1
|
Hamid S, Rhaleb IA, Kassem KM, Rhaleb NE. Role of Kinins in Hypertension and Heart Failure. Pharmaceuticals (Basel) 2020; 13:E347. [PMID: 33126450 PMCID: PMC7692223 DOI: 10.3390/ph13110347] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
The kallikrein-kinin system (KKS) is proposed to act as a counter regulatory system against the vasopressor hormonal systems such as the renin-angiotensin system (RAS), aldosterone, and catecholamines. Evidence exists that supports the idea that the KKS is not only critical to blood pressure but may also oppose target organ damage. Kinins are generated from kininogens by tissue and plasma kallikreins. The putative role of kinins in the pathogenesis of hypertension is discussed based on human mutation cases on the KKS or rats with spontaneous mutation in the kininogen gene sequence and mouse models in which the gene expressing only one of the components of the KKS has been deleted or over-expressed. Some of the effects of kinins are mediated via activation of the B2 and/or B1 receptor and downstream signaling such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF) and/or tissue plasminogen activator (T-PA). The role of kinins in blood pressure regulation at normal or under hypertension conditions remains debatable due to contradictory reports from various laboratories. Nevertheless, published reports are consistent on the protective and mediating roles of kinins against ischemia and cardiac preconditioning; reports also demonstrate the roles of kinins in the cardiovascular protective effects of the angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARBs).
Collapse
Affiliation(s)
- Suhail Hamid
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Imane A. Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Kamal M. Kassem
- Division of Cardiology, Department of Internal Medicine, University of Louisville Medical Center, Louisville, KY 40202, USA;
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
2
|
Mesquita TRR, Campos-Mota GP, Lemos VS, Cruz JS, de Jesus ICG, Camargo EA, Pesquero JL, Pesquero JB, Capettini LDSA, Lauton-Santos S. Vascular Kinin B 1 and B 2 Receptors Determine Endothelial Dysfunction through Neuronal Nitric Oxide Synthase. Front Physiol 2017; 8:228. [PMID: 28503149 PMCID: PMC5408093 DOI: 10.3389/fphys.2017.00228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/31/2017] [Indexed: 01/06/2023] Open
Abstract
B1- and B2-kinin receptors are G protein-coupled receptors that play an important role in the vascular function. Therefore, the present study was designed to evaluate the participation of kinin receptors in the acetylcholine (ACh)-induced vascular relaxation, focusing on the protein-protein interaction involving kinin receptors with endothelial and neuronal nitric oxide synthases (eNOS and nNOS). Vascular reactivity, nitric oxide (NO·) and reactive oxygen species (ROS) generation, co-immunoprecipitation were assessed in thoracic aorta from male wild-type (WT), B1- (B1R−/−), B2- (B2R−/−) knockout mice. Some vascular reactivity experiments were also performed in a double kinin receptors knockout mice (B1B2R−/−). For pharmacological studies, selective B1- and B2-kinin receptors antagonists, NOS inhibitors and superoxide dismutase (SOD) mimetic were used. First, we show that B1- and B2-kinin receptors form heteromers with nNOS and eNOS in thoracic aorta. To investigate the functionality of these protein-protein interactions, we took advantage of pharmacological tools and knockout mice. Importantly, our results show that kinin receptors regulate ACh-induced relaxation via nNOS signaling in thoracic aorta with no changes in NO· donor-induced relaxation. Interestingly, B1B2R−/− presented similar level of vascular dysfunction as found in B1R−/− or B2R−/− mice. In accordance, aortic rings from B1R−/− or B2R−/− mice exhibit decreased NO· bioavailability and increased superoxide generation compared to WT mice, suggesting the involvement of excessive ROS generation in the endothelial dysfunction of B1R−/− and B2R−/− mice. Alongside, we show that impaired endothelial vasorelaxation induced by ACh in B1R−/− or B2R−/− mice was rescued by the SOD mimetic compound. Taken together, our findings show that B1- and B2-kinin receptors regulate the endothelium-dependent vasodilation of ACh through nNOS activity and indicate that molecular disturbance of short-range interaction between B1- and B2-kinin receptors with nNOS might be involved in the oxidative pathogenesis of endothelial dysfunction.
Collapse
Affiliation(s)
| | - Gianne P Campos-Mota
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Virgínia S Lemos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Itamar C G de Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - Enilton A Camargo
- Department of Physiology, Federal University of SergipeSão Cristóvão, Brazil
| | - Jorge L Pesquero
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas GeraisBelo Horizonte, Brazil
| | - João B Pesquero
- Department of Biophysics, Federal University of São PauloSão Paulo, Brazil
| | | | | |
Collapse
|
3
|
Ienaga K, Sohn M, Naiki M, Jaffa AA. Creatinine metabolite, HMH (5-hydroxy-1-methylhydantoin; NZ-419), modulates bradykinin-induced changes in vascular smooth muscle cells. J Recept Signal Transduct Res 2014; 34:195-200. [DOI: 10.3109/10799893.2013.876039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
4
|
Rhaleb NE, Yang XP, Carretero OA. The kallikrein-kinin system as a regulator of cardiovascular and renal function. Compr Physiol 2013; 1:971-93. [PMID: 23737209 DOI: 10.1002/cphy.c100053] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autocrine, paracrine, endocrine, and neuroendocrine hormonal systems help regulate cardio-vascular and renal function. Any change in the balance among these systems may result in hypertension and target organ damage, whether the cause is genetic, environmental or a combination of the two. Endocrine and neuroendocrine vasopressor hormones such as the renin-angiotensin system (RAS), aldosterone, and catecholamines are important for regulation of blood pressure and pathogenesis of hypertension and target organ damage. While the role of vasodepressor autacoids such as kinins is not as well defined, there is increasing evidence that they are not only critical to blood pressure and renal function but may also oppose remodeling of the cardiovascular system. Here we will primarily be concerned with kinins, which are oligopeptides containing the aminoacid sequence of bradykinin. They are generated from precursors known as kininogens by enzymes such as tissue (glandular) and plasma kallikrein. Some of the effects of kinins are mediated via autacoids such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF), and/or tissue plasminogen activator (tPA). Kinins help protect against cardiac ischemia and play an important part in preconditioning as well as the cardiovascular and renal protective effects of angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARB). But the role of kinins in the pathogenesis of hypertension remains controversial. A study of Utah families revealed that a dominant kallikrein gene expressed as high urinary kallikrein excretion was associated with a decreased risk of essential hypertension. Moreover, researchers have identified a restriction fragment length polymorphism (RFLP) that distinguishes the kallikrein gene family found in one strain of spontaneously hypertensive rats (SHR) from a homologous gene in normotensive Brown Norway rats, and in recombinant inbred substrains derived from these SHR and Brown Norway rats this RFLP cosegregated with an increase in blood pressure. However, humans, rats and mice with a deficiency in one or more components of the kallikrein-kinin-system (KKS) or chronic KKS blockade do not have hypertension. In the kidney, kinins are essential for proper regulation of papillary blood flow and water and sodium excretion. B2-KO mice appear to be more sensitive to the hypertensinogenic effect of salt. Kinins are involved in the acute antihypertensive effects of ACE inhibitors but not their chronic effects (save for mineralocorticoid-salt-induced hypertension). Kinins appear to play a role in the pathogenesis of inflammatory diseases such as arthritis and skin inflammation; they act on innate immunity as mediators of inflammation by promoting maturation of dendritic cells, which activate the body's adaptive immune system and thereby stimulate mechanisms that promote inflammation. On the other hand, kinins acting via NO contribute to the vascular protective effect of ACE inhibitors during neointima formation. In myocardial infarction produced by ischemia/reperfusion, kinins help reduce infarct size following preconditioning or treatment with ACE inhibitors. In heart failure secondary to infarction, the therapeutic effects of ACE inhibitors are partially mediated by kinins via release of NO, while drugs that activate the angiotensin type 2 receptor act in part via kinins and NO. Thus kinins play an important role in regulation of cardiovascular and renal function as well as many of the beneficial effects of ACE inhibitors and ARBs on target organ damage in hypertension.
Collapse
Affiliation(s)
- Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.
| | | | | |
Collapse
|
5
|
Webb JG, Tan Y, Jaffa MA, Jaffa AA. Evidence for prostacyclin and cAMP upregulation by bradykinin and insulin-like growth factor 1 in vascular smooth muscle cells. J Recept Signal Transduct Res 2010; 30:61-71. [PMID: 20082561 DOI: 10.3109/10799890903563768] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Although bradykinin (BK) and insulin like growth factor-1 (IGF-1) have been shown to modulate the functional and structural integrity of the arterial wall, the cellular mechanisms through which this regulation occurs is still undefined. The present study examined the role of second messenger molecules generated by BK and IGF-1 that could ultimately result in proliferative or antiproliferative signals in vascular smooth muscle cells (VSMC). Activation of BK or IGF-1 receptors stimulated the synthesis and release of prostacyclin (PGI(2)) leading to increased production of cAMP in VSMC. Inhibition of p42/p44(mapk) or src kinases prevented the increase in PGI(2) and cAMP observed in response to BK or IGF-1, indicating a role for these kinases in the regulation of cPLA(2) activity in the VSMC. Inhibition of PKC failed to alter production of PGI(2) in response to BK, but further increased both p42/p44(mapk) activation and the synthesis of PGI(2) produced in response to IGF-1. In addition, both BK and IGF-1 significantly induced the expression of c-fos mRNA levels in VSMC, and this effect of BK was accentuated in the presence a cPLA(2) inhibitor. Finally, inhibition of cPLA(2) activity and/or cyclooxygenase activity enhanced the expression of collagen I mRNA levels in response to BK and IGF-1 stimulation. These findings indicate that the effect of BK or IGF-1 to stimulate VSMC growth is an integrated response to the activation of multiple signaling pathways. Thus, the excessive cell growth that occurs in certain forms of vascular disease could reflect dysfunction in one or more of these pathways.
Collapse
Affiliation(s)
- Jerry G Webb
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
6
|
Abstract
Remodeling of the arterial wall occurs mainly as a consequence of increased wall stress caused by hypertension. In this issue of the JCI, Azizi et al. report that in humans with a kallikrein gene polymorphism that lowers kallikrein activity, the brachial artery undergoes eutrophic inward remodeling in the absence of hypertension or other hemodynamic changes. It has also been reported that alterations of the kallikrein-kinin system are associated with formation of aortic aneurysms. Conversely, after vascular injury, kinins mediate the beneficial effect of angiotensin-converting enzyme inhibitors that prevent neointima formation. These findings raise the intriguing possibility that decreased kallikrein-kinin system activity may play an important role in the pathogenesis of vascular remodeling and disease, while increased activity may have a beneficial effect.
