1
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
2
|
Welch CL, Aldred MA, Balachandar S, Dooijes D, Eichstaedt CA, Gräf S, Houweling AC, Machado RD, Pandya D, Prapa M, Shaukat M, Southgate L, Tenorio-Castano J, Chung WK. Defining the clinical validity of genes reported to cause pulmonary arterial hypertension. Genet Med 2023; 25:100925. [PMID: 37422716 PMCID: PMC10766870 DOI: 10.1016/j.gim.2023.100925] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Srimmitha Balachandar
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- NIHR BioResource for Translational Research - Rare Diseases, Department of Haemotology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Divya Pandya
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Matina Prapa
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Memoona Shaukat
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Jair Tenorio-Castano
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IDiPAZ, Universidad Autonoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; ITHACA, European Reference Network, Brussels, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY; Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
3
|
Lekatz LA, Shukla P, Vasquez Hidalgo MA, O'Rourke S, Haring J, Dorsam GP, Grazul-Bilska AT, Vonnahme KA. Uterine kallikrein and arterial bradykinin activities and uterine arterial proliferation in response to acute estradiol-17β exposure in ovariectomized ewes. Domest Anim Endocrinol 2022; 81:106748. [PMID: 35842984 DOI: 10.1016/j.domaniend.2022.106748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Estradiol-17β (E2) increases kallikrein in rodent and human reproductive tissues. Kallikrein specific activity is increased in the porcine uterus when conceptus E2 is secreted at maternal recognition of pregnancy. When kallikrein acts on kininogen to liberate bradykinin, angiogenic and vasoactive factors are released. The uterus of ovariectomized ewes administered E2 undergoes rapid vascular changes via different patterns of angiogenic and vasoactive factors. Our hypothesis was that E2 would increase the specific activity and protein secretion of tissue kallikrein in endometrial explants culture media (ECM) and ewes exposed to E2 would have uterine arteries that would be more sensitive to the vasodilatory effects of bradykinin. Ovariectomized ewes received 100 mg of E2 implants for 0, 12, 24, or 48 h. After treatment, uterine weights were determined, and caruncles were processed for ECM. Uterine weights and uterine weight per ewe body weight were significantly greater in the 12 and 24 h ewes compared with the 0 h ewes, with the 48 h ewes being similar to the 24 h ewes. There were no statistically significant differences in caruncular tissue kallikrein protein secretion among the treatment groups. There was a tendency (P = 0.09) for duration of E2 exposure to influence tissue kallikrein specific activity where kallikrein activity was greater (P ≤ 0.05) in the 12 and 48 h ewes compared with the 0 h ewes, with 24 h ewes being intermediate (unprotected F test). Uterine arteries from ewes with E2 for 24 and 48 h had more sensitivity to bradykinin, via the bradykinin receptor 2, than uterine arteries from ewes with 0 or 12 h E2 exposure. We fail to reject our hypothesis as E2 did elicit a positive response in tissue kallikrein specific activity and bradykinin response. Further investigations are needed to determine how kallikrein and bradykinin may be involved in vascular remodeling of the ovine uterus.
Collapse
Affiliation(s)
- L A Lekatz
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - P Shukla
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - M A Vasquez Hidalgo
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - S O'Rourke
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - J Haring
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - G P Dorsam
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - A T Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - K A Vonnahme
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA.
| |
Collapse
|
4
|
Mohammedi K, Abouleka Y, Carpentier C, Potier L, Dubois S, Foussard N, Rigalleau V, Gautier JF, Gourdy P, Charpentier G, Roussel R, Scheen A, Bauduceau B, Hadjadj S, Alhenc-Gelas F, Marre M, Velho G. Association Between the ACE Insertion/Deletion Polymorphism and Risk of Lower-Limb Amputation in Patients With Long-Standing Type 1 Diabetes. Diabetes Care 2022; 45:407-415. [PMID: 34853028 DOI: 10.2337/dc21-0973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/01/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The ACE insertion/deletion (I/D) polymorphism has been widely studied in people with diabetes, albeit not with regard to lower-limb amputation (LLA). We examined associations among this polymorphism, plasma ACE concentration, and LLA in people with type 1 diabetes. RESEARCH DESIGN AND METHODS ACE I/D genotype and plasma ACE were assessed in three prospective cohorts of participants with type 1 diabetes. LLA was defined as minor (below-the-ankle amputation consisting of at least one ray metatarsal resection) or major (transtibial or transfemoral) amputation. Linear, logistic, and Cox regression models were computed to evaluate the likelihood of prevalent and incident LLA by ACE genotype (XD [ID or ID] vs. II) and plasma ACE, after adjusting for confounders. RESULTS Among 1,301 participants (male 54%, age 41 ± 13 years), 90 (6.9%) had a baseline history of LLA. Baseline LLA was more prevalent in XD (7.4%) than in II genotype (4.5%, odds ratio [OR] 2.17 [95 %CI 1.03-4.60]). Incident LLA occurred in 53 individuals during the 14-year follow-up and was higher in XD versus II carriers (hazard ratio 3.26 [95% CI 1.16-13.67]). This association was driven by excess risk of minor, but not major, LLA. The D allele was associated with increased prevalent LLA at the end of follow-up (OR 2.48 [1.33-4.65]). LLA was associated with higher mean (95% CI) ACE levels in II (449 [360, 539] vs. 354 [286, 423] ng/mL), but not XD (512 [454, 570] vs. 537 [488, 586]), carriers. CONCLUSIONS This report is the first of an independent association between ACE D allele and excess LLA risk, mainly minor amputations, in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Kamel Mohammedi
- Department of Endocrinology, Diabetes and Nutrition, Bordeaux University Hospital, Hôpital Haut-Lévêque, Pessac, France.,Faculty of Medicine, University of Bordeaux, Bordeaux, France.,Biology of Cardiovascular Diseases, INSERM U1034, Pessac, France
| | - Yawa Abouleka
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France.,Service d'Endocrinologie Diabétologie Nutrition, Hôpital Bichat, AP-HP, Paris, France
| | - Charlyne Carpentier
- Service d'Endocrinologie Diabétologie Nutrition, CHU d'Angers, Angers, France
| | - Louis Potier
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France.,Service d'Endocrinologie Diabétologie Nutrition, Hôpital Bichat, AP-HP, Paris, France
| | - Severine Dubois
- Service d'Endocrinologie Diabétologie Nutrition, CHU d'Angers, Angers, France
| | - Ninon Foussard
- Department of Endocrinology, Diabetes and Nutrition, Bordeaux University Hospital, Hôpital Haut-Lévêque, Pessac, France.,Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Vincent Rigalleau
- Department of Endocrinology, Diabetes and Nutrition, Bordeaux University Hospital, Hôpital Haut-Lévêque, Pessac, France.,Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Jean-François Gautier
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France.,Service de Diabétologie et d'Endocrinologie, Hôpital Lariboisière, AP-HP, Université de Paris, Paris, France
| | - Pierre Gourdy
- Service d'Endocrinologie Diabétologie Nutrition, CHU de Toulouse, Toulouse, France.,Institut des Maladies Métaboliques et Cardiovasculaires, UMR1297 INSERM/UPS, Université Toulouse 3, Toulouse, France
| | - Guillaume Charpentier
- 10Center for Study and Research for Improvement of the Treatment of Diabetes, Bioparc-Génopole Évry-Corbeil, Évry, France
| | - Ronan Roussel
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France.,Service d'Endocrinologie Diabétologie Nutrition, Hôpital Bichat, AP-HP, Paris, France
| | | | | | - Samy Hadjadj
- 13Institut du Thorax, INSERM, CNRS, Université de Nantes, CHU Nantes, Nantes, France
| | - François Alhenc-Gelas
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France
| | - Michel Marre
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France.,14Clinique Ambroise Paré, Neuilly-sur-Seine, France
| | - Gilberto Velho
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France
| |
Collapse
|
5
|
Kinins and Kinin Receptors in Cardiovascular and Renal Diseases. Pharmaceuticals (Basel) 2021; 14:ph14030240. [PMID: 33800422 PMCID: PMC8000381 DOI: 10.3390/ph14030240] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
This review addresses the physiological role of the kallikrein–kinin system in arteries, heart and kidney and the consequences of kallikrein and kinin actions in diseases affecting these organs, especially ischemic and diabetic diseases. Emphasis is put on pharmacological and genetic studies targeting kallikrein; ACE/kininase II; and the two kinin receptors, B1 (B1R) and B2 (B2R), distinguished through the work of Domenico Regoli and his collaborators. Potential therapeutic interest and limitations of the pharmacological manipulation of B1R or B2R activity in cardiovascular and renal diseases are discussed. This discussion addresses either the activation or inhibition of these receptors, based on recent clinical and experimental studies.
