1
|
Ahmad R, Chowdhury K, Kumar S, Irfan M, Reddy GS, Akter F, Jahan D, Haque M. Diabetes Mellitus: A Path to Amnesia, Personality, and Behavior Change. BIOLOGY 2022; 11:biology11030382. [PMID: 35336756 PMCID: PMC8945557 DOI: 10.3390/biology11030382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Diabetes Mellitus (DM) is a metabolic disorder resulting from a disturbance of insulin secretion, action, or both. Hyperglycemia and overproduction of superoxide induce the development and progression of chronic complications of DM. The impact of DM and its complication on the central nervous system (CNS) such as dementia and Alzheimer’s Disease (AD) still remain obscure. In dementia, there is a gradual decline in cognitive function. The incidence of dementia increases with age, and patient become socially, physically, and mentally more vulnerable and dependent. The symptoms often emerge decades after the onset of pathophysiology, thus impairing early therapeutic intervention. Most diabetic subjects who develop dementia are above the age of 65, but diabetes may also cause an increased risk of developing dementia before 65 years. Vascular dementia is the second most common form of dementia after AD. Type 2 DM (T2DM) increases the incidence of vascular dementia (since its covers the vascular system) and AD. The functional and structural integrity of the CNS is altered in T2DM due to increased synthesis of Aβ. Additionally, hyperphosphorylation of Tau protein also results from dysregulation of various signaling cascades in T2DM, thereby causing neuronal damage and AD. There is the prospect for development of a therapy that may help prevent or halt the progress of dementia resulting from T2DM. Abstract Type 2 diabetes mellitus is increasingly being associated with cognition dysfunction. Dementia, including vascular dementia and Alzheimer’s Disease, is being recognized as comorbidities of this metabolic disorder. The progressive hallmarks of this cognitive dysfunction include mild impairment of cognition and cognitive decline. Dementia and mild impairment of cognition appear primarily in older patients. Studies on risk factors, neuropathology, and brain imaging have provided important suggestions for mechanisms that lie behind the development of dementia. It is a significant challenge to understand the disease processes related to diabetes that affect the brain and lead to dementia development. The connection between diabetes mellitus and dysfunction of cognition has been observed in many human and animal studies that have noted that mechanisms related to diabetes mellitus are possibly responsible for aggravating cognitive dysfunction. This article attempts to narrate the possible association between Type 2 diabetes and dementia, reviewing studies that have noted this association in vascular dementia and Alzheimer’s Disease and helping to explain the potential mechanisms behind the disease process. A Google search for “Diabetes Mellitus and Dementia” was carried out. Search was also done for “Diabetes Mellitus”, “Vascular Dementia”, and “Alzheimer’s Disease”. The literature search was done using Google Scholar, Pubmed, Embase, ScienceDirect, and MEDLINE. Keeping in mind the increasing rate of Diabetes Mellitus, it is important to establish the Type 2 diabetes’ effect on the brain and diseases of neurodegeneration. This narrative review aims to build awareness regarding the different types of dementia and their relationship with diabetes.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka 1230, Bangladesh;
| | - Kona Chowdhury
- Department of Pediatrics, Gonoshasthaya Samaj Vittik Medical College and Hospital, Dhaka 1344, Bangladesh;
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, 907/A, Uvarsad Gandhinagar, Gujarat 382422, India;
| | - Mohammed Irfan
- Department of Forensics, Federal University of Pelotas, Pelotas 96020-010, RS, Brazil;
| | - Govindool Sharaschandra Reddy
- Department of Periodontics and Endodontics, School of Dental Medicine, University at Buffalo, Buffalo, NY 14214, USA;
| | - Farhana Akter
- Department of Endocrinology, Chittagong Medical College, Chattogram 4203, Bangladesh;
| | - Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, 111/1/A Distillery Road, Gandaria Beside Dhupkhola, Dhaka 1204, Bangladesh;
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur 57000, Malaysia
- Correspondence: or
| |
Collapse
|
2
|
Birch CA, Molinar-Inglis O, Trejo J. Subcellular hot spots of GPCR signaling promote vascular inflammation. ACTA ACUST UNITED AC 2020; 16:37-42. [PMID: 32838054 PMCID: PMC7431397 DOI: 10.1016/j.coemr.2020.07.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
G-coupled protein receptors (GPCRs) comprise the largest class of druggable targets. Signaling by GPCRs is initiated from subcellular hot spots including the plasma membrane, signalosomes, and endosomes to contribute to vascular inflammation. GPCR-G protein signaling at the plasma membrane causes endothelial barrier disruption and also cross-talks with growth factor receptors to promote proinflammatory signaling. A second surge of GPCR signaling is initiated by cytoplasmic NFκB activation mediated by β-arrestins and CARMA-BCL10-MALT1 signalosomes. Once internalized, ubiquitinated GPCRs initiate signaling from endosomes via assembly of the transforming growth factor-β-activated kinase binding protein-1 (TAB1)-TAB2-p38 MAPK complex to promote vascular inflammation. Understanding the complexities of GPCR signaling is critical for development of new strategies to treat vascular inflammation such as that associated with COVID-19.