Collapse
Affiliation(s)
- Oscar A Carretero
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| |
Collapse
|
7
|
The Kallikrein-Kinin System as a Regulator of Cardiovascular and Renal Function. Hypertension 2005. [DOI: 10.1016/b978-0-7216-0258-5.50110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
8
|
Tan Y, Hutchison FN, Jaffa AA. Mechanisms of angiotensin II-induced expression of B2 kinin receptors. Am J Physiol Heart Circ Physiol 2004; 286:H926-32. [PMID: 14766673 DOI: 10.1152/ajpheart.00757.2003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the primary roles of the kallikreinkinin system and the renin-angiotensin system are quite divergent, they are often intertwined under pathophysiological conditions. We examined the effect of ANG II on regulation of B(2) kinin receptors (B2KR) in vascular cells. Vascular smooth muscle cells (VSMC) were treated with ANG II in a concentration (10(-9)-10(-6) M)- and time (0-24 h)-dependent manner, and B2KR protein and mRNA levels were measured by Western blots and PCR, respectively. A threefold increase in B2KR protein levels was observed as early as 6 h, with a peak response at 10(-7) M. ANG II (10(-7) M) also increased B2KR mRNA levels twofold 4 h after stimulation. Actinomycin D suppressed the increase in B2KR mRNA and protein levels induced by ANG II. To elucidate the receptor subtype involved in mediating this regulation, VSMC were pretreated with losartan (AT(1) receptor antagonist) and/or PD-123319 (AT(2) receptor antagonist) at 10 microM for 30 min, followed by ANG II (10(-7) M) stimulation. Losartan completely blocked the ANG II-induced B2KR increase, whereas PD-123319 had no effect. In addition, expression of B2KR mRNA levels was decreased in AT(1A) receptor knockout mice. Finally, to determine whether ANG II stimulates B2KR expression via activation of the MAPK pathway, VSMC were pretreated with an inhibitor of p42/p44(mapk) (PD-98059) and/or an inhibitor of p38(mapk) (SB-202190), followed by ANG II (10(-7) M) for 24 h. Selective inhibition of the p42/p44(mapk) pathway significantly blocked the ANG II-induced increase in B2KR expression. These findings demonstrate that ANG II regulates expression of B2KR in VSMC and provide a rationale for studying the interaction between ANG II and bradykinin in the pathogenesis of vascular dysfunction.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/cytology
- Cells, Cultured
- Extracellular Fluid/metabolism
- Gene Expression/drug effects
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Rats
- Rats, Sprague-Dawley
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Signal Transduction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Yan Tan
- Dept. of Medicine, Endocrinology-Diabetes-Medical Genetics, Medical Univ. of South Carolina, 114 Doughty St., PO Box 250776, Charleston, SC 29425, USA
| | | | | |
Collapse
|
9
|
Diamond MP, El-Hammady E, Wang R, Saed G. Regulation of matrix metalloproteinase-1 and tissue inhibitor of matrix metalloproteinase-1 by dichloroacetic acid in human fibroblasts from normal peritoneum and adhesions. Fertil Steril 2004; 81:185-90. [PMID: 14711564 DOI: 10.1016/j.fertnstert.2003.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To examine the role of stimulation of aerobic metabolism on the differential expression of matrix metalloproteinase-1 (MMP-1) and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), which are differentially regulated in fibroblasts isolated from normal human peritoneum and adhesions. DESIGN Tissue culture study. SETTING University research laboratory. PATIENT(S) Human fibroblasts cultures from normal peritoneum and adhesions that were exposed to dichloroacetic acid (DCA; 0 and 100 microg/mL) for 24 hours under normal and hypoxic conditions. MAIN OUTCOME MEASURE(S) Real-time reverse-transcription polymerase chain reaction of MMP-1, TIMP-1, and beta-actin. RESULT(S) Dichloroacetic acid stimulated peritoneal fibroblast MMP-1 mRNA expression under normoxic conditions; this stimulation was lost during hypoxia. In adhesion fibroblasts, DCA increased MMP-1 mRNA expression; this effect was reversed by hypoxia. Expression of TIMP-1 mRNA was insignificantly increased by DCA in normal peritoneal and adhesion fibroblasts under normoxic conditions; however under hypoxic conditions, DCA reduced TIMP-1 mRNA expression from both. CONCLUSION(S) Regulation of metabolic activity of peritoneal cells may provide a target for future interventions for reduction of development of postoperative adhesions, particularly as it relates to healing of peritoneal sites that did not previously have adhesions as opposed to sites that underwent lysis of preexistent adhesions.
Collapse
Affiliation(s)
- Michael P Diamond
- Department of Obstetrics and Gynecology, Wayne State University/Detroit Medical Center, Hutzel Hospital, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
10
|
Jaffa AA, Durazo-Arvizu R, Zheng D, Lackland DT, Srikanth S, Garvey WT, Schmaier AH. Plasma prekallikrein: a risk marker for hypertension and nephropathy in type 1 diabetes. Diabetes 2003; 52:1215-21. [PMID: 12716755 DOI: 10.2337/diabetes.52.5.1215] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The relevance and significance of the plasma kallikrein/kinin system as a risk factor for the development of vascular complications in diabetic patients was explored in a cross-sectional study. We measured the circulating levels of plasma prekallikrein (PK) activity, factor XII, and high-molecular weight kininogen in the plasma of 636 type 1 diabetic patients from the Diabetes Control and Complications Trial/Epidemiology and Diabetes Intervention and Complications Study cohort. The findings demonstrated that type 1 diabetic patients with blood pressure > or =140/90 mmHg have increased PK levels compared with type 1 diabetic patients with blood pressure <140/90 (1.53 +/- 0.07 vs. 1.27 +/- 0.02 units/ml; P < 0.0001). Regression analysis also determined that plasma PK levels positively and significantly correlated with diastolic (DBP) and systolic blood pressures (SBP) as continuous variables (r = 0.17 and 0.18, respectively; P < 0.0001). In multivariate regression analysis, the semipartial r(2) value for PK was 2.93% for SBP and 2.92% for DBP (P < 0.0001). A positive correlation between plasma PK levels and the urinary albumin excretion rate (AER) was also observed (r = 0.16, P < 0.0001). In categorical analysis, patients with macroalbuminuria had a significantly higher level of plasma PK than normoalbuminuric patients (1.45 +/- 0.08 vs. 1.27 +/- 0.02 units/ml; P < 0.01), whereas microalbuminuric patients had an intermediate PK value (1.38 +/- 0.05 units/ml; P = NS). Among patients in the microalbuminuric subgroup, we observed a positive and independent correlation between PK and AER in univariate and multivariate regression analysis (r = 0.27, P < 0.03; n = 63). We concluded that in type 1 diabetes, 1) PK levels are elevated in association with increased blood pressure; 2) PK levels are independently correlated with AER and are categorically elevated in patients with macroalbuminuria; and 3) although the positive correlation between PK and AER within the subgroups of patients with microalbuminuria suggest that PK could be a marker for progressive nephropathy, longitudinal studies will be necessary to address this issue.
Collapse
Affiliation(s)
- Ayad A Jaffa
- Department of Medicine, Endocrinology-Diabetes-Medical Genetics, Medical University of South Carolina, 114 Doughty Street, PO Box 250776, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
11
|
Yayama K, Kunimatsu N, Teranishi Y, Takano M, Okamoto H. Tissue kallikrein is synthesized and secreted by human vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1593:231-8. [PMID: 12581867 DOI: 10.1016/s0167-4889(02)00393-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The generation of kinins on the surface of vascular endothelium has been postulated in two pathways involving plasma kallikrein and tissue kallikrein; the former pathway has been well documented, but the latter is controversial. To clarify the presence of a kinin-generating system on endothelium, we examined whether human umbilical vein endothelial cells (HUVEC) synthesize and release tissue kallikrein in vitro. Kallikrein-like activity hydrolyzing a peptide Pro-Phe-Arg-4-methyl-coumaryl-7-amide was detected in the culture medium of HUVEC and was inhibited by aprotinin but not by soybean trypsin inhibitor. Western blotting of HUVEC medium using anti-human tissue kallikrein antibodies demonstrated the release of tissue kallikrein from HUVEC, and the reverse transcription-polymerase chain reaction (RT-PCR) followed by Southern blotting revealed the expression of tissue kallikrein mRNA in HUVEC. HUVEC metabolically labeled with [35S]methionine released radioactive proteins corresponding to tissue kallikrein. RT-PCR also showed the expression of low-molecular-weight kininogen (L-kininogen) mRNA in HUVEC. The cGMP levels in HUVEC were significantly elevated by the incubation with angiotensin converting enzyme inhibitor, lisinopril, and the elevation was completely inhibited by aprotinin or bradykinin B2-receptor antagonist, FR172357. These results suggest that the endothelial cells continuously release an active form of tissue kallikrein which enables generation of kinins on the vascular endothelium.
Collapse
Affiliation(s)
- Katsutoshi Yayama
- Department of Pharmacology, Faculty of Pharmaceutical Sciences and High Technology Research Center, Kobe Gakuin University, Ikawadani-cho, Nishi-ku, 651-2180, Kobe, Japan
| | | | | | | | | |
Collapse
|
12
|
Monsees TK, Blöcher S, Loddo C, Steger K, Schill WB. Tissue kallikrein and bradykinin B 2
receptors in the reproductive tract of the male rat. Andrologia 2003. [DOI: 10.1111/j.1439-0272.2003.tb00823.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Monsees TK, Blöcher S, Loddo C, Steger K, Schill WB. Tissue kallikrein and bradykinin B2 receptors in the reproductive tract of the male rat. Andrologia 2003; 35:24-31. [PMID: 12558526 DOI: 10.1046/j.1439-0272.2003.00534.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The setting of a local tissue kallikrein kinin system (tKKS) within the reproductive organs of the male rat was investigated by analysing bradykinin subtype 2 receptor (B2R) gene expression and cellular distribution of B2R protein and the kinin-liberating protease tissue kallikrein (tK). Reverse transcription-polymerase chain reaction showed B2R expression in testis, epididymis and prostate from prepubertal and sexually mature rats. In mature testis, in situ hybridization and immunohistochemistry localized B2R mRNA and protein besides endothelial cells of blood vessels exclusively on pachytene spermatocytes and round and elongated spermatids. B2R expression within the seminiferous tubules was found to be dependent on the stage of the spermatogenic cycle. In pre-pubertal rat testis, B2R mRNA and protein were additionally located in peritubular cells. In the testis, specific staining for tK occurred in addition to endothelial cells of blood vessels on the acrosomal cap of round and elongated spermatids. This immunostaining was also stage-dependent. In the epididymis, tK was detected on epithelial cells near the apical surface. The stage-dependent specific expression of tK and bradykinin B2Rs in developing germ cells and peritubular cells suggests a potential role of the tKKS in the local regulation of spermatogenesis and seminiferous tubule function.