Collapse
|
6
|
Swietlik EM, Gräf S, Morrell NW. The role of genomics and genetics in pulmonary arterial hypertension. Glob Cardiol Sci Pract 2020; 2020:e202013. [PMID: 33150157 PMCID: PMC7590931 DOI: 10.21542/gcsp.2020.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom.,NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
7
|
Qian Y, Lyu Y, Jiang M, Tang B, Nie T, Lu S. Human urinary kallidinogenase or edaravone combined with butylphthalide in the treatment of acute ischemic stroke. Brain Behav 2019; 9:e01438. [PMID: 31638334 PMCID: PMC6908872 DOI: 10.1002/brb3.1438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022] Open
Abstract
AIM The effectiveness of neuroprotective agents is still unclear. Here we analyzed the clinical outcomes of acute ischemic stroke (AIS) patients treated with human urinary kallidinogenase (HUK) or edaravone (Eda) combined with butylphthalide (NBP). METHODS From January 2016 to December 2017, a total of 165 AIS patients were enrolled in this open-label, randomized controlled clinical study. Patients were randomly allocated into HUK group and Eda group in a ratio of 2:1. All the patients received basic treatments and NBP (200 mg p.o. qid) while HUK group received 0.15 PNA unit of HUK injection (ivgtt. qd) and Eda group received 30 mg Eda (ivgtt. bid) for 14 consecutive days. Independency rate [12-month modified Rankin Scale (mRS) score ≤ 1] and related factors were compared between the two groups. RESULTS Twelve-month mRS score of the HUK group (1, IQR 0~1) was significantly lower compared with Eda group (2, IQR 1~3, p < .0001). The HUK treatment achieved an independency rate of 79.1% while the Eda treatment only had 45.3% (p < .0001). Further binary logistic regression showed that recurrent stroke (RR: 0.1, 95% CI: 0.0~0.1, p = .038) and HUK treatment (RR: 4.2, 95% CI: 1.1~16.5, p = .041) could significantly affect patients' 12-month outcomes. CONCLUSION Human urinary kallidinogenase combined with NBP can enhance AIS patients' long-term independency rate, and the effectiveness of HUK combined therapy is better than Eda.
Collapse
Affiliation(s)
- Yun Qian
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Lyu
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Minhai Jiang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bo Tang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Nie
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Lu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Zhu N, Pauciulo MW, Welch CL, Lutz KA, Coleman AW, Gonzaga-Jauregui C, Wang J, Grimes JM, Martin LJ, He H, Shen Y, Chung WK, Nichols WC. Novel risk genes and mechanisms implicated by exome sequencing of 2572 individuals with pulmonary arterial hypertension. Genome Med 2019; 11:69. [PMID: 31727138 PMCID: PMC6857288 DOI: 10.1186/s13073-019-0685-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/06/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Group 1 pulmonary arterial hypertension (PAH) is a rare disease with high mortality despite recent therapeutic advances. Pathogenic remodeling of pulmonary arterioles leads to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. Mutations in bone morphogenetic protein receptor type 2 and other risk genes predispose to disease, but the vast majority of non-familial cases remain genetically undefined. METHODS To identify new risk genes, we performed exome sequencing in a large cohort from the National Biological Sample and Data Repository for PAH (PAH Biobank, n = 2572). We then carried out rare deleterious variant identification followed by case-control gene-based association analyses. To control for population structure, only unrelated European cases (n = 1832) and controls (n = 12,771) were used in association tests. Empirical p values were determined by permutation analyses, and the threshold for significance defined by Bonferroni's correction for multiple testing. RESULTS Tissue kallikrein 1 (KLK1) and gamma glutamyl carboxylase (GGCX) were identified as new candidate risk genes for idiopathic PAH (IPAH) with genome-wide significance. We note that variant carriers had later mean age of onset and relatively moderate disease phenotypes compared to bone morphogenetic receptor type 2 variant carriers. We also confirmed the genome-wide association of recently reported growth differentiation factor (GDF2) with IPAH and further implicate T-box 4 (TBX4) with child-onset PAH. CONCLUSIONS We report robust association of novel genes KLK1 and GGCX with IPAH, accounting for ~ 0.4% and 0.9% of PAH Biobank cases, respectively. Both genes play important roles in vascular hemodynamics and inflammation but have not been implicated in PAH previously. These data suggest new genes, pathogenic mechanisms, and therapeutic targets for this lethal vasculopathy.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | | | - Jiayao Wang
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Joseph M Grimes
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hua He
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA.
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
9
|
Alhenc-Gelas F, Bouby N, Girolami JP. Kallikrein/K1, Kinins, and ACE/Kininase II in Homeostasis and in Disease Insight From Human and Experimental Genetic Studies, Therapeutic Implication. Front Med (Lausanne) 2019; 6:136. [PMID: 31316987 PMCID: PMC6610447 DOI: 10.3389/fmed.2019.00136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/31/2019] [Indexed: 01/19/2023] Open
Abstract
Kallikrein-K1 is the main kinin-forming enzyme in organs in resting condition and in several pathological situations whereas angiotensin I-converting enzyme/kininase II (ACE) is the main kinin-inactivating enzyme in the circulation. Both ACE and K1 activity levels are genetic traits in man. Recent research based mainly on human genetic studies and study of genetically modified mice has documented the physiological role of K1 in the circulation, and also refined understanding of the role of ACE. Kallikrein-K1 is synthesized in arteries and involved in flow-induced vasodilatation. Endothelial ACE synthesis displays strong vessel and organ specificity modulating bioavailability of angiotensins and kinins locally. In pathological situations resulting from hemodynamic, ischemic, or metabolic insult to the cardiovascular system and the kidney K1 and kinins exert critical end-organ protective action and K1 deficiency results in severe worsening of the conditions, at least in the mouse. On the opposite, genetically high ACE level is associated with increased risk of developing ischemic and diabetic cardiac or renal diseases and worsened prognosis of these diseases. The association has been well-documented clinically while causality was established by ACE gene titration in mice. Studies suggest that reduced bioavailability of kinins is prominently involved in the detrimental effect of K1 deficiency or high ACE activity in diseases. Kinins are involved in the therapeutic effect of both ACE inhibitors and angiotensin II AT1 receptor blockers. Based on these findings, a new therapeutic hypothesis focused on selective pharmacological activation of kinin receptors has been launched. Proof of concept was obtained by using prototypic agonists in experimental ischemic and diabetic diseases in mice.
Collapse
Affiliation(s)
- Francois Alhenc-Gelas
- INSERM U1138-CRC, Paris, France.,CRC-INSERM U1138, Paris-Descartes University, Paris, France.,CRC-INSERM U1138, Sorbonne University, Paris, France
| | - Nadine Bouby
- INSERM U1138-CRC, Paris, France.,CRC-INSERM U1138, Paris-Descartes University, Paris, France.,CRC-INSERM U1138, Sorbonne University, Paris, France
| | | |
Collapse
|
10
|
Chen L, Lyu Y, Lin X, Sun M, Feng W, Tian X. Effect of HUK on the outcome of ruptured intracranial aneurysm. Brain Behav 2018; 8:e01060. [PMID: 30160055 PMCID: PMC6192394 DOI: 10.1002/brb3.1060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To evaluate the clinical efficacy of Human Urinary Kallidinogenase (HUK) on the outcome of patients with ruptured intracranial aneurysm. METHODS This was a prospective, open-label study. At the Department of Neurosurgery in our hospital, 127 patients were treated and operated due to ruptured intracranial aneurysm in the period 2015-2016. After surgery, all the patients received basic treatment and 70 patients received additional HUK treatment (HUK group) according to their willing. In detail, 0.15 PNA unit of HUK injection plus 100 ml saline in intravenous infusion was performed, with once a day for 14 consecutive days. The modified Rankin Scale (mRS) scores and favorable mRS rates (mRS 0-1) were analyzed 3-month after the treatment. RESULTS No difference was shown in the basic characteristics between the two groups (p > 0.05). Favorable mRS rate in the HUK group (71.43%) was significantly higher than that in control group (50.88%, p < 0.05). In addition, 3-month death rate was significantly lower in the HUK group. Delayed ischemic stroke rate was similar between the two groups. CONCLUSION HUK can reduce morbidity and mortality of patients with ruptured intracranial aneurysm after surgery.
Collapse
Affiliation(s)
- Luyue Chen
- Department of NeurosurgeryZhongshan Hospital Xiamen UnviersityXiamenChina
| | - Yi Lyu
- Department of Medical AffairsTechpool Bio‐pharma Co., Ltd.ShanghaiChina
| | - Xiaoning Lin
- Department of NeurosurgeryZhongshan Hospital Xiamen UnviersityXiamenChina
| | - Minghui Sun
- Department of Medical AffairsTechpool Bio‐pharma Co., Ltd.ShanghaiChina
| | - Wei Feng
- Department of NeurosurgeryZhongshan Hospital Xiamen UnviersityXiamenChina
| | - Xinhua Tian
- Department of NeurosurgeryZhongshan Hospital Xiamen UnviersityXiamenChina
| |
Collapse
|
11
|
Han D, Chen X, Li D, Liu S, Lyu Y, Feng J. Human Urinary Kallidinogenase decreases recurrence risk and promotes good recovery. Brain Behav 2018; 8:e01033. [PMID: 30030910 PMCID: PMC6085914 DOI: 10.1002/brb3.1033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/09/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To evaluate the efficacy of Human Urinary Kallidinogenase (HUK) for secondary stroke prevention in the treatment of acute ischemic stroke (AIS) patients. MATERIALS AND METHODS In this retrospective study, from October 2016 to June 2017, 300 consecutive AIS patients were registered in our database. Among them, 145 patients received HUK treatment (HUK group), and 155 patients received basic treatment (control group). Basic treatment was administrated on all patients. 0.15 PNA unit of HUK injection plus 100 ml saline in intravenous infusion was performed in the HUK group, with once a day for 14 consecutive days. The rate of recurrent stroke and modified Rankin Scale (mRS) scores in two groups were analyzed 12 months after the treatment. RESULTS No difference was found in the age, gender, comorbidities, smoking history, and NIHSS scores between two groups before treatment (p > 0.05). 12 months after treatment, 10 patients in the HUK group (10.3%) and 26 patients in the control group (16.8%) got stroke recurrence at 12 months (p = 0.009). The mRS score of HUK group was significantly lower than that in the control group (2.3 ± 1.2 vs. 3.5 ± 1.4, p = 0.011). CONCLUSION Human Urinary Kallidinogenase is able to reduce the risk of stroke recurrence and promote good recovery for AIS patients within 12 months.