Collapse
Key Words
- Arrestins
- B-cell lymphoma protein 10, (BCL10)
- COVID-19
- Endosomes
- Endothelial
- G protein-coupled receptor, GPCR
- JAK-STAT
- Janus kinase, JAK
- MALT1
- NFκB
- adherens junctions, AJ
- angiotensin II type 1 receptor, AT1
- angiotensin converting enzyme-2, ACE2
- caspase recruitment domain-containing protein, CARMA
- coronavirus disease of 2019, COVID-19
- fibroblast-growth-factor, FGF
- inhibitor of NFκB kinase, IKK
- mitogen-activated protein kinase, MAPK
- mucosa-associated lymphoid tissue lymphoma translocation protein 1, (MALT1)
- neural precursor cell expressed developmentally downregulated protein 4, NEDD4
- nuclear factor kappa-light-chain-enhancer of activated B cells, NFκB
- p38 MAPK
- platelet activating factor, PAF
- protease-activated receptor-1, PAR1
- severe acute respiratory syndrome coronavirus 2, SARS-CoV-2
- signal transducer and activator of transcription, STAT
- transforming growth factor-α-activated kinase binding protein-1, TAB1
Collapse
Affiliation(s)
- Cierra A Birch
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Olivia Molinar-Inglis
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
3
|
He L, Zhou Z, Shao Y, Yang Z, Zhou S, Zou X, Zhou Y, Tan G. Bradykinin potentially stimulates cell proliferation in rabbit corneal endothelial cells through the ZO‑1/ZONAB pathway. Int J Mol Med 2018; 42:71-80. [PMID: 29568941 PMCID: PMC5979832 DOI: 10.3892/ijmm.2018.3580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/05/2018] [Indexed: 12/20/2022] Open
Abstract
Bradykinin (BK) has been demonstrated to induce proliferation in several types of cell in ex vivo corneas. However, the mechanisms underlying the action of BK on corneal endothelial cells (CECs) remain largely unknown. The present study aimed to investigate the effect of BK on rabbit corneal endothelial cell (RCEC) proliferation, and assess the involvement of the zonula occludens‑1(ZO‑1)/ZO‑1associated nucleic acid binding protein (ZONAB) pathway. Cell proliferation and cell cycle distribution was analyzed following treatment with BK (0.01, 0.1,1.0 or 10.0 µM) for the indicated time intervals (24, 48, 72 and 96 h), or following BK treatment combined with transfection of ZONAB‑small interfering (si)RNA for 72 h. In addition, the expression of tight junction ZO‑1, nuclear ZONAB, proliferating cell nuclear antigen(PCNA) and cyclin D1 were evaluated using western blotting or immunofluorescence. BK treatment was demonstrated to induce time‑ and concentration‑dependent cell proliferation and cell cycle progression, along with the upregulation of tight junction ZO‑1 and nuclear ZONAB, as well as PCNA and cyclin D1 protein expression. Furthermore, knockdown with ZONAB‑siRNA inhibited cell proliferation, induced cell cycle arrest and downregulated PCNA and cyclin D1 protein expression. ZONAB knockdown therefore successfully reversed the increase in proliferation induced by BK treatment. Taken together, these results suggested that BK stimulated RCEC proliferation, potentially via the ZO‑1/ZONAB pathway. The signaling paradigm disclosed in the present study potentially serves as an important therapeutic target for cornea regeneration and transplantation.
