1
|
Fabiano A, Panichi D, Picone S, Lapergola G, Levantini G, D'Adamo E, Strozzi M, Gavilanes DA, Kramer BW, Gazzolo F, Abdelhameed AS, Gazzolo D. Changes in adrenomedullin in bronchoalveolar lavage fluid with chorioamnionitis in a sheep-based model. J Matern Fetal Neonatal Med 2025; 38:2456502. [PMID: 39848627 DOI: 10.1080/14767058.2025.2456502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Adrenomedullin (AM) is a potent angiogenic, antioxidant and anti-inflammatory peptide protecting the developing lung from injury due to bronchopulmonary dysplasia (BPD) of the preterm infant. At this stage, no data on the potential effects of chorioamnionitis (CA) occurrence and glucocorticoids (GC) administration on AM in developing lungs are still lacking. OBJECTIVE to investigate, in a sheep-based model, the positive/side-effects of combined exposure to CA and GC on AM concentrations measured in bronchoalveolar lavage fluid (BALF). METHODS Time-mated ewes were randomly admitted to one of six treatment groups receiving injection: saline (controls); lipopolysaccharide (L) in intra-amniotic fluid treated alone at 7 or 14 d before delivery or associated with betamethasone (B) intramuscularly; B treated alone (7d) or associated with L (14d). Lambs were surgically delivered at 120 days gestation and euthanized. BALF was used for AM measurement in the studied groups. RESULTS AM BALF levels significantly (p < 0.05, for all) changed both to B and L exposure in a time-dependent manner. The latter was characterized by AM levels at short term superimposable to controls, whilst significantly (p > 0.05) decreased at long-term. The former showed increased AM at short and decreased at long-term (p < 0.05, for all), respectively. CONCLUSIONS the present results showing AM BALF changes in a sheep-based model support the AM role in the hemodynamic patterns due to CA and BPD occurrence and open the way to further studies investigating the role of vasoactive agents as trustable markers of lung development/damage.
Collapse
Affiliation(s)
- Adele Fabiano
- Neonatal Intensive Care Unit, San Giovanni Addolorata Hospital, Rome, Italy
| | - Daniele Panichi
- Neonatal Intensive Care Unit, Maggiore Hospital, AUSL Bologna, Bologna, Italy
| | - Simonetta Picone
- Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino General Hospital, Rome, Italy
| | | | | | - Ebe D'Adamo
- Neonatal Intensive Care Unit, G d'Annunzio University of Chieti, Italy
| | | | - Danilo Aw Gavilanes
- Department of Pediatrics and Neonatology, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Ali Saber Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Diego Gazzolo
- Neonatal Intensive Care Unit, G d'Annunzio University of Chieti, Italy
| |
Collapse
|
2
|
Balint L, Patel S, Serafin DS, Zhang H, Quinn KE, Aghajanian A, Kistner BM, Caron KM. Lymphatic Activation of ACKR3 Signaling Regulates Lymphatic Response After Ischemic Heart Injury. Arterioscler Thromb Vasc Biol 2025; 45:754-768. [PMID: 40143814 PMCID: PMC12018146 DOI: 10.1161/atvbaha.124.322288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Ischemic heart disease is a prevalent cause of death and disability worldwide. Recent studies reported a rapid expansion of the cardiac lymphatic network upon ischemic heart injury and proposed that cardiac lymphatics may attenuate tissue edema and inflammatory mechanisms after ischemic heart injury. Nevertheless, the mechanisms through which hypoxic conditions affect cardiac lymphangiogenesis and function remain unclear. Here, we aimed to characterize the role of the AM (adrenomedullin) decoy receptor ACKR3 (atypical chemokine receptor-3) in the lymphatic response following ischemic heart injury. METHODS Spatial assessment of ACKR3 signaling in the heart after ischemic heart injury was conducted using ACKR3-Tango-GFP (green fluorescent protein) reporter mice. Roles of ACKR3 after ischemic heart injury were characterized in Ackr3∆Lyve1 mice and in cultured human lymphatic endothelial cells exposed to hypoxia. RESULTS Using the novel ACKR3-Tango-GFP reporter mice, we detected activation of ACKR3 signaling in cardiac lymphatics adjacent to the site of ischemic injury of left anterior descending artery ligation. Ackr3∆Lyve1 mice exhibited better survival after left anterior descending artery ligation, especially within the first couple of days post-injury, and were protected from the formation of acute tissue edema. Ackr3∆Lyve1 mice exhibited a denser cardiac lymphatic network after left anterior descending artery ligation, especially in the injured tissues. Transcriptomic analysis revealed changes in cardiac lymphatic gene expression patterns that have been associated with extracellular matrix remodeling and immune activation. We also found that ACKR3 plays a critical role in regulating continuous cell-cell junction dynamics in lymphatic endothelial cells under hypoxic conditions. CONCLUSIONS Lymphatic expression of ACKR3 governs numerous processes following ischemic heart injury, including the lymphangiogenic response, edema protection, and overall survival. These results expand our understanding of how the heart failure biomarker AM, regulated by lymphatic ACKR3, may exert its roles after ischemic cardiac injury.
