfunction in cardiovascular diseases. Cleaved high molecular weight kininogen (HKa), an activation product of the plasma kallikrein-kinin system (KKS), inhibits the functions of differentiated endothelial cells including in vitro and in vivo angiogenesis. In this study, our results provided the first evidence that HKa is able to target EPCs and inhibits their tube forming capacity. METHODS AND RESULTS We determined the effect of HKa on EPCs using a three-dimensional vasculogenesis assay. Upon stimulation with vascular endothelial growth factor (VEGF) alone, EPCs formed vacuoles and tubes, and differentiated into capillary-like networks. As detected by gelatinolytic activity assay, VEGF stimulated secretion and activation of matrix metallopeptidase 2 (MMP-2), but not MMP-9, in the conditioned medium of 3D culture of EPCs. Specific inhibition or gene ablation of MMP-2, but not MMP-9, blocked the vacuole and tube formation by EPCs. Thus, MMP-2 is selectively required for EPC vasculogenesis. In a concentration-dependent manner, HKa significantly inhibited tube formation by EPCs and the conversion of pro-MMP-2 to MMP-2. Moreover, HKa completely blocked the association between pro-MMP-2 and alphavbeta3 integrin, and its inhibition of MMP-2 activation was dependent on the presence of alphavbeta3 integrin. In a purified system, HKa did not directly inhibit MMP-2 activity. CONCLUSIONS HKa inhibits tube forming capacity of EPCs by suppression of MMP-2 activation, which may constitute a novel link between activation of the KKS and EPC dysfunction.
Collapse
Affiliation(s)
- Y Wu
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Extravasale Proteolyse: Funktion und Interaktion der Faktoren des fibrinolytischen Systems. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
38
|
Khan MM, Liu Y, Khan ME, Gilman ML, Khan ST, Bromberg M, Colman RW. Upregulation of tissue factor in monocytes by cleaved high molecular weight kininogen is dependent on TNF-alpha and IL-1beta. Am J Physiol Heart Circ Physiol 2009; 298:H652-8. [PMID: 19966052 DOI: 10.1152/ajpheart.00825.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease and arthritis are associated with contact activation that results in cleavage of kininogen to form high molecular weight kininogen (HKa) and bradykinin. We have previously demonstrated that HKa can stimulate inflammatory cytokine and chemokine secretion from human monocytes. We now show that HKa can upregulate tissue factor antigen and procoagulant activity on human monocytes as a function of time (1-4 h) and HKa concentration (75-900 nM). The amino acid sequence responsible to block HKa effects is G440-H455. The HKa receptor macrophage-1 (Mac-1; CD11b18) is the binding site as shown by inhibition by a monoclonal antibody to CD11b/18. Chemical inhibitors of JNK, ERK, and p38 signaling pathways block cell signaling, as does an inhibitor to the transcription factor NF-kappaB. A combination of monoclonal antibodies to TNF-alpha and IL-1beta but neither alone inhibited the HKa induction of tissue factor. These results suggest that HKa mimics LPS by triggering a paracrine pathway in monocytes that depends on TNF-alpha and IL-1beta. Antibodies to kininogen or peptidomimetics might be a useful and safe therapy in inflammatory diseases or sepsis involving cytokines.
Collapse
Affiliation(s)
- Mohammad M Khan
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Clapp C, Thebault S, Jeziorski MC, Martínez De La Escalera G. Peptide hormone regulation of angiogenesis. Physiol Rev 2009; 89:1177-215. [PMID: 19789380 DOI: 10.1152/physrev.00024.2009] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is now apparent that regulation of blood vessel growth contributes to the classical actions of hormones on development, growth, and reproduction. Endothelial cells are ideally positioned to respond to hormones, which act in concert with locally produced chemical mediators to regulate their growth, motility, function, and survival. Hormones affect angiogenesis either directly through actions on endothelial cells or indirectly by regulating proangiogenic factors like vascular endothelial growth factor. Importantly, the local microenvironment of endothelial cells can determine the outcome of hormone action on angiogenesis. Members of the growth hormone/prolactin/placental lactogen, the renin-angiotensin, and the kallikrein-kinin systems that exert stimulatory effects on angiogenesis can acquire antiangiogenic properties after undergoing proteolytic cleavage. In view of the opposing effects of hormonal fragments and precursor molecules, the regulation of the proteases responsible for specific protein cleavage represents an efficient mechanism for balancing angiogenesis. This review presents an overview of the actions on angiogenesis of the above-mentioned peptide hormonal families and addresses how specific proteolysis alters the final outcome of these actions in the context of health and disease.