Collapse
Affiliation(s)
- Lixian He
- Department of Ophthalmology, The First Affiliated Hospital of University of South China
| | - Zhou Zhou
- Medical College, University of South China, Hengyang, Hunan 421001
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Province Clinical Ophthalmology Institute, Nanchang, Jiangxi 330006
| | - Zhen Yang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shuangshuang Zhou
- Department of Ophthalmology, The First Affiliated Hospital of University of South China
| | - Xuexiang Zou
- Department of Ophthalmology, The First Affiliated Hospital of University of South China
| | - Ying Zhou
- Department of Ophthalmology, The First Affiliated Hospital of University of South China
| | - Gang Tan
- Department of Ophthalmology, The First Affiliated Hospital of University of South China
| |
Collapse
|
4
|
Stephan F, Aarden LA, Zeerleder S. FSAP, a new player in inflammation? Hamostaseologie 2017; 32:51-5. [DOI: 10.5482/ha-1187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/04/2011] [Indexed: 12/11/2022] Open
Abstract
SummaryFactor VII-activating protease (FSAP) is a serine protease in plasma that has a role in coagulation and fibrinolysis. FVII could be activated by purified FSAP in a tissue factor independent manner and pro-urokinase has been demonstrated to be a substrate for purified FSAP in-vitro. However, the physiological role of FSAP in haemostasis remains unclear. More recently FSAP is suggested to be involved in inflammation. It modulates vascular permeability directly and indirectly by the generation of bradykinin. Furthermore, FSAP is activated by dead cells induced by the inflammatory response and subsequently removes nucleosomes from apoptotic cells. FSAP activation can be detected in sepsis patients as well. However, whether FSAP activation upon inflammation is beneficial or detrimental remains an open question.In this review the structure, activation mechanisms and the possible role of FSAP in inflammation are discussed.
Collapse
|
5
|
Urodilatin reverses the detrimental influence of bradykinin in acute ischemic stroke. Exp Neurol 2016; 284:1-10. [DOI: 10.1016/j.expneurol.2016.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/15/2016] [Accepted: 07/14/2016] [Indexed: 02/03/2023]
|
6
|
Dobrivojević M, Špiranec K, Sinđić A. Involvement of bradykinin in brain edema development after ischemic stroke. Pflugers Arch 2014; 467:201-12. [DOI: 10.1007/s00424-014-1519-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 01/04/2023]
|
7
|
Kashuba E, Bailey J, Allsup D, Cawkwell L. The kinin-kallikrein system: physiological roles, pathophysiology and its relationship to cancer biomarkers. Biomarkers 2013; 18:279-96. [PMID: 23672534 DOI: 10.3109/1354750x.2013.787544] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The kinin-kallikrein system (KKS) is an endogenous multiprotein cascade, the activation of which leads to triggering of the intrinsic coagulation pathway and enzymatic hydrolysis of kininogens with the consequent release of bradykinin-related peptides. This system plays a crucial role in inflammation, vasodilation, smooth muscle contraction, cardioprotection, vascular permeability, blood pressure control, coagulation and pain. In this review, we will outline the physiology and pathophysiology of the KKS and also highlight the association of this system with carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Elena Kashuba
- Postgraduate Medical Institute, University of Hull, Hull, UK
| | | | | | | |
Collapse
|
8
|