Collapse
Affiliation(s)
- Laszlo Balint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Shubhangi Patel
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - D. Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Kelsey E. Quinn
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
- Department of Medicine, Division of Cardiology, University of North Carolina at Chapel Hill
| | - Bryan M. Kistner
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| |
Collapse
|
3
|
Balint L, Patel S, Serafin DS, Zhang H, Quinn KE, Aghajanian A, Kistner BM, Caron KM. Lymphatic activation of ACKR3 signaling regulates lymphatic response after ischemic heart injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626683. [PMID: 39679907 PMCID: PMC11642902 DOI: 10.1101/2024.12.04.626683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Background Ischemic heart disease is a prevalent cause of death and disability worldwide. Recent studies reported a rapid expansion of the cardiac lymphatic network upon ischemic heart injury and proposed that cardiac lymphatics may attenuate tissue edema and inflammatory mechanisms after ischemic heart injury. Nevertheless, the mechanisms through which hypoxic conditions affect cardiac lymphangiogenesis and function remain unclear. Here, we aimed to characterize the role of the adrenomedullin decoy receptor atypical chemokine receptor 3 (ACKR3) in the lymphatic response following ischemic heart injury. Methods Spatial assessment of ACKR3 signaling in the heart after ischemic heart injury was conducted using ACKR3-TangoGFP reporter mice. Roles of ACKR3 after ischemic heart injury were characterized in Ackr3 ΔLyve 1 mice and in cultured human lymphatic endothelial cells (LECs) exposed to hypoxia. Results Using the novel ACKR3-Tango-GFP reporter mice, we detected activation of ACKR3 signaling in cardiac lymphatics adjacent to the site of ischemic injury of left anterior descending artery (LAD) ligation. Ackr3 ΔLyve 1 mice exhibited better survival and were protected from the formation of acute tissue edema after ischemic cardiac injury. Ackr3 ΔLyve 1 mice exhibited a denser cardiac lymphatic network after LAD ligation, especially in the injured tissues. Transcriptomic analysis revealed changes in cardiac lymphatic gene expression patterns that have been associated with extracellular matrix remodeling and immune activation. We also found that ACKR3 plays a critical role in the regulating continuous cell-cell junction dynamics in LECs under hypoxic conditions. Conclusions Lymphatic expression of ACKR3 governs numerous processes following ischemic heart injury, including the lymphangiogenic response, edema protection and overall survival. These results expand our understanding of how the heart failure biomarker adrenomedullin, regulated by lymphatic ACKR3, may exert its cardioprotective roles after ischemic cardiac injury.
Collapse
|
4
|
van Lier D, Kox M, Pickkers P. Promotion of vascular integrity in sepsis through modulation of bioactive adrenomedullin and dipeptidyl peptidase 3. J Intern Med 2021; 289:792-806. [PMID: 33381880 PMCID: PMC8246835 DOI: 10.1111/joim.13220] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022]
Abstract
Sepsis represents one of the major medical challenges of the 21st century. Despite substantial improvements in the knowledge on pathophysiological mechanisms, this has so far not translated into novel adjuvant treatment strategies for sepsis. In sepsis, both vascular tone and vascular integrity are compromised, and contribute to the development of shock, which is strongly related to the development of organ dysfunction and mortality. In this review, we focus on dipeptidyl peptidase 3 (DPP3) and adrenomedullin (ADM), two molecules that act on the vasculature and are involved in the pathophysiology of sepsis and septic shock. DPP3 is an ubiquitous cytosolic enzyme involved in the degradation of several important signalling molecules essential for regulation of vascular tone, including angiotensin II. ADM is a key hormone involved in the regulation of vascular tone and endothelial barrier function. Previous studies have shown that circulating concentrations of both DPP3 and ADM are independently associated with the development of organ failure and adverse outcome in sepsis. We now discuss new evidence illustrating that these molecules indeed represent two distinct pathways involved in the development of septic shock. Recently, both ADM-enhancing therapies aimed at improving endothelial barrier function and vascular tone and DPP3-blocking therapies aimed at restoring systemic angiotensin responses have been shown to improve outcome in various preclinical sepsis models. Given the current lack of effective adjuvant therapies in sepsis, additional research on the therapeutic application of these peptides in humans is highly warranted.