Collapse
Affiliation(s)
- T K Monsees
- Centre of Dermatology and Andrology, Justus Liebig University, Giessen, Germany.
| | | | | | | | | |
Collapse
|
14
|
Hayashi I, Amano H, Yoshida S, Kamata K, Kamata M, Inukai M, Fujita T, Kumagai Y, Furudate SI, Majima M. Suppressed angiogenesis in kininogen-deficiencies. J Transl Med 2002; 82:871-80. [PMID: 12118089 DOI: 10.1097/01.lab.0000018885.36823.d6] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We investigated whether the kinin-generating system enhanced angiogenesis in chronic and proliferative granuloma and in tumor-surrounding stroma. In rat sponge implants, angiogenesis was gradually developed in normal Brown Norway Kitasato rats (BN-Ki). The development of angiogenesis was significantly suppressed in kininogen-deficient Brown Norway Katholiek rats (BN-Ka). The angiogenesis enhanced by basic fibroblast growth factor was also significantly less marked in BN-Ka than in BN-Ki. Naturally occurring angiogenesis was significantly suppressed by B(1) or B(2) antagonist. mRNA of vascular endothelial growth factor was more highly expressed in the granulation tissues in BN-Ki than in BN-Ka. Daily topical injections of aprotinin, but not of soy bean trypsin inhibitor, suppressed angiogenesis. Daily topical injections of low-molecular weight kininogen enhanced angiogenesis in BN-Ka. Topical injections of serum from BN-Ki, but not from BN-Ka, also facilitated angiogenesis in BN-Ka. FR190997, a nonpeptide mimic of bradykinin, promoted angiogenesis markedly, with concomitant increases in vascular endothelial growth factor mRNA. Angiogenesis in the granulation tissues around the implanted Millipore chambers containing Walker-256 cells was markedly more suppressed in BN-Ka than in BN-Ki. Our results suggest that endogenous kinin generated from the tissue kallikrein-kinin system enhances angiogenesis in chronic and proliferative granuloma and in the stroma surrounding a tumor. Thus, the agents for the kinin-generating system and/or kinin receptor signaling may become useful tools for controlling angiogenesis.
Collapse
Affiliation(s)
- Izumi Hayashi
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Di Girolamo G, González E, Livio D, de los Santos AR, Martí ML, de Gimeno MAF. The effect of Enalapril on PGI(2) and NO levels in hypertensive patients. Prostaglandins Leukot Essent Fatty Acids 2002; 66:493-8. [PMID: 12144869 DOI: 10.1054/plef.2002.0388] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effects of the angiotensin-converting enzyme inhibitors (ACEIs), may be partially mediated by the kinins' paracrine influence. Their actions may be exerted through nitric oxide and prostacyclin (PGI(2)) synthesis stimulation. The aim of our study was to determine whether the antihypertensive effect of Enalapril correlated with the increment in the plasmatic levels of NO and PGI(2) in essential moderate hypertensive patients. Normalization of blood pressure was observed in 20 patients, four on the 28th day, 15 on the 42th day and one on the 56th day. Enalapril-respondent subjects showed increased nitrate/nitrite levels on the 14th day (30% increment), on the 28th day (64%), on the 42th day (93.5%) and on the 56th day (96.2%) compared with basal levels, but they did not modify the circulating 6-keto PGF(1 alpha) levels. Four non-respondent patients showed a diminution in nitrate/nitrite and 6-keto PGF(1 alpha) circulating levels along the treatment. We conclude that the administration of 5-30 mg of Enalapril increases circulating NO metabolites in respondent-essential hypertensive subjects. The lack of responsiveness to the treatment may be related to the presence of risk factors such as those linked to an increase of oxidative stress. Finally, we consider that the evaluation of circulating NO may represent a predictive of the response to Enalapril in essential hypertensive patients.
Collapse
Affiliation(s)
- G Di Girolamo
- Segunda Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
16
|
Dedio J, Wiemer G, Rütten H, Dendorfer A, Schölkens BA, Müller-Esterl W, Wohlfart P. Tissue kallikrein KLK1 is expressed de novo in endothelial cells and mediates relaxation of human umbilical veins. Biol Chem 2001; 382:1483-90. [PMID: 11727832 DOI: 10.1515/bc.2001.182] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Bradykinin released by the endothelium is thought to play an important local role in cardiovascular regulation. However, the molecular identity of endothelial proteases liberating bradykinin from its precursors remained unclear. Using RT-PCR and Southern blotting techniques we detected mRNA for tissue kallikrein (KLK1) in human umbilical vein endothelial cells and in bovine aortic endothelial cells. Protein expression was confirmed by precipitation of KLK1 from lysates of endothelial cells pre-labeled with [35S]-cysteine/methionine. Partial purification of tissue kallikrein from total endothelial cell extracts resulted in a protein triplet of about 50 kDa in Western blots using specific anti-KLK1 antibodies. The immunodetection of tissue kallikrein antigen in the fractions from ion exchange chromatography correlated with the presence of amidolytic tissue kallikrein activity. Stimulation of endothelial cells with angiotensin II (ANG-II), which recently has been shown to activate the vascular kinin system and to cause vasodilation, resulted in the release of bradykinin and kallidin. ANG-II-dependent relaxation of pre-constricted rings from human umbilical veins was abolished in the presence of a specific tissue kallikrein inhibitor. We conclude that endothelial cells de novo express significant amounts of tissue kallikrein, which likely serves in the local generation of vasoactive kinins.
Collapse
Affiliation(s)
- J Dedio
- Aventis Pharma Deutschland GmbH, Cardiovascular Disease Group, Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Douillet CD, Velarde V, Christopher JT, Mayfield RK, Trojanowska ME, Jaffa AA. Mechanisms by which bradykinin promotes fibrosis in vascular smooth muscle cells: role of TGF-beta and MAPK. Am J Physiol Heart Circ Physiol 2000; 279:H2829-37. [PMID: 11087238 DOI: 10.1152/ajpheart.2000.279.6.h2829] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulation of extracellular matrix (ECM) is a hallmark feature of vascular disease. We have previously shown that hyperglycemia induces the expression of B(2)-kinin receptors in vascular smooth muscle cells (VSMC) and that bradykinin (BK) and hyperglycemia synergize to stimulate ECM production. The present study examined the cellular mechanisms through which BK contributes to VSMC fibrosis. VSMC treated with BK (10(-8) M) for 24 h significantly increased alpha(2)(I) collagen mRNA levels. In addition, BK produced a two- to threefold increase in alpha(2)(I) collagen promoter activity in VSMC transfected with a plasmid containing the alpha(2)(I) collagen promoter. Furthermore, treatment of VSMC with BK for 24 h produced a two- to threefold increase in the secretion rate of tissue inhibitor of metalloproteinase 1 (TIMP-1). The increase in alpha(2)(I) collagen mRNA levels and alpha(2)(I) collagen promoter activity, as well as TIMP-1 secretion, in response to BK were blocked by anti-transforming growth factor-beta (anti-TGF-beta) neutralizing antibodies. BK (10(-8) M) increased the endogenous production of TGF-beta1 mRNA and protein levels. Inhibition of the mitogen-activated protein kinase (MAPK) pathway by PD-98059 inhibited the increase of alpha(2)(I) collagen promoter activity, TIMP-1 production, and TGF-beta1 protein levels observed in response to BK. These findings provide the first evidence that BK induces collagen type I and TIMP-1 production via autocrine activation of TGF-beta1 and implicate MAPK pathway as a key player in VSMC fibrosis in response of BK.
Collapse
Affiliation(s)
- C D Douillet
- Department of Medicine, Medical University of South Carolina, Charleston, USA
| | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Nakajima S, Ito H, Hayashi I, Kuribayashi Y, Okumura T, Yajima Y, Katori M, Majima M. Inhibition of kinin degradation on the luminal side of renal tubules reduces high blood pressure in deoxycorticosterone acetate salt-treated rats. Clin Exp Pharmacol Physiol 2000; 27:80-7. [PMID: 10696533 DOI: 10.1046/j.1440-1681.2000.03209.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1. To determine whether the antihypertensive response in deoxycorticosterone acetate (DOCA) salt-treated rats was mediated by kinins on the luminal side of renal tubules or in the circulation, selective urinary kininase inhibitors were administered to normal Brown Norway Kitasato (BN-Ki) rats and kininogen-deficient Brown Norway Katholiek (BN-Ka) rats. 2. Kinins were degraded by neutral endopeptidase (NEP) and carboxypeptidase Y-like kininase (CPY) in urine, but were inactivated mainly by angiotensin-converting enzyme (ACE) in the plasma. 3. Ebelactone B inhibited CPY, while poststatin inhibited CPY and NEP. 4. Daily administration of poststatin (5 mg/kg per day, s.c.) for 3 days reduced blood pressure (BP) in DOCA salt-treated BN-Ki rats, but not in BN-Ka rats. 5. Ebelactone B (5 mg/kg per day, s.c.) also reduced BP in BN-Ki rats, which was accompanied by increased urinary sodium excretion, but had no effect on BP in BN-Ka rats. 6. Lisinopril (5 mg/kg per day, s.c.) had no effect on BP in either rat strain. 7. Arterial kinin levels in BN-Ki rats increased significantly (2.2-4.6 pg/mL) with captopril (10 mg/kg, s.c.). However, arterial kinin levels that induced hypotension following the infusion of bradykinin (1000 ng/kg per min, i.v.) were 110-fold higher than endogenous arterial kinin levels attained following captopril. 8. These results suggest that inhibition of kinin degradation on the luminal side of the renal tubules may effectively attenuate hypertension.
Collapse
Affiliation(s)
- S Nakajima
- Department of Internal Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Saed GM, Zhang W, Chegini N, Holmdahl L, Diamond MP. Alteration of type I and III collagen expression in human peritoneal mesothelial cells in response to hypoxia and transforming growth factor-beta1. Wound Repair Regen 1999; 7:504-10. [PMID: 10633010 DOI: 10.1046/j.1524-475x.1999.00504.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Overexpression and accumulation of extracellular matrix is central to peritoneal adhesion formation following surgically induced tissue trauma. Transforming growth factor-beta1 and hypoxia have been implicated in tissue fibrosis and postoperative adhesion formation. To extend this observation we examined whether transforming growth factor-beta1 and/or hypoxia regulate the expression of type I and III collagen in human peritoneal mesothelial cells. Cultured human mesothelial cells were maintained under hypoxia (2% oxygen), or treated with transforming growth factor-beta1 (1 ng/ml) or a combination of hypoxia and transforming growth factor-beta1. Total cellular RNA from treated and untreated cells were collected and subjected to multiplex reverse transcription/polymerase chain reaction to quantitate collagen I and III mRNA levels in response to these treatments. The results indicate that 6 hours of hypoxia increased collagen III mRNA by 7.2 fold which was further increased to 9.4 fold following transforming growth factor-beta1 treatment; in contrast collagen I mRNA decreased by 0.42 fold which was further decreased by 0.3 fold following transforming growth factor-beta1 treatment. Transforming growth factor-beta1 treatment under normal conditions resulted in an 8.4-fold increase and a 0.3-fold decrease in collagen III and I mRNA levels, respectively. Hypoxia treatment also resulted in a 1.9-fold increase in transforming growth factor-beta1 mRNA level compared with control. The ratio of type III/I collagen was increased in response to transforming growth factor-beta1 treatment under hypoxic condition. In conclusion, the data suggest that hypoxia may modulate extracellular matrix production by human mesothelial cells via a transforming growth factor-beta1 dependent mechanism.