Collapse
Affiliation(s)
- Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico.
| | | | | | | |
Collapse
|
40
|
Kuderer NM, Ortel TL, Francis CW. Impact of venous thromboembolism and anticoagulation on cancer and cancer survival. J Clin Oncol 2009; 27:4902-11. [PMID: 19738120 DOI: 10.1200/jco.2009.22.4584] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Changes in the hemostatic system and chronic hemostatic activation are frequently observed in patients with cancer, even in the absence of venous thromboembolism (VTE). VTE is a leading cause of death among patients with cancer and contributes to long-term mortality in patients with early as well as advanced-stage cancer. Mounting evidence suggests that components of the clotting cascade and associated vascular factors play an integral part in tumor progression, invasion, angiogenesis, and metastasis formation. Furthermore, there are intriguing in vitro and animal findings that anticoagulants, in particular the low molecular weight heparins (LMWHs), exert an antineoplastic effect through multiple mechanisms, including interference with tumor cell adhesion, invasion, metastasis formation, angiogenesis, and the immune system. Several relatively small randomized controlled clinical trials of anticoagulation as cancer therapy in patients without a VTE diagnosis have been completed. These comprise studies with LMWH, unfractionated heparin, and vitamin K antagonists, with overall encouraging but nonconclusive results and some limitations. Meta-analyses performed for the American Society of Clinical Oncology VTE Guidelines Committee and the Cochrane Collaboration suggest overall favorable effects of anticoagulation on survival of patients with cancer, mainly with LMWH. However, definitive clinical trials have been elusive and questions remain regarding the importance of tumor type and stage on treatment efficacy, the impact of fatal thromboembolic events, optimal anticoagulation therapy, and safety with differing chemotherapy regimens. Although the LMWHs and related agents hold promise for improving outcomes in patients with cancer, additional studies of their efficacy and safety in this setting are needed.
Collapse
Affiliation(s)
- Nicole M Kuderer
- Division of Hematology, Oncology and Cellular Therapy, Duke Comprehensive Cancer Center, Duke University Medical Center, DUMC 3841, Durham, NC 27710, USA.
| | | | | |
Collapse
|
41
|
Abstract
Derangement of the blood clotting system contributes strongly to multiple organ failure in severe sepsis. In this review, we examine two microbial modulators of the clotting system: polyphosphates and omptins. Polyphosphates are linear polymers of inorganic phosphate that are abundant in the acidocalcisomes of prokaryotes and unicellular organisms as well as in the dense granules of human platelets. Polyphosphates modulate haemostasis by: (1) triggering clotting via the contact pathway; (2) accelerating the activation of coagulation factor V (a key cofactor in blood clotting) and (3) causing fibrin to form clots whose fibrils are thicker and more resistant to fibrinolysis. While polyphosphates are found in all prokaryotes, omptins have a more limited distribution among certain Gram-negative species. Omptins are outer membrane aspartyl proteases which were recently found to proteolytically inactivate tissue factor pathway inhibitor (TFPI), the main inhibitor of the initiation phase of blood clotting. Omptin activity against TFPI requires lipopolysaccharide without O-antigen (rough LPS) such as is found on the surface of Yersinia pestis, the etiologic agent of plague. Interestingly, expression of Pla, the Yersinia pestis omptin, has a demonstrated virulence role in converting plasminogen into the fibrinolytic enzyme plasmin, which would seemingly antagonize any procoagulant effect of TFPI inactivation. However, since the rate of TFPI inactivation is much higher than the rate of plasminogen activation, we suggest that Pla may have a dual function in supporting the bubonic form of plague which is unique to Yersinia pestis.