Dobrivojević M, Sinđić A, Edemir B, Kalweit S, Forssmann WG, Hirsch JR. Interaction between bradykinin and natriuretic peptides via RGS protein activation in HEK-293 cells. Am J Physiol Cell Physiol 2012; 303:C1260-8. [PMID: 23054060 DOI: 10.1152/ajpcell.00033.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, the interaction of natriuretic peptides (NP) and bradykinin (BK) signaling pathways was identified by measuring membrane potential (V(m)) and intracellular Ca(2+) using the patch-clamp technique and flow cytometry in HEK-293 cells. BK and NP receptor mRNA was identified using RT-PCR. BK (100 nM) depolarized cells activating bradykinin receptor type 2 (B(2)R) and Ca(2+)-dependent Cl(-) channels inhibitable by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB; 10 μM). The BK-induced Ca(2+) signal was blocked by the B(2)R inhibitor HOE 140. [Des-Arg(9)]-bradykinin, an activator of B(1)R, had no effect on intracellular Ca(2+). NP [atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), C-type natriuretic peptide (CNP), and urodilatin] depolarized HEK-293 cells inhibiting K(+) channels. ANP, urodilatin, BNP [binding to natriuretic peptide receptor (NPR)-A] and 8-bromo-(8-Br)-cGMP inhibited the BK-induced depolarization while CNP (binding to NPR-Bi) failed to do so. The inhibitory effect on BK-triggered depolarization could be reversed by blocking PKG using the specific inhibitor KT 5823. BK-stimulated depolarization as well as Ca(2+) signaling was completely blocked by the phospholipase C (PLC) inhibitor U-73122 (10 nM). The inositol 1,4,5-trisphosphate receptor blocker 2-aminoethoxydiphenyl borate (2-APB; 50 μM) completely inhibited the BK-induced Ca(2+) signaling. UTP, another activator of the PLC-mediated Ca(2+) signaling pathway, was blocked by U-73122 as well but not by 8-Br-cGMP, indicating an intermediate regulatory step for NP via PKG in BK signaling such as regulators of G-protein signaling (RGS) proteins. When RGS proteins were inhibited by CCG-63802 in the presence of BK and 8-Br-cGMP, cells started to depolarize again. In conclusion, as natural antagonists of the B(2)R signaling pathway, NP may also positively interact in pathological conditions caused by BK.
Collapse
Affiliation(s)
- Marina Dobrivojević
- Department of Physiology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | | | | | | | | | | |
Collapse
|
9
|
Cheng CY, Tseng HC, Yang CM. Bradykinin-mediated cell proliferation depends on transactivation of EGF receptor in corneal fibroblasts. J Cell Physiol 2012; 227:1367-81. [PMID: 21604274 DOI: 10.1002/jcp.22849] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In previous studies, bradykinin (BK) has been shown to induce cell proliferation through BK B2 receptor (B2R) via p42/p44 MAPK in Statens Seruminstitut Rabbit Corneal Cells (SIRCs). In addition to this pathway, EGFR transactivation pathway has been implicated in linking a variety of G-protein coupled receptors to MAPK cascades. Here, we further investigate whether these transactivation mechanisms participating in BK-induced cell proliferation in SIRCs. Using an immunofluorescence staining and RT-PCR, we initially characterize that SIRCs were corneal fibroblasts and predominantly expressed B2R by BK. Inhibition of p42/p44 MAPK by the inhibitors of Src, EGFR, and Akt or transfection with respective siRNAs prevents BK-induced DNA synthesis in SIRCs. The mechanisms underlying these responses were mediated through phosphorylation of Src and EGFR via the formation of Src/EGFR complex which was attenuated by PP1 and AG1478. Moreover, BK-induced p42/p44 MAPK and Akt activation was mediated through EGFR transactivation, which was diminished by the inhibitors of MMP-2/9 and heparin-binding EGF-like factor (HB-EGF). Finally, increased nuclear translocation of Akt and p42/p44 MAPK turns on early gene expression leading to cell proliferation. These results suggest that BK-induced cell proliferation is mediated through c-Src-dependent transactivation of EGFR via MMP2/9-dependent pro-HB-EGF shedding linking to activation of Akt and p42/p44 MAPK in corneal fibroblasts.