Collapse
Affiliation(s)
- G M Saed
- Depatment of Obstetrics and Gynecology, Hutzel Hospital, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
22
|
Naidu PS, Velarde V, Kappler CS, Young RC, Mayfield RK, Jaffa AA. Calcium-calmodulin mediates bradykinin-induced MAPK phosphorylation and c-fos induction in vascular cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H1061-8. [PMID: 10484429 DOI: 10.1152/ajpheart.1999.277.3.h1061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The vasoactive peptide bradykinin (BK) has been implicated in the pathophysiology of a number of vascular wall abnormalities, but the cellular mechanisms by which BK generates second messengers that alter vascular function are as yet undefined. Exposure of vascular smooth muscle cells (VSMC) to BK (10(-7) M) produced a rapid and transient rise in intracellular calcium, which preceded an increase in tyrosine phosphorylation of mitogen-activated protein kinase (MAPK). MAPK activation by BK was observed as early as 1 min, peaked at 5 min, and returned to baseline by 20 min. Treatment of cells with the intracellular calcium chelator EGTA-acetoxymethyl ester inhibited BK-stimulated MAPK activation, suggesting that intracellular calcium mobilization contributes to the activation of MAPK. The calmodulin inhibitor W-7 also markedly inhibited BK-induced MAPK phosphorylation in the cytoplasm as well as in the nucleus. Moreover, the BK-induced increase in c-fos mRNA levels was significantly inhibited by the calmodulin inhibitor, indicating that calmodulin is required for BK signaling leading to c-fos induction. These results implicate the calcium-calmodulin pathway in the mechanisms for regulating MAPK activity and the resultant c-fos expression induced by BK in VSMC.
Collapse
Affiliation(s)
- P S Naidu
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
23
|
Velarde V, Ullian ME, Morinelli TA, Mayfield RK, Jaffa AA. Mechanisms of MAPK activation by bradykinin in vascular smooth muscle cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:C253-61. [PMID: 10444401 DOI: 10.1152/ajpcell.1999.277.2.c253] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation is a prominent feature of the atherosclerotic process occurring after endothelial injury. A vascular wall kallikrein-kinin system has been described. The contribution of this system to vascular disease is undefined. In the present study we characterized the signal transduction pathway leading to mitogen-activated protein kinase (MAPK) activation in response to bradykinin (BK) in VSMC. Addition of 10(-10)-10(-7) M BK to VSMC resulted in a rapid and concentration-dependent increase in tyrosine phosphorylation of several 144- to 40-kDa proteins. This effect of BK was abolished by the B(2)-kinin receptor antagonist HOE-140, but not by the B(1)-kinin receptor antagonist des-Arg(9)-Leu(8)-BK. Immunoprecipitation with anti-phosphotyrosine antibodies followed by immunoblot revealed that 10(-9) M BK induced tyrosine phosphorylation of focal adhesion kinase (p125(FAK)). BK (10(-8) M) promoted the association of p60(src) with the adapter protein growth factor receptor binding protein-2 and also induced a significant increase in MAPK activity. Pertussis and cholera toxins did not inhibit BK-induced MAPK tyrosine phosphorylation. Protein kinase C downregulation by phorbol 12-myristate 13-acetate and/or inhibitors to protein kinase C, p60(src) kinase, and MAPK kinase inhibited BK-induced MAPK tyrosine phosphorylation. These findings provide evidence that activation of the B(2)-kinin receptor in VSMC leads to generation of multiple second messengers that converge to activate MAPK. The activation of this crucial kinase by BK provides a strong rationale to investigate the mitogenic actions of BK on VSMC proliferation in disease states of vascular injury.
Collapse
MESH Headings
- Animals
- Bradykinin/pharmacology
- Calcium-Calmodulin-Dependent Protein Kinases/drug effects
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cells, Cultured
- Cholera Toxin/pharmacology
- Cytoplasm/enzymology
- Enzyme Activation/physiology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Phosphorylation/drug effects
- Protein Kinase C/physiology
- Protein-Tyrosine Kinases/drug effects
- Protein-Tyrosine Kinases/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Bradykinin/physiology
- Tyrosine/metabolism
- Virulence Factors, Bordetella/pharmacology
Collapse
Affiliation(s)
- V Velarde
- Department of Medicine, Medical University of South Carolina and Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
24
|
Pieper SJ, Fenton AM, Kurachi Y, Shen WK. Bradykinin modulates arginine vasopressin-induced calcium influx in vascular myocytes. Int J Cardiol 1999; 69:191-200. [PMID: 10549843 DOI: 10.1016/s0167-5273(99)00031-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
UNLABELLED We investigated direct, endothelium-independent effects of bradykinin on arginine vasopressin-induced calcium influx in vascular smooth muscle cells. We studied cultured rat vascular smooth muscle cells by using the whole-cell voltage-clamp and calcium fluorescence imaging methods. Exposing cultured vascular smooth muscle cells (A7r5 cell line) to arginine vasopressin (100 nM) produced a transient increase in [Ca2+]i, followed by a sustained increase in [Ca2+]i. This was readily reversible (n=28). At a holding potential of -40 to -60 mV, arginine vasopressin induced a sustained inward current correlated with a sustained increase in [Ca2+]i. Bradykinin (30 nM to 30 microM) had no effect on arginine vasopressin-induced [Ca2+]i transients. However, during the sustained phase of increased [Ca2+]i, bradykinin reversibly attenuated relative fluorescence and inward current in the presence of arginine vasopressin (n=14). This was concentration dependent and inhibited by [D-Phe7]-bradykinin (30 microM), a kinin receptor antagonist. Also, sustained arginine vasopressin-mediated increases in [Ca2+]i and inward current were attenuated by Ca2+-free or La3+-supplemented perfusate but not by nifedipine (n=5). CONCLUSIONS (1) Bradykinin can attenuate arginine vasopressin-induced and sustained Ca2+ influx and sustained inward current through a novel endothelium-independent process. (2) The direct effect of bradykinin on arginine vasopressin-induced increases in [Ca2+]i sustained Ca2+ influx in vascular smooth muscle cells is concentration dependent and kinin-receptor mediated. (3) Arginine vasopressin-induced sustained [Ca2+]i elevation correlates with the activation of a dihydropyridine-insensitive, Ca2+-conducting inward current.
Collapse
Affiliation(s)
- S J Pieper
- Division of Cardiovascular Diseases and Internal Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
25
|
Wolf WC, Harley RA, Sluce D, Chao L, Chao J. Localization and expression of tissue kallikrein and kallistatin in human blood vessels. J Histochem Cytochem 1999; 47:221-8. [PMID: 9889257 DOI: 10.1177/002215549904700210] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue kallikrein releases kinins by specific proteolysis, an activity inhibited by kallistatin. In this study, kallikrein and kallistatin were localized to endothelial and smooth muscle cells of large, medium, and small normal blood vessels by immunohistochemical techniques. Immunostaining for both proteins was strong in the endothelium of all sizes of blood vessels and was more intense in medial smooth muscle cells of small and medium-sized blood vessels than in elastic arteries. The sites of synthesis by endothelial and smooth muscle cells were demonstrated in normal blood vessels of all sizes by in situ hybridization histochemistry. Kallikrein and kallistatin levels were measured by immunoassays in homogenates of human aorta, vena cava, and iliac artery and vein. Tissue kallikrein and kallistatin transcripts were identified in human blood vessels by RT-PCR followed by Southern blot analysis with specific oligonucleotide probes. The results demonstrated the expression and co-localization of tissue kallikrein and kallistatin in human vessels and suggest a potential role of kallistatin in regulating tissue kallikrein in blood vessels.
Collapse
Affiliation(s)
- W C Wolf
- Departments of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
26
|
Okamoto H, Yayama K, Shibata H, Nagaoka M, Takano M. Kininogen expression by rat vascular smooth muscle cells: stimulation by lipopolysaccharide and angiotensin II. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1404:329-37. [PMID: 9739161 DOI: 10.1016/s0167-4889(98)00074-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To identify the presence of a local kallikrein-kinin system in vascular wall, we have studied whether rat vascular smooth muscle cells (VSMC) express kininogen in vitro and in vivo. Western blots using anti-T-kininogen antibody revealed the presence of T-kininogen in conditioned medium of cultured VSMC. T-Kininogen secretion by VSMC was markedly enhanced by the addition of lipopolysaccharide (LPS), angiotensin II (AII) and phorbol 12-myristate 13-acetate (PMA) to the culture. Experiments using specific inhibitors for protein kinases and on the PMA-induced down-regulation of protein kinase C suggested that a protein kinase C-dependent or unidentified pathway is involved in AII or LPS action, respectively. The intravenous injection of LPS (0.5 mg/kg) resulted in an increase in T-kininogen mRNA levels in the vascular smooth muscle of rat aorta, peaking at 16 h. Polyacrylamide gel electrophoresis of cDNA products generated by reverse transcription-polymerase chain reaction (RT-PCR) from aortic mRNA using primers specific for either T- or low-molecular-weight kininogen revealed that rat vascular smooth muscle expressed T-kininogen gene but not low-molecular-weight kininogen gene, and that LPS exclusively stimulated T-kininogen expression. The mRNA for high-molecular-weight kininogen was undetectable in either aortic smooth muscle or cultured VSMC by means of RT-PCR analysis. RT-PCR using specific primers for rat tissue kallikrein genes showed that aortic smooth muscle expressed KLK1 (true kallikrein) mRNA, but not KLK10 (T-kininogenase) mRNA. These results demonstrated that rat VSMC are a source of T-kininogen but not of low-molecular-weight- or high-molecular-weight kininogen, in contrast to the expression of true kallikrein but not of T-kininogenase by these cells.