Collapse
Affiliation(s)
- D van Lier
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Kox
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - P Pickkers
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Mid-Regional Proadrenomedullin as a New Biomarker of Kidney and Cardiovascular Diseases-Is It the Future? J Clin Med 2021; 10:jcm10030524. [PMID: 33540505 PMCID: PMC7867137 DOI: 10.3390/jcm10030524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 01/19/2023] Open
Abstract
The increasing prevalence of cardiovascular disease and concomitant chronic kidney disease among the aging populations is responsible for considerable growth of mortality. Additionally, frequent, prolonged hospitalizations and long-term treatment generates progressive decline in bodily functions as well as substantial public health and economic burden. Accessibility to easy, non-invasive prognostic markers able to detect patients at risk of cardiovascular events may improve effective therapy and mitigate disease progression. Moreover, an early diagnosis allows time for implementation of prophylactic and educational programs that may result in decreased morbidity, improved quality of life and reduced public health expenditure. One of the promising candidates for a novel cardiovascular biomarker is mid-regional proadrenomedullin, a derivative of adrenomedullin. Adrenomedullin is a peptide hormone known for its vasodilatory, antioxidant, antiapoptotic and antifibrotic effects. A remarkable advantage of mid-regional proadrenomedullin is its longer half-life which is a prerequisite for plasma measurements. These review aims to discuss the importance of mid-regional proadrenomedullin with reference to its usefulness as a biomarker of increased cardiovascular risk and kidney disease progression.
Collapse
|
6
|
Broeker KA, Fuchs MA, Schrankl J, Kurt B, Nolan KA, Wenger RH, Kramann R, Wagner C, Kurtz A. Different subpopulations of kidney interstitial cells produce erythropoietin and factors supporting tissue oxygenation in response to hypoxia in vivo. Kidney Int 2020; 98:918-931. [DOI: 10.1016/j.kint.2020.04.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 01/04/2023]
|
7
|
Bersani I, Pluchinotta F, Dotta A, Savarese I, Campi F, Auriti C, Chuklantseva N, Piersigilli F, Gazzolo F, Varrica A, Satriano A, Gazzolo D. Early predictors of perinatal brain damage: the role of neurobiomarkers. Clin Chem Lab Med 2020; 58:471-486. [PMID: 31851609 DOI: 10.1515/cclm-2019-0725] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/31/2019] [Indexed: 09/17/2023]
Abstract
The early detection of perinatal brain damage in preterm and term newborns (i.e. intraventricular hemorrhage, periventricular leukomalacia and perinatal asphyxia) still constitute an unsolved issue. To date, despite technological improvement in standard perinatal monitoring procedures, decreasing the incidence of perinatal mortality, the perinatal morbidity pattern has a flat trend. Against this background, the measurement of brain constituents could be particularly useful in the early detection of cases at risk for short-/long-term brain injury. On this scenario, the main European and US international health-care institutions promoted perinatal clinical and experimental neuroprotection research projects aimed at validating and including a panel of biomarkers in the clinical guidelines. Although this is a promising attempt, there are several limitations that do not allow biomarkers to be included in standard monitoring procedures. The main limitations are: (i) the heterogeneity of neurological complications in the perinatal period, (ii) the small cohort sizes, (iii) the lack of multicenter investigations, (iv) the different techniques for neurobiomarkers assessment, (iv) the lack of consensus for the validation of assays in biological fluids such as urine and saliva, and (v), the lack of reference curves according to measurement technique and biological fluid. In the present review we offer an up-to-date overview of the most promising developments in the use of biomarkers in the perinatal period such as calcium binding proteins (S100B protein), vasoactive agents (adrenomedullin), brain biomarkers (activin A, neuron specific enolase, glial fibrillary acidic protein, ubiquitin carboxyl-terminal hydrolase-L1) and oxidative stress markers.