Collapse
Affiliation(s)
- Dong Han
- Department of Neurology, The Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Dongmei Li
- Department of Medical Affairs, Techpool Bio-pharma Co., Ltd, Guangzhou, Guangdong, China
| | - Shuang Liu
- Department of Neurology, The Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yi Lyu
- Department of Medical Affairs, Techpool Bio-pharma Co., Ltd, Guangzhou, Guangdong, China
| | - Juan Feng
- Department of Neurology, The Affiliated Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
12
|
Igić R. Four decades of ocular renin-angiotensin and kallikrein-kinin systems (1977–2017). Exp Eye Res 2018; 166:74-83. [DOI: 10.1016/j.exer.2017.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/09/2017] [Accepted: 05/20/2017] [Indexed: 12/31/2022]
|
13
|
Measurement of Angiogenesis, Arteriolargenesis, and Lymphangiogenesis Phenotypes by Use of Two-Dimensional Mesenteric Angiogenesis Assay. Methods Mol Biol 2017. [PMID: 27172966 DOI: 10.1007/978-1-4939-3628-1_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Successful therapeutic angiogenesis requires an understanding of how the myriad interactions of growth factors released during angiogenesis combine to form a mature vascular bed. This requires a model in which multiple physiological and cell biological parameters can be identified. The adenoviral-mediated mesenteric angiogenesis assay as described here is ideal for that purpose. The clear, thin, and relatively avascular mesenteric panel can be used to measure increased vessel perfusion by intravital microscopy. In addition, high-powered microvessel analysis is carried out by immunostaining of features essential for the study of angiogenesis or lymphangiogenesis (including endothelium, pericyte, smooth muscle cell area, and proliferation), allowing functional data to be obtained in conjunction with high-power microvessel ultrastructural analysis. Therefore, the mesenteric angiogenesis model offers a robust system to analyze the morphological changes associated with angiogenesis, induced by different agents.
Collapse
|
14
|
Wu D, Lyu Y, Zhong P, Liu F, Liu X. Human Urinary kallidinogenase promotes good recovery in ischemic stroke patients with level 3 hypertension. Brain Behav 2017; 7:e00752. [PMID: 28828213 PMCID: PMC5561313 DOI: 10.1002/brb3.752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/05/2017] [Accepted: 05/15/2017] [Indexed: 01/08/2023] Open
Abstract
AIM To evaluate the clinical efficacy of Human Urinary kallidinogenase (HUK) in the treatment of acute ischemic stroke (AIS) patients with level 3 hypertension. METHODS In this retrospective study, from January 2015 to June 2016, 150 consecutive AIS patients were registered in our database. Among them, 47 with level 3 hypertension received either HUK treatment (HUK group, 22 cases) or basic treatment (control group, 25 cases). Basic treatment was administrated on all patients. 0.15 PNA unit of HUK injection plus 100 ml saline in intravenous infusion was performed in the HUK group, with once a day for 14 consecutive days. The modified Rankin Scale (mRS) scores in two groups were analyzed 3 months after the treatment. RESULTS No difference was found in the NIHSS scores, age, gender, and comorbidities between two groups before treatment (p > .05). While after treatment, 3-month mRS score was significantly lower in the HUK group (2.1 ± 1.4 vs. 3.1 ± 1.3, p = .012) and good recovery rate (3-month mRS score ≤2) in the HUK group was significantly higher than that in the control group (p < .05). CONCLUSION HUK is able to promote long-term recovery for AIS patients with level 3 hypertension remarkably.
Collapse
Affiliation(s)
- Danhong Wu
- Department of Neurology Shanghai Fifth People's Hospital Fudan University Shanghai China
| | - Yi Lyu
- Department of Medical Affairs Techpool Bio-pharma Co.; Ltd. Guangzhou China
| | - Ping Zhong
- Department of Neurology Shanghai TCM Integrated Hospital Affiliated Shanghai University of Chinese Medicine Shanghai China
| | - Fengdi Liu
- Department of Medical Affairs Techpool Bio-pharma Co.; Ltd. Guangzhou China
| | - Xueyuan Liu
- Department of Neurology Shanghai Tenth People's Hospital of Tongji University Shanghai China
| |
Collapse
|
15
|
Desposito D, Waeckel L, Potier L, Richer C, Roussel R, Bouby N, Alhenc-Gelas F. Kallikrein(K1)-kinin-kininase (ACE) and end-organ damage in ischemia and diabetes: therapeutic implications. Biol Chem 2017; 397:1217-1222. [PMID: 27622831 DOI: 10.1515/hsz-2016-0228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/19/2016] [Indexed: 11/15/2022]
Abstract
Genetic and pharmacological studies, clinical and experimental, focused on kallikrein-K1, kinin receptors and ACE/kininase II suggest that kinin release in the settings of ischemia or diabetes reduces organ damage, especially in the heart and kidney. Kinin bioavailability may be a limiting factor for efficacy of current kinin-potentiating drugs, like ACE inhibitors. Primary activation of kinin receptors by prototypic pharmacological agonists, peptidase-resistant, selective B1 or B2, displays therapeutic efficacy in experimental cardiac and peripheral ischemic and diabetic diseases. B1R agonism was especially efficient in diabetic animals and had no unwanted effects. Clinical development of kinin receptor agonists may be warranted.
Collapse
|
16
|
Yang X, Orgah J, Wang D, Fan G, Jingyang H, Han J, Qin G, Gao X, Zhu Y. Danhong injection reduces vascular remodeling and up-regulates the Kallikrein-kinin system in spontaneously hypertensive rats. Sci Rep 2017; 7:4308. [PMID: 28655904 PMCID: PMC5487322 DOI: 10.1038/s41598-017-04661-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/17/2017] [Indexed: 12/16/2022] Open
Abstract
Although Danhong injection (DHI) is one of the most prescribed cardiovascular medicines in China, its therapeutic indications and mechanisms remain partially defined. We now identify molecular targets of DHI in resistance vasculatures and demonstrate its role in vascular function and blood pressure (BP) regulation. BP was determined in DHI, Losartan, and placebo- treated Spontaneously Hypertensive Rats (SHR) by both noninvasive and invasive measurements. Vasorelaxation was examined both in conduit and resistance vasculature by ex vivo aortic rings. Microarray analysis was performed and gene expression changes were verified by RT-qPCR and ELISA. Diastolic, systolic and mean BPs were significantly lower in DHI-treated SHR than controls by both tail-cuff and invasive BP measurements. In ex vivo rings, aortic and mesenteric vessels from SHR treated with DHI exhibited significantly greater acetylcholine-mediated relaxation. Among the 282 genes that are differentially expressed in microarray analysis, DHI treatment up-regulated the expression of kallikrein and plasma kallikrein B genes. DHI also significantly increased serum kallikrein content in SHR. Treatment with DHI significantly increased the ratio of aortic lumen to outer diameter. Therefore, the reduction of vascular remodeling and the up-regulation of Kallikrein-kinin system contribute, at least in part, to the antihypertensive effect of DHI in SHR.
Collapse
Affiliation(s)
- Xiaohu Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China.,Department of Pharmacy, Zhejiang Hospital, 12 Lingyin Road, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
| | - John Orgah
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China
| | - Dandan Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China
| | - Guanwei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China
| | - Hu Jingyang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China
| | - Jihong Han
- College of Life Sciences, Nankai University, Tianjin, 300193, P. R. China
| | - Gangjian Qin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China.,Feinberg Cardiovascular Research Institute, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin, 300193, P. R. China. .,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, P. R. China. .,Molecular Cardiology Research Institute, Tufts Medical Center, 750 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
17
|
Stone OA, Carter JG, Lin PC, Paleolog E, Machado MJC, Bates DO. Differential regulation of blood flow-induced neovascularization and mural cell recruitment by vascular endothelial growth factor and angiopoietin signalling. J Physiol 2017; 595:1575-1591. [PMID: 27868196 PMCID: PMC5330904 DOI: 10.1113/jp273430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/15/2016] [Indexed: 12/24/2022] Open
Abstract
KEY POINTS Combining nitric oxide (NO)-mediated increased blood flow with angiopoietin-1-Tie2 receptor signalling induces arteriolargenesis - the formation of arterioles from capillaries - in a model of physiological angiogenesis. This NO-Tie-mediated arteriolargenesis requires endogenous vascular endothelial growth factor (VEGF) signalling. Inhibition of VEGF signalling increases pericyte coverage in microvessels. Together these findings indicate that generation of functional neovasculature requires close titration of NO-Tie2 signalling and localized VEGF induction, suggesting that the use of exogenous VEGF expression as a therapeutic for neovascularization may not be successful. ABSTRACT Signalling through vascular endothelial growth factor (VEGF) receptors and the tyrosine kinase with IgG and EGF domains-2 (Tie2) receptor by angiopoietins is required in combination with blood flow for the formation of a functional vascular network. We tested the hypothesis that VEGF and angiopoietin-1 (Ang1) contribute differentially to neovascularization induced by nitric oxide (NO)-mediated vasodilatation, by comparing the phenotype of new microvessels in the mesentery during induction of vascular remodelling by over-expression of endothelial nitric oxide synthase in the fat pad of the adult rat mesentery during inhibition of angiopoietin signalling with soluble Tie2 (sTie2) and VEGF signalling with soluble Fms-like tyrosine kinase receptor-1 (sFlt1). We found that NO-mediated angiogenesis was blocked by inhibition of VEGF with sFlt1 (from 881 ± 98% increase in functional vessel area to 279 ± 72%) and by inhibition of angiopoietin with sTie2 (to 337 ± 67%). Exogenous angiopoietin-1 was required to induce arteriolargenesis (8.6 ± 1.3% of vessels with recruitment of vascular smooth muscle cells; VSMCs) in the presence of enhanced flow. sTie2 and sFlt1 both inhibited VSMC recruitment (both 0%), and VEGF inhibition increased pericyte recruitment to newly formed vessels (from 27 ± 2 to 54 ± 3% pericyte ensheathment). We demonstrate that a fine balance of VEGF and angiopoietin signalling is required for the formation of a functional vascular network. Endogenous VEGF signalling prevents excess neovessel pericyte coverage, and is required for VSMC recruitment during increased nitric oxide-mediated vasodilatation and angiopoietin signalling (NO-Tie-mediated arteriogenesis). Therapeutic vascular remodelling paradigms may therefore require treatments that modulate blood flow to utilize endogenous VEGF, in combination with exogenous Ang1, for effective neovascularization.