Collapse
MESH Headings
- Angiotensin II/antagonists & inhibitors
- Angiotensin II/pharmacology
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Blotting, Southern
- Blotting, Western
- Cells, Cultured
- Culture Media, Conditioned/analysis
- Enzyme Inhibitors/pharmacology
- Kallikreins/biosynthesis
- Kallikreins/genetics
- Kininogen, High-Molecular-Weight/genetics
- Kininogen, Low-Molecular-Weight/genetics
- Kininogens/biosynthesis
- Kininogens/genetics
- Lipopolysaccharides/antagonists & inhibitors
- Lipopolysaccharides/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Polymerase Chain Reaction
- RNA, Messenger/analysis
- Rats
- Rats, Sprague-Dawley
- Tetradecanoylphorbol Acetate
Collapse
Affiliation(s)
- H Okamoto
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Ikawadani-cho, Nishi-ku, Kobe 651-2180, Japan.
| | | | | | | | | |
Collapse
|
27
|
Partovian C, Benetos A, Pommiès JP, Mischler W, Safar ME. Effects of a chronic high-salt diet on large artery structure: role of endogenous bradykinin. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:H1423-8. [PMID: 9612345 DOI: 10.1152/ajpheart.1998.274.5.h1423] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bradykinin activity could explain the blood pressure increase during NaCl loading in hypertensive animals, but its contribution on vascular structure was not evaluated. We determined cardiac mass and large artery structure after a chronic, 4-mo, high-salt diet in combination with bradykinin B2-receptor blockade by Hoe-140. Four-week-old rats were divided into eight groups according to strain [spontaneously hypertensive rats (SHR) vs. Wistar-Kyoto (WKY) rats], diet (0.4 vs. 7% NaCl), and treatment (Hoe-140 vs. placebo). In WKY rats, a high-salt diet significantly increased intra-arterial blood pressure with minor changes in arterial structure independently of Hoe-140. In SHR, blood pressure remained stable but 1) the high-salt diet was significantly associated with cardiovascular hypertrophy and increased arterial elastin and collagen, and 2) Hoe-140 alone induced carotid hypertrophy. A high-salt diet plus Hoe-140 acted synergistically on carotid hypertrophy and elastin content in SHR, suggesting that the role of endogenous bradykinin on arterial structure was amplified in the presence of a high-salt diet.
Collapse
Affiliation(s)
- C Partovian
- Department of Internal Medicine, Broussais Hospital, Paris, France
| | | | | | | | | |
Collapse
|
28
|
Sakakibara T, Hintze TH, Nasjletti A. Determinants of kinin release in isolated rat hindquarters. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:R120-5. [PMID: 9458907 DOI: 10.1152/ajpregu.1998.274.1.r120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We studied the determinants of kinin release into the venous effluent of rat hindquarters perfused with Krebs bicarbonate buffer. Kinin release in preparations perfused with control media (14.6 +/- 2.5-20.7 +/- 6.7 pg/15 min) was surpassed by that in preparations perfused with media containing kininase inhibitors (243 +/- 53 to 276 +/- 78 pg/15 min). Kinin release increased when purified kininogen (from 242 +/- 43 to 3,365 +/- 725 pg/15 min) or kallikrein (from 270 +/- 49 to 30,649 +/- 8,040 pg/15 min) was added to the perfusate. Conversely, kinin release fell when the kallikrein inhibitor aprotinin (from 272 +/- 58 to 122 +/- 27 pg/15 min) or soybean trypsin inhibitor (from 273 +/- 52 to 195 +/- 25 pg/15 min) was added. Both basal and kininogen-induced kinin release were attenuated in preparations perfused with media containing cycloheximide, a protein synthesis inhibitor, but kallikrein-induced kinin release was not. These data suggest that kinin release from perfused rat hindquarters reflects the activity of both the kinin-degrading and kinin-generating pathways and that the latter is sustained by a kallikrein manufactured de novo and by preexistent kininogen(s).
Collapse
Affiliation(s)
- T Sakakibara
- Department of Physiology, New York Medical College, Valhalla 10595, USA
| | | | | |
Collapse
|
29
|
Abstract
The kallikrein-kinin system (KKS) plays an important role in the regulation of renal function. Endogenous kinins modulate renal microvascular resistance, medullary blood flow, and distal nephron sodium and water reabsorption. All the components of the KKS, including tissue kallikrein, kininogen, kininase II, and kinin receptors are expressed within the kidney, establishing a paracrine system capable of controlling local nephron functions. In this review, data will be presented demonstrating that the developing kidney expresses an endogenous, functionally active KKS. Molecular studies have shown that gene expression of the renal KKS in the rat is activated postnatally, and that the intrarenal distribution of KKS components is subject to developmental control. Furthermore, the developmental expression of KKS appears to be regulated primarily at the transcriptional level. Ontogenetic studies have also revealed that the bradykinin B-2 receptor gene is overexpressed in the developing rat kidney. As kinins are potent vasoactive and growth-promoting factors, it is proposed that endogenous kinins mediate developmental renal growth and differentiation, and modulate the maturational changes which occur in renal hemodynamics.
Collapse
Affiliation(s)
- S S el-Dahr
- Tulane University School of Medicine, Department of Pediatrics, New Orleans, Louisiana 70112, USA
| |
Collapse
|
30
|
Abstract
Angiotensin converting-enzyme (ACE) inhibitors attenuate cardiac hypertrophy and prolong survival in animal models and patients after myocardial infarction (MI). Considering the dual function of the ACE, the therapeutic efficacy of ACE inhibitors after MI implicates the renin-angiotensin system and/or the kallikrein-kinin system in the pathophysiology of postinfarction cardiac remodeling. We evaluated the role of kinins, and their potential contribution to the antiremodeling effects of ACE inhibition in this setting. Rats underwent coronary artery ligation followed by chronic B2 kinin receptor blockade with icatibant (HOE 140). Additional groups of MI rats were treated with the ACE inhibitor lisinopril, alone or in combination with icatibant. B2 kinin receptor blockade enhanced the deposition of collagen (morphometric analysis) in the left ventricular interstitial space after MI, whereas markers of cardiomyocyte hypertrophy (left ventricular weights and prepro-atrial natriuretic factor [ANF] expression) were not affected. Chronic ACE inhibition reduced collagen deposition and cardiomyocyte hypertrophy after MI. The inhibitory action of ACE inhibition on interstitial collagen was partially reversed by B2 kinin receptor blockade. However, B2 kinin receptor blockade did not attenuate the effects of ACE inhibition on cardiomyocyte hypertrophy. In conclusion, kinins inhibit the interstitial accumulation of collagen, but do not modulate cardiomyocyte hypertrophy after MI. Kinins contribute to the reduction of myocardial collagen accumulation by ACE inhibition; however, the effects of ACE inhibition on cardiomyocyte hypertrophy are related to reduced generation of angiotensin II.
Collapse
Affiliation(s)
- K C Wollert
- Abteilung Kardiologie, Medizinische Hochschule Hannover, Germany
| | | |
Collapse
|
31
|
Dixon BS, Dennis MJ. Regulation of mitogenesis by kinins in arterial smooth muscle cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:C7-20. [PMID: 9252437 DOI: 10.1152/ajpcell.1997.273.1.c7] [Citation(s) in RCA: 156] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recent evidence suggests that bradykinin (BK) plays a role in regulating neointimal formation after vascular injury. The present study examined the mechanism whereby BK regulates platelet-derived growth factor (PDGF) AB-induced mitogenesis in smooth muscle cells from rat mesenteric artery. BK, but not other activators of phosphoinositidase C (e.g., angiotensin II), inhibited PDGF-stimulated mitogenesis. The B1 receptor agonist des-Arg9-BK (DABK) was more potent than the B2 agonist BK; smaller BK fragments had no activity. In studies in which the B2 receptor antagonist HOE-140 {D-Arg0[Hyp3,beta-(2-thienyl)-Ala5,D-Tic7,Oic 8]BK} and the B1 receptor antagonist DHOE [[D-Arg0,Hyp3,beta-(2-thienyl)-Ala5,D-Tic7,Oi c8,des-Arg9]BK] were used, both receptors independently mediated inhibition of PDGF-induced mitogenesis. There was no evidence for metabolism of BK to DABK. The rank potency for activating phosphoinositidase C and increasing intracellular Ca2+ (BK > DABK) was opposite that for inhibiting mitogenesis (DABK > BK). Inhibition of cyclooxygenase did not prevent the kinin-mediated inhibition. Kinetic analysis of the cell cycle effects of kinins on PDGF-stimulated mitogenesis revealed that continuous exposure to DABK or BK was inhibitory even when added shortly before the cells initiated DNA synthesis (S phase). However, short-term exposure (5-60 min) to DABK or BK was inhibitory only when added after exposure to PDGF. These data suggest that the B1 and B2 receptors potently inhibited PDGF-stimulated mitogenesis and proliferation by activating an alternative signal transduction cascade not involving phosphoinositidase C or prostaglandins. The inhibition occurred at a point late in progression of the cell cycle from G1 to S and was dependent on the presence of kinins after exposure to PDGF.
Collapse
MESH Headings
- Animals
- Bradykinin/analogs & derivatives
- Bradykinin/pharmacology
- Bradykinin Receptor Antagonists
- Carotid Arteries/cytology
- Carotid Arteries/drug effects
- Carotid Arteries/physiology
- Cell Division/drug effects
- Cells, Cultured
- Cyclooxygenase Inhibitors/pharmacology
- Enzyme Activation
- In Vitro Techniques
- Mesenteric Arteries/cytology
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiology
- Mitosis/drug effects
- Muscle Contraction/drug effects
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Phosphoric Diester Hydrolases/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Rats
- Rats, Wistar
- Receptor, Bradykinin B1
- Receptor, Bradykinin B2
- Receptors, Bradykinin/agonists
Collapse
Affiliation(s)
- B S Dixon
- Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | | |
Collapse
|
32
|
Raidoo DM, Ramsaroop R, Naidoo S, Müller-Esterl W, Bhoola KD. Kinin receptors in human vascular tissue: their role in atheromatous disease. IMMUNOPHARMACOLOGY 1997; 36:153-60. [PMID: 9228540 DOI: 10.1016/s0162-3109(97)00015-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Using samples of many human blood vessels, obtained at autopsy and specific antibodies directed to peptide sequences of the kinin B1 and B2 receptors, we demonstrate the localisation of these receptors within the human vascular system using standard immunolabelling techniques. In large elastic arteries and veins, kinin receptors are present only in the endothelial cells whereas in all muscular arteries and arterioles, these receptors are present in both the endothelial and smooth muscle cells. The identification of kinin receptors in human blood vessels confirms that kinins may modulate both vascular permeability and contractility. The incidental finding at histology, of patchy atheromatous disease in the coronary, femoral, vertebral and pericallosal arteries, assisted in elucidating the role of these receptors in the commonest disease affecting human blood vessels. Intense labelling for B1 receptors was observed in the endothelial cells, foamy macrophages, inflammatory cells and fibroblasts within the thickened intima of the plaque as well as in smooth muscle cells of the underlying tunica media. Immunoreactive B2 receptors were also observed in these cells but with reduced intensity. The intense immunolabelling of B1 receptors in these regions suggest that these may be induced by atheromatous disease and may have therapeutic importance for the B1 receptor antagonists.