Collapse
Affiliation(s)
- Thomas H Yun
- Biochemistry Department, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | |
Collapse
|
42
|
Schousboe I, Nystrøm B. High molecular weight kininogen binds to laminin--characterization and kinetic analysis. FEBS J 2009; 276:5228-38. [PMID: 19691495 DOI: 10.1111/j.1742-4658.2009.07218.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
High molecular weight kininogen (HK) is an abundant plasma protein that plays a central role for the function of the kallikrein/kinin/kininogen system. Thus, cleavage of HK by kallikrein liberates bradykinin, which stimulates vascular repair and a two-chain protein, activated HK (HKa), which induces apoptosis in proliferating endothelial cells. The localization of these events remains obscure, although the basement membrane may be of importance. Analyzing the interaction between HK and HKa and selected basement membrane proteins, we observed that they bound to the major noncollageneous proteins laminin, but not to vitronectin or fibronectin coated on microtiter plates. The binding to laminin was Zn2+ independent. However, at low but not at high concentrations of albumin, Zn2+ increased the affinity for the binding by abolishing an inhibitory effect of Ca2+. Recombinant human kininostatin encompassing the amino acid sequence, Arg439-Ser532 but not the endothelial cell binding peptide sequence (His479-His498; HKH20) within kininostatin inhibited the binding of HKa to laminin. This established that the amino acid sequence Arg439-Lys478 in domain 5 of HK is of importance for its binding to laminin. Extensive proteolytic cleavage of HK and HKa with kallikrein abolished the binding to laminin, releasing a 12 kDa anti-kininostatin reacting peptide. On the basis of these results, we propose that the binding of HK to laminin is a primary event, which secures proper localization of the cleavage products for subsequent interaction with the endothelium to promote inflammatory and pro- and anti-angiogenic activities.
Collapse
Affiliation(s)
- Inger Schousboe
- Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark.
| | | |
Collapse
|
43
|
Vosgerau U, Lauer D, Unger T, Kaschina E. Cleaved high molecular weight kininogen, a novel factor in the regulation of matrix metalloproteinases in vascular smooth muscle cells. Biochem Pharmacol 2009; 79:172-9. [PMID: 19682438 DOI: 10.1016/j.bcp.2009.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 08/03/2009] [Accepted: 08/05/2009] [Indexed: 01/09/2023]
Abstract
We previously reported that Brown Norway Katholiek rats, which feature a deficiency of plasma kininogens, develop severe abdominal aortic aneurysm. Increased activity of matrix metalloproteinases (MMPs) in the aortic wall, leading to degradation of extracellular matrix components, is considered to play a crucial role in aneurysm formation. Using an in vitro model of vascular smooth muscle cells (VSMCs), cultured from the rat aorta, we investigated whether the cleaved form of high molecular weight kininogen, designated HKa, affects the expression of MMP-9 and MMP-2 and their tissue inhibitors (TIMPs). Treatment of VSMCs with HKa reduced in a concentration-dependent manner IL-1alpha-induced release of MMP-9 and MMP-2, associated with decreased MMP enzymatic activity levels in conditioned media, as demonstrated by gelatin zymography and fluorescein-labeled gelatin substrate assay, respectively. Real-time PCR revealed that HKa reduced corresponding MMP-9 mRNA levels. Further investigations showed that this effect did not result from a modified rate of MMP-9 mRNA degradation. TIMP-1 mRNA levels, already increased as a result of cytokine-stimulation, were significantly enhanced by HKa. Furthermore, we found elevated basal mRNA expression levels of MMP-2 and TIMP-2 in VSMCs derived from kininogen-deficient Brown Norway Katholiek rats. These results demonstrate for the first time that HKa affects the regulation of MMPs in VSMCs.
Collapse
Affiliation(s)
- Uwe Vosgerau
- Center for Cardiovascular Research/Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | | | | | | |
Collapse
|
44
|
Blumenstein M, Prakash R, Cooper GJS, North RA. Aberrant processing of plasma vitronectin and high-molecular-weight kininogen precedes the onset of preeclampsia. Reprod Sci 2009; 16:1144-52. [PMID: 19657137 DOI: 10.1177/1933719109342756] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To date, there is no reliable test to identify women in early pregnancy at risk of developing preeclampsia. Difference gel electrophoresis (DIGE) identified the plasma proteins vitronectin (VN) and high-molecular-weight kininogen (HK) in association with preeclampsia. In a longitudinal proteomics study, the plasma of preeclamptic patients (n = 6) was compared to healthy control participants (n = 6) before the onset of preeclampsia (week 20) and at the time of presentation with clinical disease (weeks 33-36). The 75-kd single-chain VN molecule increased 1.6- to 1.9-fold in preeclampsia, whereas the 65-kd moiety of the 2-chain VN molecule decreased 1.5- to 1.7-fold compared to healthy controls (P < .05). Immunoblots revealed differences in proteolytic processing of VN and/or HK in women who develop preeclampsia or preeclampsia further complicated by small-for-gestational-age. Vitronectin and HK may prove to be useful as early markers of fibrinolytic activity and neutrophil activation, which are known to be associated with preeclampsia.