Collapse
Affiliation(s)
- Oliver A Stone
- Microvascular Research Laboratories, Bristol Heart Institute, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - James G Carter
- Microvascular Research Laboratories, Bristol Heart Institute, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - P Charles Lin
- Center for Cancer Research, National Institute of Cancer, Frederick, MD, 2170, USA
| | - Ewa Paleolog
- Kennedy Institute of Rheumatology, University of Oxford, 65 Aspenlea Road, Hammersmith, London, UK
| | - Maria J C Machado
- Microvascular Research Laboratories, Bristol Heart Institute, School of Physiology and Pharmacology, University of Bristol, Bristol, UK.,Cancer Biology, Division of Oncology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - David O Bates
- Microvascular Research Laboratories, Bristol Heart Institute, School of Physiology and Pharmacology, University of Bristol, Bristol, UK.,Cancer Biology, Division of Oncology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
18
|
Guihard PJ, Yao J, Blazquez-Medela AM, Iruela-Arispe L, Boström KI, Yao Y. Endothelial-Mesenchymal Transition in Vascular Calcification of Ins2Akita/+ Mice. PLoS One 2016; 11:e0167936. [PMID: 27936229 PMCID: PMC5148029 DOI: 10.1371/journal.pone.0167936] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
Endothelial-mesenchymal transition (EndMT) drives endothelium to contribute to normal development and disease processes. Here, we report that EndMTs occur in the diabetic endothelium of Ins2Akita/wt mouse, and show that induction of sex determining region Y-box 2 (Sox2) is a mediator of excess BMP signaling that results in activation of EndMTs and increased vascular calcification. We also find an induction of a complex of serine proteases in the diabetic endothelium, required for the up-regulation of Sox2. Our results suggest that EndMTs contribute to vascular calcification in diabetic arteries.
Collapse
Affiliation(s)
- Pierre J. Guihard
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ana M. Blazquez-Medela
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Luisa Iruela-Arispe
- The Molecular Biology Institute at UCLA, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California, United States of America
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- The Molecular Biology Institute at UCLA, Los Angeles, California, United States of America
- * E-mail: (YY); (KB)
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California, United States of America
- * E-mail: (YY); (KB)
| |
Collapse
|
19
|
Bolinger MT, Antonetti DA. Moving Past Anti-VEGF: Novel Therapies for Treating Diabetic Retinopathy. Int J Mol Sci 2016; 17:E1498. [PMID: 27618014 PMCID: PMC5037775 DOI: 10.3390/ijms17091498] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 12/25/2022] Open
Abstract
Diabetic retinopathy is the leading cause of blindness in working age adults, and is projected to be a significant future health concern due to the rising incidence of diabetes. The recent advent of anti-vascular endothelial growth factor (VEGF) antibodies has revolutionized the treatment of diabetic retinopathy but a significant subset of patients fail to respond to treatment. Accumulating evidence indicates that inflammatory cytokines and chemokines other than VEGF may contribute to the disease process. The current review examines the presence of non-VEGF cytokines in the eyes of patients with diabetic retinopathy and highlights mechanistic pathways in relevant animal models. Finally, novel drug targets including components of the kinin-kallikrein system and emerging treatments such as anti-HPTP (human protein tyrosine phosphatase) β antibodies are discussed. Recognition of non-VEGF contributions to disease pathogenesis may lead to novel therapeutics to enhance existing treatments for patients who do not respond to anti-VEGF therapies.
Collapse
Affiliation(s)
- Mark T Bolinger
- Departments of Ophthalmology and Visual Sciences, Kellogg Eye Center, and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| | - David A Antonetti
- Departments of Ophthalmology and Visual Sciences, Kellogg Eye Center, and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
20
|
Kallikrein in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Filippou PS, Karagiannis GS, Musrap N, Diamandis EP. Kallikrein-related peptidases (KLKs) and the hallmarks of cancer. Crit Rev Clin Lab Sci 2016; 53:277-91. [PMID: 26886390 DOI: 10.3109/10408363.2016.1154643] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kallikrein-related peptidases (KLKs) represent the largest family of serine proteases within the human genome and are expressed in various tissues. Although they regulate several important physiological functions, KLKs have also been implicated in numerous pathophysiological processes, including cancer. Growing evidence describing the deregulation of KLK expression and secretion, as well as activation in various malignancies, has uncovered their potential as mediators of cancer progression, biomarkers of disease and as candidate therapeutic targets. The diversity of signalling pathways and proteolytic cascades involving KLKs and their downstream targets appears to affect cancer biology through multiple mechanisms, including those related to the hallmarks of cancer. The aim of this review is to provide an update on the importance of KLK-driven molecular pathways in relation to cancer cell traits associated with the hallmarks of cancer and to highlight their potential in personalized therapeutics.
Collapse
Affiliation(s)
- Panagiota S Filippou
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - George S Karagiannis
- b Department of Anatomy & Structural Biology , Albert Einstein College of Medicine, Yeshiva University Bronx , New York , NY , USA
| | - Natasha Musrap
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - Eleftherios P Diamandis
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada .,c Department of Clinical Biochemistry , University Health Network , Toronto , ON , Canada , and.,d Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
22
|
Involvement of Kallikrein-Related Peptidases in Normal and Pathologic Processes. DISEASE MARKERS 2015; 2015:946572. [PMID: 26783378 PMCID: PMC4689925 DOI: 10.1155/2015/946572] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
Human kallikrein-related peptidases (KLKs) are a subgroup of serine proteases that participate in proteolytic pathways and control protein levels in normal physiology as well as in several pathological conditions. Their complex network of stimulatory and inhibitory interactions may induce inflammatory and immune responses and contribute to the neoplastic phenotype through the regulation of several cellular processes, such as proliferation, survival, migration, and invasion. This family of proteases, which includes one of the most useful cancer biomarkers, kallikrein-related peptidase 3 or PSA, also has a protective effect against cancer promoting apoptosis or counteracting angiogenesis and cell proliferation. Therefore, they represent attractive therapeutic targets and may have important applications in clinical oncology. Despite being intensively studied, many gaps in our knowledge on several molecular aspects of KLK functions still exist. This review aims to summarize recent data on their involvement in different processes related to health and disease, in particular those directly or indirectly linked to the neoplastic process.
Collapse
|
23
|
Murakami T. Kallikrein-Kinin System: An Emerging Competitor or Collaborator for VEGF in Diabetic Macular Edema? Diabetes 2015; 64:3350-2. [PMID: 26405275 DOI: 10.2337/db15-0746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Tomoaki Murakami
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Abbas OL, Borman H, Terzi YK, Terzi A, Bayraktar N, Yazıcı AC. The Notch pathway is a critical regulator of angiogenesis in a skin model of ischemia. Vasc Med 2015; 20:205-11. [PMID: 25834117 DOI: 10.1177/1358863x15570723] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The Notch pathway is definitely required for normal vascular development. Although the contribution of Notch in postnatal angiogenesis is the focus of intense investigation, the implication of Notch in reparative neovascularization in the skin remains unexplored. In this study, we investigated Notch changes using a skin model of ischemia. Thirty Sprague-Dawley rats were divided into two groups. In the surgery group (n = 24), a caudally based dorsal skin flap was raised and sutured back into its initial position. In the control group, no surgical procedure was performed. Tissue biopsies were obtained at different time intervals. Tissue specimens were assessed for Delta-like ligand 4 (DLL4) and vascular endothelial growth factor (VEGF) gene expression by real-time polymerase chain reaction (PCR). Immunohistochemical staining was used for detection of DLL4 in tissue materials. Quantitative assessment of skin flap microvasculature was made. Compared with normoperfused tissue, VEGF and DLL4 expressions increased significantly (p < 0.01). Immunohistochemical analysis revealed weak and patchy expression of DLL4 in microvascular endothelial cells of normoperfused tissues. Conversely, DLL4 expression was upregulated in capillary endothelial cells after ischemia. In conclusion, in this study we have shown that the Notch ligand DLL4 is upregulated in skin tissue after ischemia. A deeper understanding of these fundamental principles will aid in the development of new avenues for the treatment of blood vessel-related skin pathologies.