Collapse
Affiliation(s)
- D M Raidoo
- Department of Physiology, University of Natal Medical School, South Africa
| | | | | | | | | |
Collapse
|
33
|
Nolly H, Miatello R, Damiani MT, Abate CD. Possible protective effects of kinins and converting enzyme inhibitors in cardiovascular tissues. IMMUNOPHARMACOLOGY 1997; 36:185-91. [PMID: 9228545 DOI: 10.1016/s0162-3109(97)00020-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The main objective of this study was to determine if the components of the kallikrein-kinin system are released into the venous effluent from isolated perfused rat hearts. To assess the contribution of kinins and the vascular and cardioprotective effects of the ACE inhibitor ramipril, we determined the status of cardiac kallikrein (CKK), potent kinin-generating enzyme, in rats with right ventricular hypertrophy induced by chronic volume overload and left ventricular hypertrophy by aortic banding. CKK was measured as previously described (Nolly, H.L., Carbini, L., Carretero, O.A., Scicli, A.G., 1994). Kininogen by a modification of the technique of Dinitz and Carvalho (1963) and kinins were extracted with a Sep-Pak C18 cartridge and measured by RIA. CKK (169 +/- 9 pg Bk/30 min), kininogen (670 +/- 45 pg Bk/30 min) and immunoreactive kinins (62 +/- 10 pg Bk/30 min) were released into the perfusate. The release was almost constant over a 120 min period. Pretreatment with the protein synthesis inhibitor puromycin (10 mg i.p.) lowered the release of kallikrein (42 +/- 12 pg Bk/30 min, p < 0.001) and kininogen (128 +/- 56 pg Bk/30 min, p < 0.001). Addition of ramiprilat (10 micrograms/ml) increased kinin release from 54 +/- 18 to 204 +/- 76 pg Bk/30 min (p < 0.001). Aortic banding of rats increased their blood pressure (BP) (p < 0.001), relative heart weight (RHW) (p < 0.001) and CKK (p < 0.001). Ramipril treatment induced a reduction in BP (p < 0.05) and RHW (p < 0.005) while CKK remained elevated. Aortocaval shunts increased their ANF plasma levels (p < 0.05), RHW (p < 0.001) and CKK (p < 0.01). Ramipril treatment induced a reduction in RHW (p < 0.05), while CKK and ANF increased significantly (p < 0.05). The present data show that the components of the kallikrein-kinin system are continuously formed in the isolated rat heart and that ramipril reduces bradykinin breakdown with subsequent increase in bradykinin outflow. The experiments with aorta caval shunt and aortic banding show that cardiac tissues increase their kinin-generating activity and this was even higher in ramipril-treated animals. This may suggest that the actual level of kinins is finely tuned to the local metabolic demands. In this experimental model of cardiac hypertrophy. ACE inhibitors potentiate the actions of kinins and probably try to normalise endothelial cell function.
Collapse
Affiliation(s)
- H Nolly
- Laboratory of Experimental Hypertension and Vasoactive Substances, School of Medicine and National Council of Research, Mendoza, Argentina
| | | | | | | |
Collapse
|
34
|
Kuo JJ, Zimmerman BG. Increased kallikrein activity in the rabbit renal vasculature during low sodium intake. Clin Exp Pharmacol Physiol 1997; 24:153-6. [PMID: 9075588 DOI: 10.1111/j.1440-1681.1997.tb01799.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
1. Vascular tissue has been shown to possess a kallikrein--kinin system that may participate in the kinin-mediated increase in renal sodium excretion. As sodium deprivation has been demonstrated to increase kallikrein content in the kidney and urine we hypothesized that during low sodium intake, kallikrein should increase in the renal vasculature. 2. Kininogenase activity, reflecting kallikrein enzymatic content, was measured in a homogenate of a microdissected intrarenal arterial network (IAN) from the rabbit kidney. Kininogenase activity was determined in rabbits on a normal sodium (n = 14) or sodium-restricted (n = 9) diet. 3. Total kininogenase activity in rabbits on a normal sodium diet was 15.0 +/- 2.7 pg kinin/mg per 30 min, while it was much higher in rabbits on a sodium-restricted diet (90.7 +/- 16.5 pg kinin/mg per 30 min). Specific tissue kallikrein activity was measured by comparing the difference in kininogenase activity in homogenates treated with soybean-trypsin inhibitor (SBTI) compared with homogenates treated with SBTI and aprotinin. This difference was much larger in the sodium-restricted rabbits than in rabbits on a normal sodium diet (29.5 +/- 3.8 vs 5.1 +/- 1.7 pg kinin/mg per 30 min, respectively). 4. We conclude that the rabbit IAN produces kallikrein, which is markedly increased in response to sodium restriction. Increased kinins during sodium restriction may modulate the pressor and anti-natriuretic systems activated during negative sodium balance.
Collapse
Affiliation(s)
- J J Kuo
- Department of Pharmacology, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
35
|
Abstract
BACKGROUND A variety of immunologic abnormalities have been described in systemic and experimental lupus erythematosus (LE). Several T-cell defects, especially in helper T (Th) cell cytokines, have been reported. OBJECTIVE Our purpose was to identify the Th cytokine profile in cutaneous LE. METHOD Total RNA was extracted from punch biopsy specimens from 19 patients with cutaneous LE (nine, discoid LE; two, subacute cutaneous LE; and eight, systemic LE) and from four healthy control subjects. RNA was reverse transcribed into complementary DNA and amplified with polymerase chain reaction (PCR) primers specific for interleukin-2 (IL-2), IL-4, IL-5, IL-10, interferon gamma (IFN-gamma), and beta actin. PCR products were detected by agarose gel electrophoresis and Southern blot with 32P-labeled, nested probes. RESULTS Sixteen of 19 cutaneous LE specimens lacked IL-2, all were negative for IL-4, and 10 of 19 had detectable IL-10, whereas IFN-gamma and IL-5 messenger RNAs were present in the majority of LE specimens. IFN-gamma and IL-10 mRNAs were found in all normal skin controls, whereas IL-2, IL-4, and IL-5 mRNAs were undetectable. Functional IFN-gamma protein was evidenced by intercellular adhesion molecule-1 and HLA-DR staining of keratinocytes in nine of nine LE specimens but not in normal skin. The pattern of cytokine mRNAs, intercellular adhesion molecule-1, and/or HLA-DR expression in cutaneous LE specimens did not vary with different subtypes of LE, antinuclear antibody titer, or the magnitude of inflammation. CONCLUSION The presence of IL-5 mRNA in cutaneous LE specimens suggests that Th type 2 cells combine with local IFN-gamma production to augment disease and may be related to the pathophysiology of cutaneous LE.
Collapse
Affiliation(s)
- L F Stein
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | |
Collapse
|
36
|
Holte HR, Bjørnstad-Ostensen A, Berg T. The role of endogenous bradykinin in blood pressure homeostasis in spontaneously hypertensive rats. Br J Pharmacol 1996; 118:1925-30. [PMID: 8864525 PMCID: PMC1909879 DOI: 10.1111/j.1476-5381.1996.tb15626.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1. The role of endogenous bradykinin in mean arterial blood pressure (BP) homeostasis was studied in spontaneously hypertensive (SHR) and normotensive (WKY) rats by the use of a bradykinin B2-receptor antagonist (BKant; Hoe 140, 11.6 micrograms kg-1) and converting enzyme (kininase II) inhibitor (captopril, 10 mg). To obtain a response to captopril that was induced through inhibition of kinin-degradation only and not through inhibition of angiotensin II-formation, the studies were performed on binephrectomized male rats to eliminate the renin-angiotensin system. 2. The role of the nitric oxide (NO) and the adrenergic systems were evaluated by the use of NO-synthase inhibitor (L-NAME, 0.3 g kg-1) and phentolamine (2 mg kg-1), respectively. 3. The rats were anaesthetized and pretreated with two injections of vehicle (PBS) or drugs spaced 5 min apart: PBS + PBS; BKant + PBS; PBS + L-NAME; BKant + L-NAME; or phentolamine + L-NAME. All rats were given captopril 15 min later. Time-control groups were treated with L-NAME but not captopril. 4. In WKY rats, captopril did not significantly alter BP in any of the groups. In the SHR-PBS + PBS group, on the other hand, captopril induced an immediate fall in BP (delta BP = -23 +/- 4 mmHg, P < 0.0017) which was completely blocked by BKant (delta BP = 2 +/- 2 mmHg) (P < 0.0011). L-NAME did not significantly alter the immediate hypotensive response to captopril but disclosed a later hypertensive reaction. In L-NAME + BKant-treated rats, both the hypotensive response and the late hypertension was abolished. In rats treated with phentolamine + L-NAME, the immediate fall in BP was not different from the controls whereas the late hypertension was absent. 5. BKant itself had no effect on basal BP in either WKY or SHR even when a 10 times higher dose was tested in a separate set of experiments. This was true also for conscious, nonnephrectomized SHR rats. 6. It was concluded that endogenous production of bradykinin was demonstrable through kininase II-inhibition in hypertensive but not in normotensive rats. However, this endogenous bradykinin did not play a role in basal BP homeostasis. The captopril-induced hypotension depended on kinin but, under the present conditions, not on NO as a mediator. The fall in BP induced a compensatory adrenergic hypertensive response which was revealed when the continuous NO-synthesis was blocked by L-NAME.
Collapse
Affiliation(s)
- H R Holte
- Department of Physiology, University of Oslo, Norway
| | | | | |
Collapse
|
37
|
MacDonald RJ, Southard-Smith EM, Kroon E. Disparate tissue-specific expression of members of the tissue kallikrein multigene family of the rat. J Biol Chem 1996; 271:13684-90. [PMID: 8662704 DOI: 10.1074/jbc.271.23.13684] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
To understand the regulatory diversity of the rat family of linked kallikrein genes, we have assayed the expression of family members in 20 major organs. Reverse transcription-polymerase chain reaction analysis using primers and hybridization probes specific for each of the 10 expressed kallikrein genes showed that no two family members share the same organ-specific pattern of expression. The only common site of expression for all 10 known active genes is the submandibular gland. The presence of the mRNA for at least one family member is detected in 19 of these 20 organs (liver excepted), from as few as three organs to as many as 18 for individual family members. For individual genes there can be more than a 10(5)-fold variation in mRNA levels among organs, from a limit of detection of slightly less than 1 mRNA molecule/10 cells to more than 10,000 mRNA molecules/cell. Despite high sequence conservation and close linkage, the members of this family are expressed in very different and complex patterns. A gradient of diversity of expression corresponds to the order of the genes within the kallikrein family locus.