Collapse
Affiliation(s)
- Iliana Bersani
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Pluchinotta
- Laboratory Research Department of Pediatric Cardiovascular Surgery, SanDonato Milanese Univerity Hospital, San Donato Milanese, Milan, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Immacolata Savarese
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Campi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cinzia Auriti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Natalia Chuklantseva
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fiammetta Piersigilli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Alessandro Varrica
- Laboratory Research Department of Pediatric Cardiovascular Surgery, SanDonato Milanese Univerity Hospital, San Donato Milanese, Milan, Italy
| | - Angela Satriano
- Laboratory Research Department of Pediatric Cardiovascular Surgery, SanDonato Milanese Univerity Hospital, San Donato Milanese, Milan, Italy
| | - Diego Gazzolo
- Neonatal Intensive Care Unit, G. d'Annunzio University, Chieti, Italy
- Neonatal Intesive Care Unit, AO S.S. Antonio, Biagio, C. Arrigo Hospital, Spalto Marengo 46, 15100 Alessandria, Italy
| |
Collapse
|
8
|
Abstract
Sepsis remains a major scientific and medical challenge, for which, apart from significant refinements in supportive therapy, treatment has barely changed over the last few decades. During sepsis, both vascular tone and vascular integrity are compromised, and contribute to the development of shock. The free circulating peptide adrenomedullin (ADM) is involved in the regulation of the endothelial barrier function and tone of blood vessels. Several animal studies have shown that ADM administration improves outcome of sepsis. However, in higher dosages, ADM administration may cause hypotension, limiting its clinical applicability. Moreover, ADM has a very short half-life and easily adheres to surfaces, further hampering its clinical use. The non-neutralizing anti-ADM antibody Adrecizumab (HAM8101) which causes a long-lasting increase of plasma ADM has shown promising results in animal models of systemic inflammation and sepsis; it reduced inflammation, attenuated vascular leakage, and improved hemodynamics, kidney function, and survival. Combined with an excellent safety profile derived from animal and phase I human studies, Adrecizumab represents a promising candidate drug for the adjunctive treatment of sepsis. In this review, we first provide a brief overview of the currently available data on the role of adrenomedullin in sepsis and describe its effects on endothelial barrier function and vasodilation. Furthermore, we provide a novel hypothesis concerning the mechanisms of action through which Adrecizumab may exert its beneficial effects in sepsis.
Collapse
|
9
|
Geven C, Kox M, Pickkers P. Adrenomedullin and Adrenomedullin-Targeted Therapy As Treatment Strategies Relevant for Sepsis. Front Immunol 2018; 9:292. [PMID: 29520277 PMCID: PMC5827550 DOI: 10.3389/fimmu.2018.00292] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/01/2018] [Indexed: 12/29/2022] Open
Abstract
Sepsis remains a major medical challenge, for which, apart from improvements in supportive care, treatment has not relevantly changed over the last few decades. Vasodilation and vascular leakage play a pivotal role in the development of septic shock, with vascular leakage being caused by disrupted endothelial integrity. Adrenomedullin (ADM), a free circulating peptide involved in regulation of endothelial barrier function and vascular tone, is implicated in the pathophysiology of sepsis. ADM levels are increased during sepsis, and correlate with extent of vasodilation, as well as with disease severity and mortality. In vitro and preclinical in vivo data show that administration of ADM exerts anti-inflammatory, antimicrobial, and protective effects on endothelial barrier function during sepsis, but other work suggests that it may also decrease blood pressure, which could be detrimental for patients with septic shock. Work has been carried out to negate ADMs putative negative effects, while preserving or even potentiating its beneficial actions. Preclinical studies have demonstrated that the use of antibodies that bind to the N-terminus of ADM results in an overall increase of circulating ADM levels and improves sepsis outcome. Similar beneficial effects were obtained using coadministration of ADM and ADM-binding protein-1. It is hypothesized that the mechanism behind the beneficial effects of ADM binding involves prolongation of its half-life and a shift of ADM from the interstitium to the circulation. This in turn results in increased ADM activity in the blood compartment, where it exerts beneficial endothelial barrier-stabilizing effects, whereas its detrimental vasodilatory effects in the interstitium are reduced. Up till now, in vivo data on ADM-targeted treatments in humans are lacking; however, the first study in septic patients with an N-terminus antibody (Adrecizumab) is currently being conducted.
Collapse
Affiliation(s)
- Christopher Geven
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
10
|
Abstract
Adrenomedullin (ADM), the product of the vascular endothelial and smooth muscle cells, and cardiomyocytes, is considered to be a local factor controlling vascular tone, cardiac contractility and renal sodium excretion. The aim of this article was to review the existing data on the effect of different types of exercise on plasma ADM concentration in healthy men. The results of studies on the effect of dynamic exercise on the plasma ADM are contradictory. Some authors reported an increase in plasma ADM, while others showed a slight decrease or did not observe any changes. The inverse relationship between plasma ADM and mean blood pressure observed during maximal exercise support the concept that ADM might blunt the exercise-induced systemic blood pressure increase. Positive relationships between increases in plasma ADM and those in noradrenaline, atrial natriuretic peptide (ANP) or interleukin-6 observed during prolonged exercise suggest that the sympathetic nervous system and cytokine induction may be involved in ADM release. Increased secretion of ADM and ANP during this type of exercise may be a compensatory mechanism attenuating elevation of blood pressure and preventing deterioration of cardiac function. Studies performed during static exercise have showed an increase in plasma ADM only in older healthy men. Positive correlations between increases in plasma ADM and those in noradrenaline and endothelin-1 may indicate the interaction of these hormones in shaping the cardiovascular response to static exercise. Inverse relationships between exercise-induced changes in plasma ADM and those in cardiovascular indices may be at least partly associated with inotropic action of ADM on the heart. Interactions of ADM with vasoactive peptides, catecholamines and hemodynamic factors demonstrate the potential involvement of this peptide in the regulation of blood pressure and myocardial contractility during exercise.