Collapse
Affiliation(s)
- Ozan L Abbas
- Department of Plastic and Reconstructive Surgery, Ahi Evran University, Kirsehir, Turkey
| | - Hüseyin Borman
- Department of Plastic and Reconstructive Surgery, Başkent University, Ankara, Turkey
| | - Yunus K Terzi
- Department of Medical Genetics, Başkent University, Ankara, Turkey
| | - Ayşen Terzi
- Department of Pathology, Başkent University, Ankara, Turkey
| | | | - Ayşe C Yazıcı
- Department of Biostatistics, Başkent University, Ankara, Turkey
| |
Collapse
|
25
|
Desposito D, Potier L, Chollet C, Gobeil F, Roussel R, Alhenc-Gelas F, Bouby N, Waeckel L. Kinin receptor agonism restores hindlimb postischemic neovascularization capacity in diabetic mice. J Pharmacol Exp Ther 2014; 352:218-26. [PMID: 25398240 DOI: 10.1124/jpet.114.219196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Limb ischemia is a major complication of thromboembolic diseases. Diabetes worsens prognosis by impairing neovascularization. Genetic or pharmacological inactivation of the kallikrein-kinin system aggravates limb ischemia in nondiabetic animals, whereas angiotensin I-converting enzyme/kininase II inhibition improves outcome. The role of kinins in limb ischemia in the setting of diabetes is not documented. We assessed whether selective activation of kinin receptors by pharmacological agonists can influence neovascularization in diabetic mice with limb ischemia and have a therapeutic effect. Selective pseudopeptide kinin B1 or B2 receptor agonists resistant to peptidase action were administered by osmotic minipumps at a nonhypotensive dosage for 14 days after unilateral femoral artery ligation in mice previously rendered diabetic by streptozotocin. Comparison was made with ligatured, nonagonist-treated nondiabetic and diabetic mice. Diabetes reduced neovascularization, assessed by microangiography and histologic capillary density analysis, by roughly 40%. B1 receptor agonist or B2 receptor agonist similarly restored neovascularization in diabetic mice. Neovascularization in agonist-treated diabetic mice was indistinguishable from nondiabetic mice. Both treatments restored blood flow in the ischemic hindfoot, measured by laser-Doppler perfusion imaging. Macrophage infiltration increased 3-fold in the ischemic gastrocnemius muscle during B1 receptor agonist or B2 receptor agonist treatment, and vascular endothelial growth factor (VEGF) level increased 2-fold. Both treatments increased, by 50-100%, circulating CD45/CD11b-positive monocytes and CD34(+)/VEGFR2(+) progenitor cells. Thus, selective pharmacological activation of B1 or B2 kinin receptor overcomes the effect of diabetes on postischemic neovascularization and restores tissue perfusion through monocyte/macrophage mobilization. Kinin receptors are potential therapeutic targets in limb ischemia in diabetes.
Collapse
Affiliation(s)
- Dorinne Desposito
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Louis Potier
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Catherine Chollet
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Fernand Gobeil
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ronan Roussel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Francois Alhenc-Gelas
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Nadine Bouby
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ludovic Waeckel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| |
Collapse
|
26
|
Naidu N, Botha JH, Naidoo S. B1 but not B2 bradykinin receptor agonists promote DU145 prostate cancer cell proliferation and migration. Afr Health Sci 2014; 14:657-62. [PMID: 25352885 DOI: 10.4314/ahs.v14i3.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The kallikrein-kinin system (KKS) is an endogenous pathway involved in angiogenesis and tumourigenesis, both vital for cancer growth and progression. OBJECTIVES To investigate the effect of two bradykinin receptor (B1R and B2R) agonists on growth and motility of prostate tumour (DU145) and micro-vascular endothelial cells (dMVECs). METHODS Increasing concentrations of selective B1R and B2R agonists were added to cultured cells. Cell proliferation and migration were assessed using the 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) and modified Boyden Chamber assays, respectively. Where significant stimulation was found, the influence of an antagonist was also investigated. RESULTS Neither growth nor motility of endothelial cells was affected by either agonist. In DU145 cells, while the B2R agonist was without any significant effect, the B1R agonist stimulated proliferation and migration at concentrations of 10nM and 50nM respectively. Further, this effect was abrogated when cells were pre-incubated with a B1R antagonist. CONCLUSIONS Unlike the physiologically-active B2R, the pathologically-inducible B1R may be implicated in prostate tumourigenic events. The involvement of the KKS in malignant prostate pathology supports on-going exploration of bradykinin receptor antagonists as target candidates in the development of alternate approaches to cancer therapy.
Collapse
Affiliation(s)
- N Naidu
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - J H Botha
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - S Naidoo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| |
Collapse
|
27
|
Ahimastos AA, Latouche C, Natoli AK, Reddy-luthmoodoo M, Golledge J, Kingwell BA. Potential Vascular Mechanisms of Ramipril Induced Increases in Walking Ability in Patients With Intermittent Claudication. Circ Res 2014; 114:1144-55. [DOI: 10.1161/circresaha.114.302420] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
We recently reported that ramipril more than doubled maximum walking times in patients with peripheral artery disease with intermittent claudication.
Objective:
Our aim was to conduct exploratory analyses of the effects of ramipril therapy on circulating biomarkers of angiogenesis/arteriogenesis, thrombosis, inflammation, and leukocyte adhesion in patients with intermittent claudication.
Methods and Results:
One hundred sixty-five patients with intermittent claudication (mean, 65.3 [SD, 6.7] years) were administered ramipril 10 mg per day (n=82) or matching placebo (n=83) for 24 weeks in a randomized, double-blind study. Plasma biomarkers of angiogenesis/arteriogenesis (vascular endothelial growth factor-A, fibroblast growth factor-2), thrombosis (D-dimer, von Willebrand factor, thrombin-antithrombin III), inflammation (high-sensitivity C-reactive protein, osteopontin), and leukocyte adhesion (soluble vascular cell adhesion molecule-1, soluble intracellular adhesion molecule-1) were measured at baseline and 24 weeks. Relative to placebo, ramipril was associated with increases in vascular endothelial growth factor-A by 38% (95% confidence interval [CI], 34%–42%) and fibroblast growth factor-2 by 64% (95% CI, 44–85%;
P
<0.001 for both), and reductions in D-dimer by 24% (95% CI, −30% to −18%), von Willebrand factor by 22% (95% CI, −35% to −9%), thrombin-antithrombin III by 16% (95% CI, −19% to −13%), high-sensitivity C-reactive protein by 13% (95% CI, −14% to −9%), osteopontin by 12% (95% CI, −14% to −10%), soluble vascular cell adhesion molecule-1 by 14% (95% CI, −18% to −10%), and soluble intracellular adhesion molecule-1 by 15% (95% CI, −17% to −13%; all
P
<0.001). With the exception of von Willebrand factor, all the above changes correlated significantly with the change in maximum walking time (
P
=0.02−0.001) in the group treated with ramipril.
Conclusions:
Ramipril is associated with an increase in the biomarkers of angiogenesis/arteriogenesis and reduction in the markers of thrombosis, inflammation, and leukocyte adhesion. This study informs strategies to improve mobility in patients with intermittent claudication.
Clinical Trial Registration Information:
URL:
http://clinicaltrials.gov
. Unique identifier: NCT00681226.
Collapse
Affiliation(s)
- Anna A. Ahimastos
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Celine Latouche
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Alaina K. Natoli
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Medini Reddy-luthmoodoo
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Jonathan Golledge
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Bronwyn A. Kingwell
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
28
|
Bhat M, Pouliot M, Couture R, Vaucher E. The kallikrein-kinin system in diabetic retinopathy. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:111-43. [PMID: 25130041 DOI: 10.1007/978-3-319-06683-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic retinopathy (DR) is a major microvascular complication associated with type 1 and type 2 diabetes mellitus, which can lead to visual impairment and blindness. Current treatment strategies for DR are mostly limited to laser therapies, steroids, and anti-VEGF agents, which are often associated with unwanted side effects leading to further complications. Recent evidence suggests that kinins play a primary role in the development of DR through enhanced vascular permeability, leukocytes infiltration, and other inflammatory mechanisms. These deleterious effects are mediated by kinin B1 and B2 receptors, which are expressed in diabetic human and rodent retina. Importantly, kinin B1 receptor is virtually absent in sane tissue, yet it is induced and upregulated in diabetic retina. These peptides belong to the kallikrein-kinin system (KKS), which contains two separate and independent pathways of regulated serine proteases, namely plasma kallikrein (PK) and tissue kallikrein (TK) that are involved in the biosynthesis of bradykinin (BK) and kallidin (Lys-BK), respectively. Hence, ocular inhibition of kallikreins or antagonism of kinin receptors offers new therapeutic avenues in the treatment and management of DR. Herein, we present an overview of the principal features and known inflammatory mechanisms associated with DR along with the current therapeutic approaches and put special emphasis on the KKS as a new and promising therapeutic target due to its link with key pathways directly associated with the development of DR.
Collapse
|
29
|
Girolami JP, Blaes N, Bouby N, Alhenc-Gelas F. Genetic manipulation and genetic variation of the kallikrein-kinin system: impact on cardiovascular and renal diseases. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:145-196. [PMID: 25130042 DOI: 10.1007/978-3-319-06683-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Genetic manipulation of the kallikrein-kinin system (KKS) in mice, with either gain or loss of function, and study of human genetic variability in KKS components which has been well documented at the phenotypic and genomic level, have allowed recognizing the physiological role of KKS in health and in disease. This role has been especially documented in the cardiovascular system and the kidney. Kinins are produced at slow rate in most organs in resting condition and/or inactivated quickly. Yet the KKS is involved in arterial function and in renal tubular function. In several pathological situations, kinin production increases, kinin receptor synthesis is upregulated, and kinins play an important role, whether beneficial or detrimental, in disease outcome. In the setting of ischemic, diabetic or hemodynamic aggression, kinin release by tissue kallikrein protects against organ damage, through B2 and/or B1 bradykinin receptor activation, depending on organ and disease. This has been well documented for the ischemic or diabetic heart, kidney and skeletal muscle, where KKS activity reduces oxidative stress, limits necrosis or fibrosis and promotes angiogenesis. On the other hand, in some pathological situations where plasma prekallikrein is inappropriately activated, excess kinin release in local or systemic circulation is detrimental, through oedema or hypotension. Putative therapeutic application of these clinical and experimental findings through current pharmacological development is discussed in the chapter.