Collapse
Affiliation(s)
- R J MacDonald
- Department of Biochemistry, Molecular Immunology Center, the University of Texas Southwestern Medical Center, Dallas, Texas 75235-9140, USA
| | | | | |
Collapse
|
38
|
Dixon BS, Dennis M. Interaction between growth factors and kinins in arterial smooth muscle cells. IMMUNOPHARMACOLOGY 1996; 33:16-23. [PMID: 8856109 DOI: 10.1016/0162-3109(96)00076-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Bradykinin receptors are present on vascular smooth muscle cells; however, the regulation and biological function of these receptors is unclear. To address these questions the interaction between growth factors and kinins in cultured arterial smooth muscle cells has been examined. Based upon the data a hypothesis is presented that platelet-derived growth factor (PDGF) upregulates cell surface bradykinin B2 receptors on arterial smooth muscle cells. The biological effect of the increase in B2 receptors is currently unclear but under certain conditions they may enhance mitogenesis. These mitogenic effects however, are strongly opposed by the effects of bradykinin acting via a B1-type of receptor which mediates potent inhibition of growth factor-induced mitogenesis.
Collapse
MESH Headings
- Animals
- Bradykinin/pharmacology
- Cell Division/drug effects
- Cells, Cultured
- Growth Substances/pharmacology
- Kinins/metabolism
- Kinins/pharmacology
- Models, Biological
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Rats
- Receptor, Bradykinin B2
- Receptors, Bradykinin/drug effects
- Receptors, Bradykinin/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- B S Dixon
- Department of Medicine, University of Iowa College of Medicine, Iowa City, USA
| | | |
Collapse
|
39
|
Chao J, Jin L, Chen LM, Chen VC, Chao L. Systemic and portal vein delivery of human kallikrein gene reduces blood pressure in hypertensive rats. Hum Gene Ther 1996; 7:901-11. [PMID: 8727504 DOI: 10.1089/hum.1996.7.8-901] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
There is an inverse correlation between systemic blood pressure and urinary kallikrein levels in humans and hypertensive animal models, suggesting that the tissue kallikrein-kinin system plays an important role in blood pressure regulation. In this study, we explored the potential of human kallikrein gene delivery on blood pressure reduction in spontaneously hypertensive rats (SHR). The human tissue kallikrein gene or cDNA was placed under the control of following promoters: the metallothionein gene metal response-element (MRE-pHK), albumin gene (ALB-pHK), Rous sarcoma virus 3' long terminal repeat (LTR) (RSV-cHK), and cytomegalovirus (CMV-cHK). A single injection of these kallikrein DNAs results in a significant reduction of blood pressure in SHR, which lasts for 5-6 weeks. Systemic delivery of CMV-cHK, RSV-cHK, and MRE-pHK has a greater effect on blood pressure reduction than ALB-pHK, whereas intraportal vein gene delivery of ALB-pHK is more effective than the other kallikrein DNA constructs. The degree of blood pressure reduction depends on the amount of administered DNA and the age of the animals. Reduction of blood pressure was observed in adult, but not young, SHR. The expression of human tissue kallikrein in rats was identified by an ELISA that is specific for human tissue kallikrein. No antibodies to either human tissue kallikrein or its DNA were detected in rat sera after somatic gene delivery. These results show that somatic gene delivery of human tissue kallikrein causes a lowering effect of systolic blood pressure in genetically hypertensive rats and provide valuable information for kallikrein gene therapy in the treatment of hypertension.
Collapse
Affiliation(s)
- J Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston 29425-2211, USA
| | | | | | | | | |
Collapse
|
40
|
Deperthes D, Gauthier ER, Chapdelaine P, Lazure C, Tremblay RR, Dubé JY. Identification of glandular kallikrein in dog pancreas and determination of its tissue distribution. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1243:291-4. [PMID: 7727502 DOI: 10.1016/0304-4165(94)00128-k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In order to establish a formal link between previously purified canine urinary kallikrein and dog pancreatic kallikrein whose cDNA sequence has recently been published, we have isolated the pancreatic kallikrein from that animal species. Pancreatic cytosol proteins were sequentially subjected to chromatography on DEAE-Sepharose CL-6B and Concanavalin A-Sepharose, to an autolysis step and finally to two-dimensional gel electrophoresis. Kallikrein immunoreactive spots were identified with an antibody directed against canine urinary kallikrein. These proteins were isolated after electroblotting and the amino acid sequence of their NH2-terminal portion was determined by microsequencing. The sequence was found to be identical to the one deduced from pancreatic kallikrein cDNA. Using the same antibody and immunohistochemical procedures, kallikrein was found to be present in the pancreas, the salivary glands, the kidney, the colon, the lungs and the testis. These results thus confirm the molecular nature of a glandular kallikrein in the canine species.
Collapse
Affiliation(s)
- D Deperthes
- Laboratory of Hormonal Bioregulation, CHUL Research Center, Sainte-Foy, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
41
|
Wang C, Chao L, Chao J. Direct gene delivery of human tissue kallikrein reduces blood pressure in spontaneously hypertensive rats. J Clin Invest 1995; 95:1710-6. [PMID: 7535795 PMCID: PMC295685 DOI: 10.1172/jci117847] [Citation(s) in RCA: 111] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Hypertension is a multigene and multifactorial disorder affecting approximately 25% of the population. To demonstrate potential therapeutic effects of human tissue kallikrein in hypertension, spontaneously hypertensive rats were subjected to somatic gene therapy. Two human tissue kallikrein DNA constructs, one under the promoter control of the metallothionein metal response element and the other under the control of the Rous sarcoma virus 3'-LTR, were generated. We delivered naked DNA constructs into spontaneously hypertensive rats via intravenous injection. The expression of human tissue kallikrein in rats was identified in the heart, lung, and kidney by reverse transcription polymerase chain reaction followed by Southern blot analysis and an ELISA specific for human tissue kallikrein. A single injection of both human kallikrein plasmid DNA constructs caused a sustained reduction of blood pressure which began 1 wk after injection and continued for 6 wk. A maximal effect of blood pressure reduction of 46 mmHg in rats was observed 2-3 wk after injection with kallikrein DNA as compared to rats with vector DNA (n = 6, P < 0.05). The hypotensive effect caused by somatic gene delivery of human tissue kallikrein in hypertensive rats is reversed by subcutaneous injection of aprotinin, a potent tissue kallikrein inhibitor. No antibodies to either human tissue kallikrein or kallikrein DNA were detected in rat sera after injection of the human kallikrein gene. These results show that direct gene delivery of human tissue kallikrein causes a sustained reduction in systolic blood pressure in genetically hypertensive rats and indicate that the feasibility of kallikrein gene therapy for treating human hypertension should be studied.
Collapse
Affiliation(s)
- C Wang
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston 29425-2211, USA
| | | | | |
Collapse
|
42
|
Abstract
Kinins are endothelium-dependent vasodilators and natriuretic paracrine peptides that participate in the regulation of blood pressure, renal hemodynamics and sodium excretion. Several lines of evidence suggest an important role for intrarenal kinins and their receptors in kidney growth and development. (1) The developing rat kidney expresses all the components of the tissue kallikrein-kinin system: tissue kallikrein, low molecular weight (LMW) kininogen, kininase II and kinin receptors. Also, the developing liver expresses high molecular weight and LMW kininogens. Thus, a complete kinin-generating system exists in the developing kidney. (2) Gene transcription, mRNA and protein abundance, and enzymatic activity of renal kallikrein are all markedly up-regulated during postnatal kidney growth, and a positive correlation exists between renal kallikrein synthesis and the maturational rise in renal blood flow. (3) Rat glomerular mesangial cells in culture express the kinin receptors and proliferate in response to bradykinin, suggesting that endogenous kinins and their receptors modulate glomerular growth. (4) The newborn period is characterized by an activation of kinin receptor gene expression, and chronic pharmacological blockade of kinin receptors suppresses DNA synthesis in the developing but not adult kidney. Collectively, these data provide the basis for the hypothesis that endogenous kinins and the kinin receptors play an important role in the developmental biology of the metanephric kidney.