Collapse
Affiliation(s)
- Krzysztof Krzeminski
- Department of Applied Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
11
|
García-Ponce A, Chánez Paredes S, Castro Ochoa KF, Schnoor M. Regulation of endothelial and epithelial barrier functions by peptide hormones of the adrenomedullin family. Tissue Barriers 2016; 4:e1228439. [PMID: 28123925 DOI: 10.1080/21688370.2016.1228439] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 01/16/2023] Open
Abstract
The correct regulation of tissue barriers is of utmost importance for health. Barrier dysfunction accompanies inflammatory disorders and, if not controlled properly, can contribute to the development of chronic diseases. Tissue barriers are formed by monolayers of epithelial cells that separate organs from their environment, and endothelial cells that cover the vasculature, thus separating the blood stream from underlying tissues. Cells within the monolayers are connected by intercellular junctions that are linked by adaptor molecules to the cytoskeleton, and the regulation of these interactions is critical for the maintenance of tissue barriers. Many endogenous and exogenous molecules are known to regulate barrier functions in both ways. Proinflammatory cytokines weaken the barrier, whereas anti-inflammatory mediators stabilize barriers. Adrenomedullin (ADM) and intermedin (IMD) are endogenous peptide hormones of the same family that are produced and secreted by many cell types during physiologic and pathologic conditions. They activate certain G-protein-coupled receptor complexes to regulate many cellular processes such as cytokine production, actin dynamics and junction stability. In this review, we summarize current knowledge about the barrier-stabilizing effects of ADM and IMD in health and disease.
Collapse
Affiliation(s)
- Alexander García-Ponce
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Sandra Chánez Paredes
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Karla Fabiola Castro Ochoa
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN) , Mexico City, Mexico
| |
Collapse
|
12
|
Bersani I, Auriti C, Ronchetti MP, Prencipe G, Gazzolo D, Dotta A. Use of early biomarkers in neonatal brain damage and sepsis: state of the art and future perspectives. BIOMED RESEARCH INTERNATIONAL 2015; 2015:253520. [PMID: 25685774 PMCID: PMC4313065 DOI: 10.1155/2015/253520] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/11/2014] [Indexed: 12/19/2022]
Abstract
The identification of early noninvasive biochemical markers of disease is a crucial issue of the current scientific research, particularly during the first period of life, since it could provide useful and precocious diagnostic information when clinical and radiological signs are still silent. The ideal biomarker should be practical and sensitive in the precocious identification of at risk patients. An earlier diagnosis may lead to a larger therapeutic window and improve neonatal outcome. Brain damage and sepsis are common causes of severe morbidity with poor outcome and mortality during the perinatal period. A large number of potential biomarkers, including neuroproteins, calcium binding proteins, enzymes, oxidative stress markers, vasoactive agents, and inflammatory mediators, have been so far investigated. The aim of the present review was to provide a brief overview of some of the more commonly investigated biomarkers used in case of neonatal brain damage and sepsis.
Collapse
Affiliation(s)
- Iliana Bersani
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, Rome, Italy
| | - Cinzia Auriti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, Rome, Italy
| | - Maria Paola Ronchetti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, Rome, Italy
| | - Giusi Prencipe
- Department of Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, Rome, Italy
| | - Diego Gazzolo
- Department of Maternal, Fetal and Neonatal Medicine, C. Arrigo Children's Hospital, Alessandria, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, Rome, Italy
| |
Collapse
|
13
|
Potentiated adrenomedullin-induced vasorelaxation during hypoxia in organ cultured porcine coronary arteries. J Cardiovasc Pharmacol 2014; 63:58-67. [PMID: 24084221 DOI: 10.1097/fjc.0000000000000025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This study describes the effect of variable oxygen supply on relaxing responses induced by α-calcitonin gene-related peptide (CGRP) and adrenomedullin (AM) on isolated pig coronary arteries in vitro. Organ culture during normoxia (21% of O₂) and hypoxia (5% of O₂) induced a significant leftward shift of the AM concentration-response curves compared with fresh vessels altering the pEC₅₀ values from 6.9 ± 0.04 to 8.0 ± 0.04, whereas the potency (pEC₅₀) of αCGRP was attenuated from 8.8 ± 0.04 to 7.6 ± 0.04. AM₂₂₋₅₂ exerted significant antagonistic effect on AM-induced vasorelaxation in hypoxic and normoxic conditions (apparent pK(B) = 6.8-7.2), whereas no antagonistic effect was observed in fresh and hyperoxic (95%) organ cultured vessels. The antagonistic effect exerted by αCGRP₈₋₃₇ (10⁻⁶·⁵-10⁻⁵·⁵ M) on αCGRP-induced vasodilatation in fresh vessels (derived from Schild plot pA₂ = 7.4 ± 0.1) was unaltered during organ culture. The antagonistic effect exerted by αCGRP₈₋₃₇ (10⁻⁶ M) on AM-induced vasorelaxation in fresh vessels (apparent pK(B) = 7.4 ± 0.1) was absent during hypoxic organ culture. The receptor activity-modifying proteins 1 (RAMP1)/calcitonin-like receptor (CLR) messenger RNA ratio was reduced and RAMP2/CLR messenger RNA ratio was increased during hypoxic and normoxic organ culture compared with fresh vessels. Hypoxic organ culture for 24-72 hours potentiated the AM-induced vasorelaxation through an AM₂₂₋₅₂-sensitive receptor but attenuated the vasorelaxant effect of CGRP through the CGRP receptors. This could possibly be explained by relatively decreased levels of RAMP1, thus favoring RAMP2 + CLR complex (=AM receptor) formation during hypoxic organ culture.