Collapse
|
30
|
Fu SS, Li FJ, Wang YY, You AB, Qie YL, Meng X, Li JR, Li BC, Zhang Y, Da Li Q. Kallikrein gene-modified EPCs induce angiogenesis in rats with ischemic hindlimb and correlate with integrin αvβ3 expression. PLoS One 2013; 8:e73035. [PMID: 24019890 PMCID: PMC3760867 DOI: 10.1371/journal.pone.0073035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 07/16/2013] [Indexed: 12/29/2022] Open
Abstract
Background Human tissue kallikrein (hTK) plays an essential role in the physiological and pathological mechanisms of blood vessels. This study aimed to determine whether angiogenesis induced by endothelial progenitor cells (EPCs) transduced with the adenovirus-mediated hTK gene could improve blood flow in rat hindlimb ischemia in vivo and to establish a promising mechanism in vitro. Methods EPCs transduced with adenovirus encoding hTK-162 (i.e., Ad/hTK-transduced EPCs or Ad/GFP-transduced EPCs) were administered to Wister rats with hindlimb ischemia through therapeutic neovascularization. Muscular capillary density (MCD), blood flow (BF), and the number of myofibers were measured at days 7, 14, and 21 after treatment. Expressions of integrin αvβ3 and endothelial nitric oxide synthase (eNOS) were detected on the surface of EPCs. Results MCD, BF, and the number of myofibers in rats with Ad/hTK-transduced EPCs remarkably increased at day 21 after treatment compared with rats with Ad/GFP-transduced EPCs or the control group (P<0.01). Expressions of integrin αvβ3 and eNOS protein on the surface of EPCs also increased in rats with Ad/hTK-transduced EPCs. The levels of integrin αvβ3 expression were reduced by PI3K and eNOS blockade, and the inhibitor of integrin αvβ3 abrogated the migration and adhesion of hTK-transduced EPCs (P<0.05). Conclusion hTK gene delivery in vivo improves the natural angiogenic response to ischemia. The ability of hTK gene-transduced EPCs can be enhanced in vitro, in which integrin αvβ3 plays a role in the process.
Collapse
Affiliation(s)
- Shen Shen Fu
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Fu Ji Li
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yuan Yuan Wang
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - An Bei You
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yi Liang Qie
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiao Meng
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Jian Rui Li
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bao Chuan Li
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yun Zhang
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qing Da Li
- Department of Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
- * E-mail:
| |
Collapse
|
31
|
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
|
32
|
van den Hengel LG, Hellingman AA, Nossent AY, van Oeveren-Rietdijk AM, de Vries MR, Spek CA, van Zonneveld AJ, Reitsma PH, Hamming JF, de Boer HC, Versteeg HH, Quax PHA. Protease-activated receptor (PAR)2, but not PAR1, is involved in collateral formation and anti-inflammatory monocyte polarization in a mouse hind limb ischemia model. PLoS One 2013; 8:e61923. [PMID: 23637930 PMCID: PMC3630144 DOI: 10.1371/journal.pone.0061923] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 03/14/2013] [Indexed: 12/14/2022] Open
Abstract
AIMS In collateral development (i.e. arteriogenesis), mononuclear cells are important and exist as a heterogeneous population consisting of pro-inflammatory and anti-inflammatory/repair-associated cells. Protease-activated receptor (PAR)1 and PAR2 are G-protein-coupled receptors that are both expressed by mononuclear cells and are involved in pro-inflammatory reactions, while PAR2 also plays a role in repair-associated responses. Here, we investigated the physiological role of PAR1 and PAR2 in arteriogenesis in a murine hind limb ischemia model. METHODS AND RESULTS PAR1-deficient (PAR1-/-), PAR2-deficient (PAR2-/-) and wild-type (WT) mice underwent femoral artery ligation. Laser Doppler measurements revealed reduced post-ischemic blood flow recovery in PAR2-/- hind limbs when compared to WT, while PAR1-/- mice were not affected. Upon ischemia, reduced numbers of smooth muscle actin (SMA)-positive collaterals and CD31-positive capillaries were found in PAR2-/- mice when compared to WT mice, whereas these parameters in PAR1-/- mice did not differ from WT mice. The pool of circulating repair-associated (Ly6C-low) monocytes and the number of repair-associated (CD206-positive) macrophages surrounding collaterals in the hind limbs were increased in WT and PAR1-/- mice, but unaffected in PAR2-/- mice. The number of repair-associated macrophages in PAR2-/- hind limbs correlated with CD11b- and CD115-expression on the circulating monocytes in these animals, suggesting that monocyte extravasation and M-CSF-dependent differentiation into repair-associated cells are hampered. CONCLUSION PAR2, but not PAR1, is involved in arteriogenesis and promotes the repair-associated response in ischemic tissues. Therefore, PAR2 potentially forms a new pro-arteriogenic target in coronary artery disease (CAD) patients.
Collapse
Affiliation(s)
- Lisa G. van den Hengel
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Alwine A. Hellingman
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne Yael Nossent
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemarie M. van Oeveren-Rietdijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet R. de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - C. Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter H. Reitsma
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap F. Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hetty C. de Boer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H. Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | - Paul H. A. Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
33
|
Luo Y, Kumar P, Mendelson CR. Estrogen-related receptor γ (ERRγ) regulates oxygen-dependent expression of voltage-gated potassium (K+) channels and tissue kallikrein during human trophoblast differentiation. Mol Endocrinol 2013; 27:940-52. [PMID: 23584901 DOI: 10.1210/me.2013-1038] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Estrogen-related receptor γ (ERRγ) serves a critical O2-dependent regulatory role in the differentiation of human cytotrophoblasts to syncytiotrophoblast. In this study, we investigated expression of genes encoding tissue kallikrein (KLK1) and voltage-gated K(+) channels (KV7) during differentiation of human trophoblasts in culture and the roles of ERRγ and O2 tension in their regulation. Expression of KLK1 and the KV7 channel subunits, KCNQ1, KCNE1, KCNE3, and KCNE5, increased during differentiation of cultured human trophoblast cells in a 20% O2 environment. Notably, together with ERRγ, expression of KLK1, KCNQ1, KCNE1, KCNE3, and KCNE5 was markedly reduced when cells were cultured in a hypoxic environment (2% O2). Moreover, upon transduction of trophoblast cells with short hairpin RNAs for endogenous ERRγ, KLK1, KCNQ1, KCNE1, and KCNE3 expression was significantly decreased. Promoter and site-directed mutagenesis studies in transfected cells identified putative ERRγ response elements within the KLK1 and KCNE1 5'-flanking regions required for ERRγ-stimulated transcriptional activity. Binding of endogenous ERRγ to these ERRγ response elements increased during trophoblast differentiation in culture and was inhibited by hypoxia. The KV7 blocker linopirdine reduced human chorionic gonadotropin secretion and aggregation of cultured human trophoblasts, suggesting a possible role of KV7 channels in cell fusion and differentiation. Illumina gene expression arrays of cultured human trophoblast cells revealed several genes upregulated during syncytiotrophoblast differentiation and downregulated upon ERRγ knockdown involved in cell differentiation, adhesion, and synthesis of steroid and peptide hormones required for placental development and function. Collectively, these findings suggest that ERRγ mediates O2-dependent expression of genes involved in human trophoblast differentiation, function, and vascular homeostasis.
Collapse
Affiliation(s)
- Yanmin Luo
- Department of Biochemistry, North Texas March of Dimes Birth Defects Center, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | | | | |
Collapse
|
34
|
Waeckel L, Potier L, Richer C, Roussel R, Bouby N, Alhenc-Gelas F. Pathophysiology of genetic deficiency in tissue kallikrein activity in mouse and man. Thromb Haemost 2013; 110:476-83. [PMID: 23572029 DOI: 10.1160/th12-12-0937] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/14/2013] [Indexed: 12/30/2022]
Abstract
Study of mice rendered deficient in tissue kallikrein (TK) by gene inactivation and human subjects partially deficient in TK activity as consequence of an active site mutation has allowed recognising the physiological role of TK and its peptide products kinins in arterial function and in vasodilatation, in both species. TK appears as the major kinin forming enzyme in arteries, heart and kidney. Non-kinin mediated actions of TK may occur in epithelial cells in the renal tubule. In basal condition, TK deficiency induces mild defective phenotypes in the cardiovascular system and the kidney. However, in pathological situations where TK synthesis is typically increased and kinins are produced, TK deficiency has major, deleterious consequences. This has been well documented experimentally for cardiac ischaemia, diabetes renal disease, peripheral ischaemia and aldosterone-salt induced hypertension. These conditions are all aggravated by TK deficiency. The beneficial effect of ACE/kininase II inhibitors or angiotensin II AT1 receptor antagonists in cardiac ischaemia is abolished in TK-deficient mice, suggesting a prominent role for TK and kinins in the cardioprotective action of these drugs. Based on findings made in TK-deficient mice and additional evidence obtained by pharmacological or genetic inactivation of kinin receptors, development of novel therapeutic approaches relying on kinin receptor agonism may be warranted.