Collapse
Affiliation(s)
- Ching-Yi Cheng
- Department of Biomedical Engineering, Chung Yuan Christian University, Tao-Yuan, Taiwan
| | | | | |
Collapse
|
10
|
Vascular pathology and blood-brain barrier disruption in cognitive and psychiatric complications of type 2 diabetes mellitus. Cardiovasc Psychiatry Neurol 2011; 2011:609202. [PMID: 21350721 PMCID: PMC3042607 DOI: 10.1155/2011/609202] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 12/28/2010] [Indexed: 01/13/2023] Open
Abstract
Vascular pathology is recognized as a principle insult in type 2 diabetes mellitus (T2DM). Co-morbidities such as structural brain abnormalities, cognitive, learning and memory deficits are also prevailing in T2DM patients. We previously suggested that microvascular pathologies involving blood-brain barrier (BBB) breakdown results in leakage of serum-derived components into the brain parenchyma, leading to neuronal dysfunction manifested as psychiatric illnesses. The current postulate focuses on the molecular mechanisms controlling BBB permeability in T2DM, as key contributors to the pathogenesis of mental disorders in patients. Revealing the mechanisms underlying BBB dysfunction and inflammatory response in T2DM and their role in metabolic disturbances, abnormal neurovascular coupling and neuronal plasticity, would contribute to the understanding of the mechanisms underlying psychopathologies in diabetic patients. Establishing this link would offer new targets for future therapeutic interventions.
Collapse
|
11
|
Komarova Y, Malik AB. Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu Rev Physiol 2010; 72:463-93. [PMID: 20148685 DOI: 10.1146/annurev-physiol-021909-135833] [Citation(s) in RCA: 488] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endothelium functions as a semipermeable barrier regulating tissue fluid homeostasis and transmigration of leukocytes and providing essential nutrients across the vessel wall. Transport of plasma proteins and solutes across the endothelium involves two different routes: one transcellular, via caveolae-mediated vesicular transport, and the other paracellular, through interendothelial junctions. The permeability of the endothelial barrier is an exquisitely regulated process in the resting state and in response to extracellular stimuli and mediators. The focus of this review is to provide a comprehensive overview of molecular and signaling mechanisms regulating endothelial barrier permeability with emphasis on the cross-talk between paracellular and transcellular transport pathways.
Collapse
Affiliation(s)
- Yulia Komarova
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | |
Collapse
|
12
|
Current concepts of neurohormonal activation in heart failure: mediators and mechanisms. AACN Adv Crit Care 2009; 19:364-85; quiz 386-7. [PMID: 18981739 DOI: 10.1097/01.aacn.0000340718.93742.c4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurohormonal activation is a commonly cited array of phenomena in the body's physiologic response to heart failure. Although various neurohormones and pharmacologic agents that moderate their pathophysiologic effects have been reviewed in the nursing literature, both the mechanisms of neurohormonal system activation and cellular and organ system effects have been described only in brief. Accordingly, this article reviews mechanisms of neurohormonal activation and describes cellular and cardiovascular effects of the (1) sympathetic nervous system, (2) renin-angiotensin-aldosterone system, (3) kallikrein-kininogen-kinin system, (4) vasopressinergic system, (5) natriuretic peptide systems, and (6) endothelin in the context of heart failure. This article implicitly details the physiologic basis for numerous current and potential future pharmacologic agents used in the management of heart failure. It is intended that this article be used as a reference for advanced clinical nursing practice, research, and education.
Collapse
|
13
|
Oeseburg H, Iusuf D, van der Harst P, van Gilst WH, Henning RH, Roks AJM. Bradykinin protects against oxidative stress-induced endothelial cell senescence. Hypertension 2008; 53:417-22. [PMID: 19075096 DOI: 10.1161/hypertensionaha.108.123729] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Premature aging (senescence) of endothelial cells might play an important role in the development and progression of hypertension and atherosclerosis. We hypothesized that bradykinin, a hormone that mediates vasoprotective effects of angiotensin-converting enzyme inhibitors, protects endothelial cells from oxidative stress-induced senescence. Bradykinin treatment (0.001 to 1 nmol/L) dose-dependently decreased senescence induced by 25 micromol/L of H(2)O(2) in cultured bovine aortic endothelial cells, as witnessed by a complete inhibition of increased senescent cell numbers and a 34% reduction of the levels of the senescence-associated cell cycle protein p21. Because H(2)O(2) induces senescence through superoxide-induced DNA damage, single-cell DNA damage was measured by comet assay. Bradykinin reduced DNA damage to control levels. The protective effect of bradykinin also resulted in a significant increase in the migration of H(2)O(2)-treated bovine aorta endothelial cells in an in vitro endothelial injury model, or "scratch" assay. The protective effect of bradykinin was abolished by the bradykinin B2 receptor antagonist HOE-140 and the NO production inhibitor N(omega)-methyl-L-arginine acetate salt. Therefore, we conclude that bradykinin protects endothelial cells from superoxide-induced senescence through bradykinin B2 receptor- and NO-mediated inhibition of DNA damage.