Collapse
Affiliation(s)
- Marion Blumenstein
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Ohta T, Ikemoto Y, Usami A, Koide T, Wakabayashi S. High affinity interaction between histidine-rich glycoprotein and the cell surface type ATP synthase on T-cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:1099-107. [DOI: 10.1016/j.bbamem.2009.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 02/19/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022]
|
47
|
Serum ferritin regulates blood vessel formation: a role beyond iron storage. Proc Natl Acad Sci U S A 2009; 106:1683-4. [PMID: 19193849 DOI: 10.1073/pnas.0813318106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
48
|
Abstract
Angiogenesis, the synthesis of new blood vessels from preexisting vessels, plays a critical role in normal wound healing and tumor growth. HKa (cleaved high molecular weight kininogen) is an endogenous inhibitor of angiogenesis formed by the cleavage of kininogen on endothelial cells. Ferritin is a protein principally known for its central role in iron storage. Here, we demonstrate that ferritin binds to HKa with high affinity (K(d) 13 nM). Further, ferritin antagonizes the antiangiogenic effects of HKa, enhancing the migration, assembly, and survival of HKa-treated endothelial cells. Effects of ferritin were independent of its iron content. Peptide mapping revealed that ferritin binds to a 22-aa subdomain of HKa that is critical to its antiangiogenic activity. In vivo, ferritin opposed HKa's antiangiogenic effects in a human prostate cancer xenograft, restoring tumor-dependent vessel growth. Ferritin-mediated regulation of angiogenesis represents a new angiogenic regulatory pathway, and identifies a new role for ferritin in cell biology.
Collapse
|
49
|
Clements JA, Willemsen NM, Myers SA, Dong Y. The Tissue Kallikrein Family of Serine Proteases: Functional Roles in Human Disease and Potential as Clinical Biomarkers. Crit Rev Clin Lab Sci 2008; 41:265-312. [PMID: 15307634 DOI: 10.1080/10408360490471931] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostate specific antigen (PSA) or human kallikrein 3 (hK3) has long been an effective biomarker for prostate cancer. Now, other members of the tissue kallikrein (KLK) gene family are fast becoming of clinical interest due to their potential as prognostic biomarkers. particularly for hormone dependent cancers. The tissue kallikreins are serine proteases that are encoded by highly conserved multi-gene family clusters in rodents and humans. The rat and mouse loci contain 10 and 25 functional genes, respectively, while the human locus at 19q 13.4 contains 15 genes. The structural organization and size of these genes are similar across species; all genes have 5 coding exons that encode a prepro-enzyme. Although the physiological activators of these zymogens have not been described, in vitro biochemical studies show that some kallikreins can auto-activate and others can activate each other, suggesting that the kallikreins may participate in an enzymatic cascade similar to that of the coagulation cascade. These genes are expressed, to varying degrees, in a wide range of tissues suggesting a functional involvement in a diverse range of physiological and pathophysiological processes. These include roles in normal skin desquamation and psoriatic lesions, tooth development, neural plasticity, and Alzheimer's disease (AD). Of particular interest is the expression of many kallikreins in prostate, ovarian, and breast cancers where they are emerging as useful prognostic indicators of disease progression.
Collapse
Affiliation(s)
- Judith A Clements
- Hormone Dependent Cancer Program, Cluster for Molecular Biotechnology, School of Life Sciences & Science Research Centre, Queensland University of Technology, Brisbane, Australia.
| | | | | | | |
Collapse
|
50
|
A systematic methodology for proteome-wide identification of peptides inhibiting the proliferation and migration of endothelial cells. Proc Natl Acad Sci U S A 2008; 105:13775-80. [PMID: 18780781 DOI: 10.1073/pnas.0803241105] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We introduce a systematic computational methodology based on bioinformatics that has enabled us to identify and classify >120 endogenous peptide inhibitors of endothelial cell proliferation and migration. These peptides are derived from members of the type IV collagen, thrombospondin, and CXC chemokine protein families, as well as somatotropin hormones, serpins, and various kringle-containing proteins. Their activity in suppressing the proliferation and migration of endothelial cells in vitro provides proof of principle for the validity of this computational method. Interestingly, some of the peptides are derived from proteins known to be proangiogenic. By performing receptor neutralization studies, we have identified receptors to which these peptides bind. On the basis of this receptor-binding information, we evaluated several examples of peptide-based combinatorial screening strategies. In some cases, this combinatorial screening identified strong synergism between peptides. The current work provides a guideline for a computational-based peptidomics approach for the discovery of endogenous bioactive peptides.
Collapse
|