Collapse
|
14
|
Myojo M, Nagata D, Fujita D, Kiyosue A, Takahashi M, Satonaka H, Morishita Y, Akimoto T, Nagai R, Komuro I, Hirata Y. Telmisartan activates endothelial nitric oxide synthase via Ser1177 phosphorylation in vascular endothelial cells. PLoS One 2014; 9:e96948. [PMID: 24827148 PMCID: PMC4020804 DOI: 10.1371/journal.pone.0096948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/13/2014] [Indexed: 12/13/2022] Open
Abstract
Because endothelial nitric oxide synthase (eNOS) has anti-inflammatory and anti-arteriosclerotic functions, it has been recognized as one of the key molecules essential for the homeostatic control of blood vessels other than relaxation of vascular tone. Here, we examined whether telmisartan modulates eNOS function through its pleiotropic effect. Administration of telmisartan to mice significantly increased the phosphorylation level of eNOS (Ser1177) in the aortic endothelium, but administration of valsartan had no effect. Similarly, telmisartan treatment of human umbilical vein endothelial cells significantly increased the phosphorylation levels of AMP-activated protein kinase (Thr172) and eNOS and the concentration of intracellular guanosine 3′,5′-cyclic monophosphate (cGMP). Furthermore, pretreatment with a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor suppressed the increased phosphorylation level of eNOS and intracellular cGMP concentration. These data show that telmisartan increases eNOS activity through Ser1177 phosphorylation in vascular endothelial cells mainly via p38 MAPK signaling.
Collapse
Affiliation(s)
- Masahiro Myojo
- Department of Cardiovascular Medicine, The University of Tokyo, Graduate school of Medicine, Tokyo, Japan
| | - Daisuke Nagata
- Department of Nephrology, Jichi Medical University, Tochigi, Japan
- * E-mail:
| | - Daishi Fujita
- Department of Cardiovascular Medicine, The University of Tokyo, Graduate school of Medicine, Tokyo, Japan
| | - Arihiro Kiyosue
- Department of Cardiovascular Medicine, The University of Tokyo, Graduate school of Medicine, Tokyo, Japan
| | - Masao Takahashi
- Department of Cardiovascular Medicine, The University of Tokyo, Graduate school of Medicine, Tokyo, Japan
| | - Hiroshi Satonaka
- Department of Cardiology and Nephrology, Dokkyo Medical University, Tochigi, Japan
| | | | - Tetsu Akimoto
- Department of Nephrology, Jichi Medical University, Tochigi, Japan
| | | | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo, Graduate school of Medicine, Tokyo, Japan
| | | |
Collapse
|
15
|
Goldfarb-Rumyantzev AS, Alper SL. Short-term responses of the kidney to high altitude in mountain climbers. Nephrol Dial Transplant 2014; 29:497-506. [PMID: 23525530 PMCID: PMC3938295 DOI: 10.1093/ndt/gft051] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/24/2013] [Indexed: 01/07/2023] Open
Abstract
In high-altitude climbers, the kidneys play a crucial role in acclimatization and in mountain sickness syndromes [acute mountain sickness (AMS), high-altitude cerebral edema, high-altitude pulmonary edema] through their roles in regulating body fluids, electrolyte and acid-base homeostasis. Here, we discuss renal responses to several high-altitude-related stresses, including changes in systemic volume status, renal plasma flow and clearance, and altered acid-base and electrolyte status. Volume regulation is considered central both to high-altitude adaptation and to maladaptive development of mountain sickness. The rapid and powerful diuretic response to the hypobaric hypoxic stimulus of altitude integrates decreased circulating concentrations of antidiuretic hormone, renin and aldosterone, increased levels of natriuretic hormones, plasma and urinary epinephrine, norepinephrine, endothelin and urinary adrenomedullin, with increased insensible fluid losses and reduced fluid intake. The ventilatory and hormonal responses to hypoxia may predict susceptibility to AMS, also likely influenced by multiple genetic factors. The timing of altitude increases and adaptation also modifies the body's physiologic responses to altitude. While hypovolemia develops as part of the diuretic response to altitude, coincident vascular leak and extravascular fluid accumulation lead to syndromes of high-altitude sickness. Pharmacological interventions, such as diuretics, calcium blockers, steroids, phosphodiesterase inhibitors and β-agonists, may potentially be helpful in preventing or attenuating these syndromes.