Collapse
Affiliation(s)
- S S el-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana 70112
| |
Collapse
|
43
|
Morgan K. Diverse factors influencing angiotensin metabolism during ACE inhibition: insights from molecular biology and genetic studies. Heart 1994; 72:S3-10. [PMID: 7946800 PMCID: PMC1025586 DOI: 10.1136/hrt.72.3_suppl.s3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- K Morgan
- Department of Cardiology, Royal Postgraduate Medical School, Hammersmith Hospital, London
| |
Collapse
|
44
|
Graf K, Gräfe M, Auch-Schwelk W, Baumgarten CR, Scheffer H, Hildebrandt A, Fleck E. Tissue kallikrein activity and kinin release in human endothelial cells. EUROPEAN JOURNAL OF CLINICAL CHEMISTRY AND CLINICAL BIOCHEMISTRY : JOURNAL OF THE FORUM OF EUROPEAN CLINICAL CHEMISTRY SOCIETIES 1994; 32:495-500. [PMID: 7981328 DOI: 10.1515/cclm.1994.32.7.495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The kininogenase, tissue kallikrein (EC 3.4.21.8), has been identified in different blood vessels. The enzyme was mainly found in vascular smooth muscle cells. It is not known whether it is present and functionally active in vascular endothelial cells. The following study investigates the presence of tissue kallikrein in endothelial cells from human umbilical veins and pulmonary arteries. Tissue kallikrein was demonstrated in three ways: 1) by immunostaining in endothelial cells; 2) by measurement of tissue kallikrein activity using a colorimetric assay; 3) by the measurement of kinin release in intact and homogenised endothelial cells with a radioimmunoassay. Immunostaining demonstrated the presence of tissue kallikrein in endothelial cells from human umbilical veins and endothelial cells from human pulmonary arteries. Tissue kallikrein-like activity, measured by the degradation of D-Val-cyclohexyl-Ala-Arg-4-nitraniline, was 3.57 +/- 0.5 mU/10(6) endothelial cells from human umbilical veins and 7.52 +/- 0.84 mU/10(6) endothelial cells from human pulmonary arteries. Intracellular kinin concentrations were 424 +/- 83 pg/10(6) cells in endothelial cells from human umbilical veins and 576 +/- 146 pg/10(6) cells in endothelial cells from human pulmonary arteries, and they increased in a time-dependent manner after homogenisation. The increase was abolished by aprotinin (1000 kIU), an inhibitor of tissue kallikrein in both cell types. Addition of exogenous kallikrein (5 mU) to homogenised cells led to a five fold increase of kinin concentrations after five minutes, indicating a sufficient resource of cellular kininogen. Removal of extracellularly bound kininogen by washing with dextran sulphate (100 mg/l) resulted in an approximately 75% reduction of the cellular kinin release.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- K Graf
- Department of Internal Medicine and Cardiology, German Heart Institute, Berlin
| | | | | | | | | | | | | |
Collapse
|
45
|
Saed G, Fivenson DP, Naidu Y, Nickoloff BJ. Mycosis fungoides exhibits a Th1-type cell-mediated cytokine profile whereas Sezary syndrome expresses a Th2-type profile. J Invest Dermatol 1994; 103:29-33. [PMID: 8027577 DOI: 10.1111/1523-1747.ep12388985] [Citation(s) in RCA: 134] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We determined T-cell cytokine profiles in the epidermis, dermis, and blood of cutaneous T-cell lymphoma to differentiate whether unique cytokine profiles were associated with mycosis fungoides (MF) versus Sezary syndrome. Punch biopsy specimens from plaque stage MF (n = 7) were compared to Sezary skin (n = 3) after undergoing rapid heat-saline separation of epidermis from dermis. Normal adult skin (n = 11), neonatal foreskin (n = 4), untreated psoriatic plaques (n = 6), and normal donor peripheral blood leukocytes (n = 3) were studied as controls. Total RNA was extracted from all skin specimens, as well as peripheral blood leukocytes from MF (n = 3) and Sezary patients (n = 7), and was converted to cDNA by reverse transcriptase. Polymerase chain reaction amplification of cDNAs using interleukin 2 (IL-2), IL-4, IL-5, IL-10, and interferon gamma-specific primers was used to differentiate Th1-type responses (IL-2+ and interferon gamma +) from Th2-type responses (IL-4+, IL-5+, and IL-10+). beta-actin specific primers were included as a positive control for mRNA integrity. All MF specimens contained mRNAs for IL-2 and interferon gamma limited to epidermis but not IL-4, IL-5, or IL-10. In contrast, Sezary skin and blood showed a cytokine mRNA pattern dominated by IL-4, IL-5, and IL-10. MF blood showed a pattern similar to normal peripheral blood T cells with mixed detection of all T-helper cell cytokine mRNAs. All psoriasis samples contained mRNAs for IL-2 and interferon gamma in both epidermis and dermis with no IL-4 or IL-10 in either compartment. These findings demonstrate that the cutaneous lesions of MF are characterized by an epidermal Th1-type cytokine profile, whereas both the blood and skin of patients with Sezary syndrome is characterized by a Th2-type profile. This work suggests that differences in cytokine production may be related to the pathophysiology and clinical presentation in cutaneous T-cell lymphoma.
Collapse
Affiliation(s)
- G Saed
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202
| | | | | | | |
Collapse
|
46
|
Dixon BS. Cyclic AMP selectively enhances bradykinin receptor synthesis and expression in cultured arterial smooth muscle. Inhibition of angiotensin II and vasopressin response. J Clin Invest 1994; 93:2535-44. [PMID: 8200990 PMCID: PMC294477 DOI: 10.1172/jci117264] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Bradykinin receptors on vascular smooth muscle may play an important role in regulating the endogenous effects of the vascular kallikrein-kinin system. The present study examined the effect of cyclic nucleotides on bradykinin-stimulated responses in cultured arterial smooth muscle cells. Short term stimulation (1 min) with cyclic AMP produced a variable inhibition of bradykinin-stimulated calcium mobilization which was lost in later passaged cells. However, long-term stimulation (24 h) produced a consistent increase in bradykinin-stimulated calcium mobilization in both early and late passaged cells. Further analysis demonstrated that chronic exposure to cAMP produced a twofold increase in both the number of cell surface bradykinin receptors and in bradykinin-stimulated phosphoinositide hydrolysis. The increase in bradykinin receptors was time dependent (> 7 h) and blocked by protein synthesis inhibitors, suggesting that cAMP enhanced the synthesis of new bradykinin receptors. The increase in bradykinin receptor binding and calcium mobilization was also stimulated by cholera toxin, forskolin, and isobutylmethylxanthine, but not isoproterenol or prostaglandin E2. Of considerable interest, prolonged exposure to cAMP inhibited both angiotensin II and arginine vasopressin-stimulated phosphoinositide hydrolysis and intracellular calcium mobilization. In summary, prolonged treatment with cAMP selectively stimulates the synthesis and expression of bradykinin receptors on arterial smooth muscle while decreasing the responsiveness to vasoconstrictor agonists such as angiotensin II and vasopressin.
Collapse
Affiliation(s)
- B S Dixon
- Department of Medicine, University of Iowa College of Medicine, Iowa City 52242
| |
Collapse
|
47
|
Fivenson DP, Saed GM, Beck ER, Dunstan RW, Moore PF. T-cell receptor gene rearrangement in canine mycosis fungoides: further support for a canine model of cutaneous T-cell lymphoma. J Invest Dermatol 1994; 102:227-30. [PMID: 7906287 DOI: 10.1111/1523-1747.ep12371767] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Canine cutaneous T-cell lymphoma (CTCL) is a morphologic and immunophenotypic simulant of human mycosis fungoides (MF) characterized by an infiltrate of atypical, hyperconvoluted, epidermotropic T cells. To further support our hypothesis that canine MF is a useful model for the study of human CTCL, we have used Southern blotting to search for clonal T-cell proliferations in canine MF. Cellular DNA was extracted from normal dog buffy coat cells (n = 8), lesional canine MF skin (n = 8), canine MF buffy coat cells (n = 7), normal dog skin (n = 3), and normal human buffy coat cells (n = 5), digested with a panel of restriction enzymes and Southern blotted onto nylon membranes. All cases of canine MF were also immunophenotyped with anti-canine monoclonal antibodies to CD4, CD8, CD18, CD45RA, canine class II, T-cell activation antigens, and pan-B-cell antigens. Normal dogs gave reproducible digestion patterns in blood and skin, which differed from the human germline patterns when probed with a human T-cell receptor (TCR), beta chain constant region (C beta) cDNA. Common germline bands between the species included the 3.5-kb Eco RI, 3.4-kb Bam HI, 5.4-kb Sac I. These results confirmed that the TCR-beta gene is evolutionarily conserved between dog and man. Immunostaining revealed that 3/7 cases were CD4+ canine CTCL and 4/7 were CD8+ canine CTCL. Rearranged bands, deletion of germline bands, as well as minor alterations in electrophoretic mobility were observed in lesional DNA from seven of eight cases of canine MF, with at least two restriction digests in each case. Dog rearrangements were best detected with Bgl II, Eco RI, Eco RV, and Sac I, whereas deletions were detected with Bgl II, Sac I, Eco RV, and Bam HI. These studies demonstrate the presence of clonal TCR rearrangement in canine MF, further supporting the similarity of this tumor to human MF and its role as an animal model of CTCL.
Collapse
MESH Headings
- Animals
- Antigens, CD/analysis
- Biological Evolution
- Blotting, Southern
- CD18 Antigens
- CD4 Antigens/analysis
- CD8 Antigens/analysis
- DNA, Neoplasm/analysis
- DNA, Neoplasm/genetics
- Disease Models, Animal
- Dog Diseases/genetics
- Dogs
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics
- Humans
- Immunophenotyping
- Leukocyte Common Antigens/analysis
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/pathology
- Lymphoma, T-Cell, Cutaneous/veterinary
- Mycosis Fungoides/genetics
- Mycosis Fungoides/veterinary
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Skin Neoplasms/veterinary
- T-Lymphocytes/chemistry
- T-Lymphocytes/pathology
- T-Lymphocytes/ultrastructure
Collapse
Affiliation(s)
- D P Fivenson
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202
| | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- J A Clements
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
49
|
Abstract
Kinins are vasoactive paracrine peptides which participate in a wide range of functions, including the regulation of local organ blood flow, systemic blood pressure, transepithelial water and electrolyte transport, cellular growth, capillary permeability and inflammatory response, and pain. The recent introduction of specific bradykinin receptor subtype antagonists has greatly advanced our understanding of the role of the kallikrein-kinin system (KKS) in various physiological and disease states. However, a major gap remains in our knowledge of the role of kinins in early development. In this review, evidence is presented that the developing nephron expresses both tissue kallikrein and kininogen, and that the genes encoding the components of the KKS are subject to considerable developmental regulation. The activity of the intrarenal kinin-generating system is lowest in the developing kidney and increases with age. Completion of nephrogenesis is characterized by a marked surge in intrarenal kallikrein synthesis and gene transcription. Maturation is associated with redistribution of intrarenal kallikrein and its messenger RNA from the inner to outer cortical nephrons following the centrifugal pattern of nephron development. Challenges for the future include delineation of the direct role of kinins in the maturation of renal functions and elucidation of the molecular mechanisms underlying the developmental expression of the KKS.
Collapse
Affiliation(s)
- S S el-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA 70112
| | | |
Collapse
|
50
|
Abstract
Kallikrein was identified in the adrenal glands of the rat. The enzyme was present in active and inactive forms (n = 9), since preincubation with trypsin increased kininogenase activity from 54.8 +/- 11.8 to 230 +/- 23 pg bradykinin per milligram protein per minute. Adrenal kininogenase activity was inhibited by 91% by phenylmethylsulfonyl fluoride (2 mM), 81% by D-Phe-Phe-Arg-chloromethyl ketone (1 microM), 88% by aprotinin (1,000 KIU), and only 16% by soybean trypsin inhibitor (50 microM). Preincubation with antibodies against rat urinary kallikrein resulted in over 90% inhibition of kininogenase activity. Immunoreactive glandular kallikrein was 30.7 +/- 4.8 ng/mg protein (n = 11). The apparent molecular weight of the adrenal kininogenase on gel filtration chromatography was 33,000 +/- 500 D. Both the adrenal enzyme and the purified submandibular gland kallikrein used as a control had the same mobility on alkaline polyacrylamide gel electrophoresis. To determine whether messenger RNA (mRNA) for glandular kallikrein is present in adrenal gland RNA, we used the polymerase chain reaction employing oligonucleotide primers and glandular kallikrein 32P complementary DNA (cDNA) as a probe, which should give a cDNA fragment of 370 bp. Southern blots of the amplified products revealed a fragment of the predicted size. In conclusion, glandular kallikrein has been identified in the adrenal glands. The presence of mRNA for glandular kallikrein suggests that kallikrein is synthesized locally in this tissue. This provides an anatomic basis for possible participation of a local kallikrein-kinin pathway in the regulation of adrenal function.
Collapse
Affiliation(s)
- H Nolly
- Henry Ford Hospital, Hypertension and Vascular Research Division, Detroit, MI 48202
| | | | | | | | | |
Collapse
|