Collapse
Affiliation(s)
- L Waeckel
- Francois Alhenc-Gelas, INSERM U872, Centre de Recherche des Cordeliers, 15 rue de l'Ecole de Médecine 75006 Paris, France, E-mail:
| | | | | | | | | | | |
Collapse
|
35
|
Unique expression patterns associated with preferential recruitment of immature myeloid cells into angiogenic versus dormant tumors. Genes Immun 2013; 14:90-8. [PMID: 23303247 DOI: 10.1038/gene.2012.59] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cancer progression from microscopic dormant tumors into disseminated disease involves tumor angiogenesis, and is commonly referred to as the 'angiogenic switch'. CD11b(+)Gr1(+) immature myeloid cells (IMCs) were reported to promote angiogenesis and tumor progression. Here, we studied a model of tumor dormancy, in which Lewis Lung Carcinoma tumor cells were inoculated intra-abdominally into C57Bl/6J mice. Dormancy versus expansive growth was determined by the site of tumor implantation (lower vs upper abdomen). Global gene expression of IMCs was evaluated in different stages of recruitment, starting in the bone marrow, followed by the peripheral blood and finally in the vascular versus dormant tumors. We first demonstrated a ∼3 fold enrichment of IMCs within vascular tumors as compared with dormant tumors, correlating with tumor-infiltrating CD31(+) endothelial cells. Although their migration from the PB into dormant tumors led to differential expression of a relatively small number of genes, recruitment of IMCs into the upper tumors was associated with a profound transcriptional response. Importantly, a large set of proangiogenic genes were significantly upregulated in IMCs derived from vascular tumors compared with those derived from dormant tumors. We therefore, suggest that proangiogenic versus nonangiogenic transcriptional patterns is associated with the ability of IMCs to promote tumor angiogenesis.
Collapse
|
36
|
Early detection biomarkers for ovarian cancer. JOURNAL OF ONCOLOGY 2012; 2012:709049. [PMID: 23319948 PMCID: PMC3540796 DOI: 10.1155/2012/709049] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 11/19/2012] [Indexed: 12/12/2022]
Abstract
Despite the widespread use of conventional and contemporary methods to detect ovarian cancer development, ovarian cancer remains a common and commonly fatal gynecological malignancy. The identification and validation of early detection biomarkers highly specific to ovarian cancer, which would permit development of minimally invasive screening methods for detecting early onset of the disease, are urgently needed. Current practices for early detection of ovarian cancer include transvaginal ultrasonography, biomarker analysis, or a combination of both. In this paper we review recent research on novel and robust biomarkers for early detection of ovarian cancer and provide specific details on their contributions to tumorigenesis. Promising biomarkers for early detection of ovarian cancer include KLK6/7, GSTT1, PRSS8, FOLR1, ALDH1, and miRNAs.
Collapse
|
37
|
Effects of a novel bradykinin B1 receptor antagonist and angiotensin II receptor blockade on experimental myocardial infarction in rats. PLoS One 2012; 7:e51151. [PMID: 23236443 PMCID: PMC3517424 DOI: 10.1371/journal.pone.0051151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/29/2012] [Indexed: 01/06/2023] Open
Abstract
Background The aim of the present study was to evaluate the cardiovascular effects of the novel bradykinin B1 receptor antagonist BI-113823 following myocardial infarction (MI) and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin II type 1 (AT1) receptor antagonist after MI in rats. Methodology/Principal Findings Sprague Dawley rats were subjected to permanent occlusion of the left descending coronary artery. Cardiovascular function was determined at 7 days post MI. Treatment with either B1 receptor antagonist (BI-113823) or AT1 receptor antagonist (irbesartan) alone or in combination improved post-MI cardiac function as evidenced by attenuation of elevated left ventricular end diastolic pressure (LVEDP); greater first derivative of left ventricular pressure (± dp/dt max), left ventricle ejection fraction, fractional shorting, and better wall motion; as we as reductions in post-MI up-regulation of matrix metalloproteinases 2 (MMP-2) and collagen III. In addition, the cardiac up-regulation of B1 receptor and AT1 receptor mRNA were markedly reduced in animals treated with BI 113823, although bradykinin B2 receptor and angiotensin 1 converting enzyme (ACE1) mRNA expression were not significantly affected by B1 receptor blockade. Conclusions/Significance The present study demonstrates that treatment with the novel B1 receptor antagonist, BI-113823 improves post-MI cardiac function and does not influence the cardiovascular effects of AT1 receptor antagonist following MI.
Collapse
|
38
|
Yao Y, Sheng Z, Li Y, Yan F, Fu C, Li Y, Ma G, Liu N, Chao J, Chao L. Tissue kallikrein promotes cardiac neovascularization by enhancing endothelial progenitor cell functional capacity. Hum Gene Ther 2012; 23:859-70. [PMID: 22435954 DOI: 10.1089/hum.2011.123] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue kallikrein (TK) has been demonstrated to improve neovasculogenesis after myocardial infarction (MI). In the present study, we examined the role and underlying mechanisms of TK in peripheral endothelial progenitor cell (EPC) function. Peripheral blood-derived mononuclear cells containing EPCs were isolated from rat. The in vitro effects of TK on EPC differentiation, apoptosis, migration, and vascular tube formation capacity were studied in the presence or absence of TK, kinin B(2) receptor antagonist (icatibant), and phosphatidylinositol-3 kinase inhibitor (LY294002). Apoptosis was evaluated by flow-cytometry analysis using Annexin V-FITC/PI staining, as well as western-blot analysis of Akt phosphorylation and cleaved caspase-3. Using an MI mouse model, we then examined the in vivo effects of human TK gene adenoviral vector (Ad.hTK) administration on the number of CD34(+)Flk-1(+) progenitors in the peripheral circulation, heart tissue, extent of vasculogenesis, and heart function. Administration of TK significantly increased the number of Dil-LDL/UEA-lectin double-positive early EPCs, as well as their migration and tube formation properties in vitro. Transduction of TK in cultured EPCs attenuated apoptosis induced by hypoxia and led to an increase in Akt phosphorylation and a decrease in cleaved caspase-3 levels. The beneficial effects of TK were blocked by pretreatment with icatibant and LY294002. The expression of recombinant human TK in the ischemic mouse heart significantly improved cardiac contractility and reduced infarct size 7 days after gene delivery. Compared with the Ad.Null group, Ad.hTK reduced mortality and preserved left ventricular function by increasing the number of CD34(+)Flk-1(+) EPCs and promoting the growth of capillaries and arterioles in the peri-infarct myocardium. These data provide direct evidence that TK promotes vessel growth by increasing the number of EPCs and enhancing their functional properties through the kinin B(2) receptor-Akt signaling pathway.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Julian Ilcheff Borissoff
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute of Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | |
Collapse
|
40
|
Wang YD, Lu RY, Huang XX, Yuan F, Hu T, Peng Y, Huang SQ. Human tissue kallikrein promoted activation of the ipsilesional sensorimotor cortex after acute cerebral infarction. Eur Neurol 2011; 65:208-14. [PMID: 21422759 DOI: 10.1159/000325735] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 02/15/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Kallikrein, a serine proteinase, has been reported to have many functions, such as selectively dilating arterioles in the ischemic area and enhancing angiogenesis and neurogenesis. Therefore, it may promote cerebral poststroke reorganization. We observed the effect of human tissue kallikrein on the brain motor activation of acute ischemic stroke patients and evaluated patient condition severity and prognosis. METHODS Forty-four cases suffering from cerebral infarction between 6 and 72 h of onset were randomly assigned into the kallikrein group (n = 24) and the control group (n = 20). The control group was given conventional treatment, whereas the kallikrein group was given both conventional treatment and human tissue kallikrein over the course of 12-14 days. The activation of the sensorimotor cortex (SMC) and cerebellum, the affected forefinger strength and the NIHSS scores were evaluated before and after treatment. The MBI and MRS scores were assessed at 30 and 90 days after stroke onset. RESULTS There were no differences between the two groups in activation volume, patient condition and scores before treatment. After treatment, the ipsilesional SMC activation volume was significantly larger and the increase in the volume was significantly greater in the kallikrein group than in the control group (p < 0.05 for both). The NIHSS score was significantly smaller and the improvement in the score was significantly greater in the kallikrein group after treatment (p < 0.05 for both). Moreover, the MBI scores at 30 days were significantly higher, whereas the MRS scores at 30 days were significantly lower in the kallikrein group than in the control group (p < 0.05 for both). CONCLUSIONS Kallikrein improved neural function effectively and quickly after stroke, and promoting cerebral reorganization might be an important mechanism for kallikrein in the treatment of acute cerebral infarction.
Collapse
Affiliation(s)
- Yi-Dong Wang
- Department of Neurology, Sun Yat-Sen Hospital of Sun Yat-Sen University, Guangzhou, PR China
| | | | | | | | | | | | | |
Collapse
|
41
|
Valdés G, Corthorn J. Review: The angiogenic and vasodilatory utero-placental network. Placenta 2011; 32 Suppl 2:S170-5. [DOI: 10.1016/j.placenta.2011.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/31/2010] [Accepted: 01/11/2011] [Indexed: 01/23/2023]
|
42
|
Spinetti G, Fortunato O, Cordella D, Portararo P, Kränkel N, Katare R, Sala-Newby GB, Richer C, Vincent MP, Alhenc-Gelas F, Tonolo G, Cherchi S, Emanueli C, Madeddu P. Tissue kallikrein is essential for invasive capacity of circulating proangiogenic cells. Circ Res 2010; 108:284-93. [PMID: 21164105 DOI: 10.1161/circresaha.110.236786] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
RATIONALE Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.