Collapse
Affiliation(s)
- Alexander S. Goldfarb-Rumyantzev
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Transplant Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Seth L. Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Risso FM, Sannia A, Gavilanes DAW, Vles HJ, Colivicchi M, Ricotti A, Li Volti G, Gazzolo D. Biomarkers of brain damage in preterm infants. J Matern Fetal Neonatal Med 2012; 25 Suppl 4:101-104. [PMID: 22958034 DOI: 10.3109/14767058.2012.715024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE There is growing evidence on the usefulness of biomarkers in the early detection of preterm infants at risk for brain damage. However, among different tools Activin A, S100B protein and adrenomedullin assessment offer the possibility to investigate brain/multiorgan function and development. This could be especially useful in perinatal medicine that requires even more non-invasive techniques in order to fulfill the minimal handling in diagnostic and therapeutic strategy performance. MATERIALS AND METHODS The concept of Unconventional Biological Fluid (UBF: urine and saliva) is becoming even stronger and regards the assessment in non-invasive biological fluids of biochemical markers involved in the cascade of events leading to brain damage. RESULTS Activin A, S100B protein and adrenomedullin in UBF were increased in preterm newborns developing brain damage and/or ominous outcome. CONCLUSIONS The present manuscript offers an update on the usefulness of Activin A, S100B protein an adrenomedullin in UBF as brain damage markers. The findings open a new cue on the use of these markers in daily neonatal intensive care unit (NICU) activities.
Collapse
MESH Headings
- Activins/analysis
- Activins/genetics
- Activins/metabolism
- Adrenomedullin/analysis
- Adrenomedullin/genetics
- Adrenomedullin/metabolism
- Biomarkers/analysis
- Biomarkers/cerebrospinal fluid
- Biomarkers/metabolism
- Biomarkers/urine
- Brain Injuries/cerebrospinal fluid
- Brain Injuries/diagnosis
- Brain Injuries/metabolism
- Brain Injuries/urine
- Humans
- Infant, Newborn
- Infant, Premature/cerebrospinal fluid
- Infant, Premature/metabolism
- Infant, Premature/urine
- Infant, Premature, Diseases/cerebrospinal fluid
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/metabolism
- Infant, Premature, Diseases/urine
- Nerve Growth Factors/analysis
- Nerve Growth Factors/genetics
- Nerve Growth Factors/metabolism
- S100 Calcium Binding Protein beta Subunit
- S100 Proteins/analysis
- S100 Proteins/genetics
- S100 Proteins/metabolism
- Saliva/chemistry
- Saliva/metabolism
Collapse
Affiliation(s)
- Francesco M Risso
- Department of Neonatology, G Gaslini Children's University Hospital, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Geiger K, Leiherer A, Muendlein A, Stark N, Geller-Rhomberg S, Saely CH, Wabitsch M, Fraunberger P, Drexel H. Identification of hypoxia-induced genes in human SGBS adipocytes by microarray analysis. PLoS One 2011; 6:e26465. [PMID: 22039494 PMCID: PMC3198480 DOI: 10.1371/journal.pone.0026465] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/27/2011] [Indexed: 01/04/2023] Open
Abstract
Hypoxia in adipose tissue is suggested to be involved in the development of a chronic mild inflammation, which in obesity can further lead to insulin resistance. The effect of hypoxia on gene expression in adipocytes appears to play a central role in this inflammatory response observed in obesity. However, the global impact of hypoxia on transcriptional changes in human adipocytes is unclear. Therefore, we compared gene expression profiles of human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes under normoxic or hypoxic conditions to detect hypoxia-responsive genes in adipocytes by using whole human genome microarrays. Microarray analysis showed more than 500 significantly differentially regulated mRNAs after incubation of the cells under low oxygen levels. To gain further insight into the biological processes, hypoxia-regulated genes after 16 hours of hypoxia were classified according to their function. We identified an enrichment of genes involved in important biological processes such as glycolysis, response to hypoxia, regulation of cellular component movement, response to nutrient levels, regulation of cell migration, and transcription regulator activity. Real-time PCR confirmed eight genes to be consistently upregulated in response to 3, 6 and 16 hours of hypoxia. For adipocytes the hypoxia-induced regulation of these genes is shown here for the first time. Moreover in six of these eight genes we identified HIF response elements in the proximal promoters, specific for the HIF transcription factor family members HIF1A and HIF2A. In the present study, we demonstrated that hypoxia has an extensive effect on gene expression of SGBS adipocytes. In addition, the identified hypoxia-regulated genes are likely involved in the regulation of obesity, the incidence of type 2 diabetes, and the metabolic syndrome.