Collapse
Affiliation(s)
- Hisko Oeseburg
- Department of Experimental Cardiology, AB 43, University Medical Center Groningen, Antonius Deusinglaan 1, Building 3211, 9713 AV Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
14
|
Chen LY, Woszczek G, Nagineni S, Logun C, Shelhamer JH. Cytosolic phospholipase A2alpha activation induced by S1P is mediated by the S1P3 receptor in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2008; 295:L326-35. [PMID: 18502815 DOI: 10.1152/ajplung.00393.2007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cytosolic phospholipase A(2)alpha (cPLA(2)alpha) activation is a regulatory step in the control of arachidonic acid (AA) liberation for eicosanoid formation. Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator involved in the regulation of many important proinflammatory processes and has been found in the airways of asthmatic subjects. We investigated the mechanism of S1P-induced AA release and determined the involvement of cPLA(2)alpha in these events in A549 human lung epithelial cells. S1P induced AA release rapidly within 5 min in a dose- and time-dependent manner. S1P-induced AA release was inhibited by the cPLA(2)alpha inhibitors methyl arachidonyl fluorophosphonate (MAFP) and pyrrolidine derivative, by small interfering RNA-mediated downregulation of cPLA(2)alpha, and by inhibition of S1P-induced calcium flux, suggesting a significant role of cPLA(2)alpha in S1P-mediated AA release. Knockdown of the S1P3 receptor, the major S1P receptor expressed on A549 cells, inhibited S1P-induced calcium flux and AA release. The S1P-induced calcium flux and AA release was associated with sphingosine kinase 1 (Sphk1) expression and activity. Furthermore, Rho-associated kinase, downstream of S1P3, was crucial for S1P-induced cPLA(2)alpha activation. Our data suggest that S1P acting through S1P3, calcium flux, and Rho kinase activates cPLA(2)alpha and releases AA in lung epithelial cells. An understanding of S1P-induced cPLA(2)alpha activation mechanisms in epithelial cells may provide potential targets to control inflammatory processes in the lung.
Collapse
Affiliation(s)
- Li-Yuan Chen
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
15
|
Shabir S, Southgate J. Calcium signalling in wound-responsive normal human urothelial cell monolayers. Cell Calcium 2008; 44:453-64. [PMID: 18417211 DOI: 10.1016/j.ceca.2008.02.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 02/21/2008] [Accepted: 02/28/2008] [Indexed: 11/15/2022]
Abstract
Epithelial tissue repair requires coordination of migratory and proliferative activity both adjacent to and remote from the wound edge. Although calcium signalling is implicated, the specific mechanisms are poorly understood. This study characterises the calcium signal invoked in response to scratch wounding of normal human urothelial (NHU) cells and relates it to the localised cellular response. Immediately after wounding of confluent NHU cell monolayers, cells adjacent to the wound edge showed a sustained (>30 min) rise in [Ca(2+)](i), while there was an independent, but simultaneous calcium wave that propagated out from the wound edge. The transient signal involved release of calcium from intracellular stores and was not mediated via gap junctions, but by diffusion of extracellular agonists. We demonstrated that ATP was partially responsible for the initiation and propagation of the calcium wave and showed that the calcium release mechanism was mediated in part via activation of inositol-1,4,5-triphosphate (IP(3)) receptors. By contrast, the sustained calcium signal originated from the extracellular milieu and correlated with an increased rate of migration by these cells. The work presented here provides supportive evidence that the calcium signature, defined by its temporal and amplitude characteristics, is important in co-ordinating the response of cells within an epithelial cell monolayer after wounding.