Collapse
Affiliation(s)
- Gaia Spinetti
- Chair of Experimental Cardiovascular Medicine, University of Bristol, Bristol BS28HW, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
BACKGROUND Human tissue kallikrein (hK1) generates vasodilator kinins from kininogen and promotes angiogenesis by kinin-dependent and kinin-independent mechanisms. Here, we investigate the expression and functional relevance of hK1 in human gastrointestinal stromal tumour (GIST). METHODS Vascularisation and hK1 expression of GIST samples were assessed by immunohistochemistry. In two GIST cell lines, hK1 expression was assessed by PCR, and hK1 protein levels and activity were measured by ELISA and an amidolytic assay, respectively. The effect of hK1 silencing, inhibition or overexpression on GIST cell proliferation, migration and paracrine induction of angiogenesis was studied. Finally, local and systemic levels of hK1 were assessed in mice injected with GIST cells. RESULTS Human tissue kallikrein was detected in 19 out of 22 human GIST samples. Moreover, GIST cells express and secrete active hK1. Titration of hK1 demonstrated its involvement in GIST invasive behaviour, but not proliferation. Furthermore, hK1 released by GIST cells promoted endothelial cell migration and network formation through kinin-dependent mechanisms. Gastrointestinal stromal tumour implantation in nude mice resulted in local and systemic hK1 expression proportional to tumour dimension. CONCLUSIONS Human tissue kallikrein is produced and released by GIST and participates in tumour invasion. Further studies are needed to validate hK1 as a diagnostic biomarker and therapeutic target in GIST.
Collapse
|
44
|
Savvatis K, Westermann D, Schultheiss HP, Tschöpe C. Kinins in cardiac inflammation and regeneration: insights from ischemic and diabetic cardiomyopathy. Neuropeptides 2010; 44:119-25. [PMID: 20036002 DOI: 10.1016/j.npep.2009.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 11/05/2009] [Accepted: 11/12/2009] [Indexed: 11/24/2022]
Abstract
The kallikrein-kinin system (KKS) is a system of vasoactive peptides, the kinins, involved in different aspects of remodeling, inflammation and angiogenesis. Kinins mediate their actions through two receptors, B1R and B2R. It is increasingly recognized that the KKS is involved in the inflammatory processes of the heart. Evidence shows that the B2R is beneficial in myocardial diseases, protecting from inflammation, fibrosis and apoptosis, while B1R shows a proinflammatory character contributing to the disease progression by increasing the production of cytokines and stimulating the migration of immune cells. Furthermore, novel important actions of the KKS and its receptors contribute to neovascularization and recruitment of endothelial progenitor cells in ischemic areas and endothelial dysfunction. The kinin receptors could therefore constitute potential therapeutic targets in the treatment of myocardial ischemia and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Konstantinos Savvatis
- Charité - Universitätsmedizin Berlin, Department of Cardiology and Pneumonology, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | | | | | | |
Collapse
|
45
|
Cristofaro B, Stone OA, Caporali A, Dawbarn D, Ieronimakis N, Reyes M, Madeddu P, Bates DO, Emanueli C. Neurotrophin-3 is a novel angiogenic factor capable of therapeutic neovascularization in a mouse model of limb ischemia. Arterioscler Thromb Vasc Biol 2010; 30:1143-50. [PMID: 20360537 DOI: 10.1161/atvbaha.109.205468] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To investigate the novel hypothesis that neurotrophin-3 (NT-3), an established neurotrophic factor that participates in embryonic heart development, promotes blood vessel growth. METHODS AND RESULTS We evaluated the proangiogenic capacity of recombinant NT-3 in vitro and of NT-3 gene transfer in vivo (rat mesenteric angiogenesis assay and mouse normoperfused adductor muscle). Then, we studied whether either transgenic or endogenous NT-3 mediates postischemic neovascularization in a mouse model of limb ischemia. In vitro, NT-3 stimulated endothelial cell survival, proliferation, migration, and network formation on the basement membrane matrix Matrigel. In the mesenteric assay, NT-3 increased the number and size of functional vessels, including vessels covered with mural cells. Consistently, NT-3 overexpression increased muscular capillary and arteriolar densities in either the absence or the presence of ischemia and improved postischemic blood flow recovery in mouse hind limbs. NT-3-induced microvascular responses were accompanied by tropomyosin receptor kinase C (an NT-3 high-affinity receptor) phosphorylation and involved the phosphatidylinositol 3-kinase-Akt kinase-endothelial nitric oxide synthase pathway. Finally, endogenous NT-3 was shown to be essential in native postischemic neovascularization, as demonstrated by using a soluble tropomyosin receptor kinase C receptor domain that neutralizes NT-3. CONCLUSIONS Our results provide the first insight into the proangiogenic capacity of NT-3 and propose NT-3 as a novel potential agent for the treatment of ischemic disease.
Collapse
Affiliation(s)
- Brunella Cristofaro
- Experimental Cardiovascular Medicine Division, University of Bristol, Bristol, England, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Siragusa M, Katare R, Meloni M, Damilano F, Hirsch E, Emanueli C, Madeddu P. Involvement of phosphoinositide 3-kinase gamma in angiogenesis and healing of experimental myocardial infarction in mice. Circ Res 2010; 106:757-68. [PMID: 20056919 DOI: 10.1161/circresaha.109.207449] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Phosphoinositide 3-kinase (PI3K)gamma is expressed in hematopoietic cells, endothelial cells (ECs), and cardiomyocytes and regulates different cellular functions relevant to inflammation, tissue remodeling and cicatrization. Recently, PI3Kgamma inhibitors have been indicated for the treatment of chronic inflammatory/autoimmune diseases and atherosclerosis. OBJECTIVE We aimed to determine PI3Kgamma contribution to the angiogenic capacity of ECs and the effect of PI3Kgamma inhibition on healing of myocardial infarction (MI). METHODS AND RESULTS Human umbilical ECs were treated with a selective PI3Kgamma inhibitor, AS605240, or a pan-phosphoinositide 3-kinases inhibitor, LY294002. Both inhibitory treatments and small interfering RNA-mediated PI3Kgamma knockdown strongly impaired ECs angiogenic capacity, because of suppression of the PI3K/Akt and mitogen-activated protein kinase pathways. Constitutive activation of Akt rescued the angiogenic defect. Reparative angiogenesis was studied in vivo in a model of MI. AS605240 did not affect MI-induced PI3Kgamma upregulation, whereas it suppressed Akt activation and downstream signaling. AS605240 strongly reduced inflammation, enhanced cardiomyocyte apoptosis, and impaired survival and proliferation of ECs in peri-infarct zone, which resulted in defective reparative neovascularization. As a consequence, AS605240-treated MI hearts showed increased infarct size and impaired recovery of left ventricular function. Similarly, PI3Kgamma-deficient mice showed impaired reparative neovascularization, enhanced cardiomyocyte apoptosis and marked deterioration of cardiac function following MI. Mice expressing catalytically inactive PI3Kgamma also failed to mount a proper neovascularization, although cardiac dysfunction was similar to wild-type controls. CONCLUSIONS PI3Kgamma expression and catalytic activity are involved at different levels in reparative neovascularization and healing of MI.
Collapse
Affiliation(s)
- Mauro Siragusa
- Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Upper Maudlin Street, Bristol, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
47
|
Katori M, Majima M. A Novel Category of Anti-Hypertensive Drugs for Treating Salt-Sensitive Hypertension on the Basis of a New Development Concept. Pharmaceuticals (Basel) 2010; 3:59-109. [PMID: 27713243 PMCID: PMC3991021 DOI: 10.3390/ph3010059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 12/24/2009] [Accepted: 01/06/2010] [Indexed: 12/20/2022] Open
Abstract
Terrestrial animals must conserve water and NaCl to survive dry environments. The kidney reabsorbs 95% of the sodium filtered from the glomeruli before sodium reaches the distal connecting tubules. Excess sodium intake requires the renal kallikrein-kinin system for additional excretion. Renal kallikrein is secreted from the distal connecting tubule cells of the kidney, and its substrates, low molecular kininogen, from the principal cells of the cortical collecting ducts (CD). Formed kinins inhibit reabsorption of NaCl through bradykinin (BK)-B₂ receptors, localized along the CD. Degradation pathway of BK by kinin-destroying enzymes in urine differs completely from that in plasma, so that ACE inhibitors are ineffective. Urinary BK is destroyed mainly by a carboxypeptidase-Y-like exopeptidase (CPY) and partly by a neutral endopeptidase (NEP). Inhibitors of CPY and NEP, ebelactone B and poststatin, respectively, were found. Renal kallikrein secretion is accelerated by potassium and ATP-sensitive potassium (KATP) channel blockers, such as PNU-37883A. Ebelactone B prevents DOCA-salt hypertension in rats. Only high salt intake causes hypertension in animals deficient in BK-B2 receptors, tissue kallikrein, or kininogen. Hypertensive patients, and spontaneously hypertensive rats, excrete less kallikrein than normal subjects, irrespective of races, and become salt-sensitive. Ebelactone B, poststatin, and KATP channel blockers could become novel antihypertensive drugs by increase in urinary kinin levels. Roles of kinin in cardiovascular diseases were discussed.
Collapse
Affiliation(s)
- Makoto Katori
- Department of Pharmacology, School of Medicine, Kitasato University, Sagamihara, Kanagawa 228-8555, Japan.
| | - Masataka Majima
- Department of Pharmacology, School of Medicine, Kitasato University, Sagamihara, Kanagawa 228-8555, Japan
| |
Collapse
|
48
|
Abstract
Notch signalling represents a key pathway essential for normal vascular development. Recently, great attention has been focused on the implication of Notch pathway components in postnatal angiogenesis and regenerative medicine. This paper critically reviews the most recent findings supporting the role of Notch in ischaemia-induced neovascularization. Notch signalling reportedly regulates several steps of the reparative process occurring in ischaemic tissues, including sprouting angiogenesis, vessel maturation, interaction of vascular cells with recruited leucocytes and skeletal myocyte regeneration. Further characterization of Notch interaction with other signalling pathways might help identify novel targets for therapeutic angiogenesis.
Collapse
|
49
|
|