Collapse
Affiliation(s)
- Saqib Shabir
- Jack Birch Unit of Molecular Carcinogenesis, Department of Biology, University of York YO10 5YW, UK
| | | |
Collapse
|
16
|
Mueller S, Paegelow I, Reissmann S. Hypothesized and found mechanisms for potentiation of bradykinin actions. SIGNAL TRANSDUCTION 2006; 6:5-18. [PMID: 32327962 PMCID: PMC7169587 DOI: 10.1002/sita.200500061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Revised: 11/17/2005] [Indexed: 11/06/2022]
Abstract
Potentiation of hormone actions can occur by different mechanisms, including inhibition of degrading enzymes, interaction with the hormone receptor leading to stabilization of bioactive conformation or leading to receptor homo- and hetero-oligomerization, receptor phosphorylation and dephosphorylation or can occur by directly influencing the signal transduction and ion channels. In this review the potentiation of bradykinin actions in different systems by certain compounds will be reviewed. Despite many long years of experimental research and investigation the mechanisms of potentiating action remain not fully understood. One of the most contradictory findings are the distinct differences between the inhibition of the angiotensin I-converting enzyme and the potentiation of the bradykinin induced smooth muscle reaction. Contradictory findings and hypothesized mechanisms in the literature are discussed in this review and in some cases compared to own results. Investigation of potentiating actions was extended from hypotension, smooth muscle reaction and cellular actions to activation of immunocompetent cells. In our opinion the potentiation of bradykinin action can occur by different mechanisms, depending on the system and the applied potentiating factor used.
Collapse
Affiliation(s)
- Sylvia Mueller
- Institute of Biochemistry and Biophysics, Biological and Pharmaceutical Faculty, Friedrich‐Schiller‐University Jena, Jena, Germany. Fax: +49 3641 949352
| | - Inge Paegelow
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Rostock, Rostock, Germany
| | | |
Collapse
|
17
|
Fang Y, Li GG, Peng J. Optical biosensor provides insights for bradykinin B(2) receptor signaling in A431 cells. FEBS Lett 2005; 579:6365-6374. [PMID: 16263113 DOI: 10.1016/j.febslet.2005.10.019] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 10/10/2005] [Accepted: 10/13/2005] [Indexed: 11/18/2022]
Abstract
The spatial and temporal targeting of proteins or protein assemblies to appropriate sites is crucial to regulate the specificity and efficiency of protein-protein interactions, thus dictating the timing and intensity of cell signaling and responses. The resultant dynamic mass redistribution could be manifested by label free optical biosensor, and lead to a novel and functional optical signature for studying cell signaling. Here we applied this technology, termed as mass redistribution cell assay technology (MRCAT), to study the signaling networks of bradykinin B(2) receptor in A431 cells. Using MRCAT, the spatial and temporal relocation of proteins and protein assemblies mediated by bradykinin was quantitatively monitored in microplate format and in live cells. The saturability to bradykinin, together with the specific and dose-dependent inhibition by a B(2) specific antagonist HOE140, suggested that the optical signature is a direct result of B(2) receptor activation. The sensitivity of the optical signature to cholesterol depletion by methyl-beta-cyclodextrin argued that B(2) receptor signaling is dependent on the integrity of lipid rafts; disruption of these microdomains hinders the B(2) signaling. Modulations of several important intracellular targets with specific inhibitors suggested that B(2) receptor activation results in signaling via at least dual pathways - G(s)- and G(q)-mediated signaling. Remarkably, the two signaling pathways counter-regulate each other. Several critical downstream targets including protein kinase C, protein kinase A, and epidermal growth factor receptor had been identified to involve in B(2) signaling. The roles of endocytosis and cytoskeleton modulation in B(2) signaling were also demonstrated.
Collapse
Affiliation(s)
- Ye Fang
- Biochemical Technologies, Science and Technology Division, Corning Incorporated, Sullivan Park, Corning, NY 14831, USA.
| | | | | |
Collapse
|
18
|
|