Collapse
Affiliation(s)
- Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
- Medical Central Laboratories, Feldkirch, Austria
| | - Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Nicole Stark
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Simone Geller-Rhomberg
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Christoph H. Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Department of Medicine and Cardiology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University of Ulm, Ulm, Germany
| | | | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Department of Medicine and Cardiology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
- Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Kiyosue A, Nagata D, Myojo M, Sato T, Takahashi M, Satonaka H, Nagai R, Hirata Y. Aldosterone-induced osteopontin gene transcription in vascular smooth muscle cells involves glucocorticoid response element. Hypertens Res 2011; 34:1283-7. [PMID: 21814207 DOI: 10.1038/hr.2011.119] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteopontin (OPN) is known to be one of the cytokines that is involved in the vascular inflammation caused by aldosterone (Aldo). Previous reports have shown that Aldo increases OPN transcripts, and the mechanisms for this remain to be clarified. In this study, we investigated how Aldo increases OPN transcripts in the vascular smooth muscle cells of rats. Aldosterone increased OPN transcripts time-dependently as well as dose-dependently. This increase was diminished by eplerenone, a mineralocorticoid receptor (MR) antagonist. Luciferase promoter assays showed that the OPN promoter deleted to the -1599 site retained the same promoting ability as the full-length OPN promoter when stimulated by 10(-7) M Aldo, but the promoter deleted to the -1300 site lost the promoting ability. A glucocorticoid response element (GRE) is located in that deleted region. Luciferase assays of a mutated promoter without the GRE lost the luciferase upregulation, although mutated promoters with the deletion of other consensus sites maintained the promoter activity. The binding of the Aldo-MR complex to the GRE fragment was confirmed by an electrophoretic-mobility shift assay. This is the first report showing that Aldo regulates the transcriptional levels of OPN and inflammatory responses in the vasculature through a specific GRE site in the OPN promoter region.
Collapse
Affiliation(s)
- Arihiro Kiyosue
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Maybin JA, Battersby S, Hirani N, Nikitenko LL, Critchley HOD, Jabbour HN. The expression and regulation of adrenomedullin in the human endometrium: a candidate for endometrial repair. Endocrinology 2011; 152:2845-56. [PMID: 21558311 PMCID: PMC3192419 DOI: 10.1210/en.2010-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 04/21/2011] [Indexed: 12/29/2022]
Abstract
After menstruation, the endometrium has a remarkable capacity for repair, but the factors involved remain undefined. We hypothesize adrenomedullin (AM) plays a role in this process. Premenstrually progesterone levels decline, stimulating prostaglandin (PG) synthesis, vasoconstriction, and hypoxia. This study aimed to determine 1) AM expression throughout the menstrual (M) cycle and 2) its regulation by PG and hypoxia. Human endometrial biopsies (n = 51) were collected with ethical approval and consent. AM mRNA expression was examined by quantitative RT-PCR and was found to be selectively elevated in endometrium from the menstrual (M) phase (P < 0.001). AM immunohistochemical staining was maximal in M and proliferative (P) endometrium. Culture of secretory, but not P, explants with 100 nm PGF(2α) or hypoxia (0.5% O2) increased AM mRNA (P < 0.05). P explants were induced to increase AM expression using in vitro progesterone withdrawal but required the presence of hypoxia (P < 0.05). Short hairpin sequences against hypoxia-inducible factor-1α (HIF-1α) inhibited AM hypoxic up-regulation but did not alter PGF(2α)-induced expression. The AM receptor was immunolocalized to endothelial cells in both lymphatic and blood vessels. Conditioned medium from PGF(2α)-treated cells increased endothelial cell proliferation and branching (P < 0.05). This was abolished by AM receptor antagonists. In conclusion, AM is elevated at the time of endometrial repair and induces both angiogenesis and lymphangiogenesis by stimulating endothelial cell proliferation and tube formation. In the human endometrium, AM expression is up-regulated by two mechanisms: a HIF-1α-mediated hypoxic induction and a HIF-1α-independent PGF(2α) pathway. These physiological mechanisms may provide novel therapeutic targets for disorders such as heavy menstrual bleeding.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- Centre for Reproductive Biology, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
The human endometrium is exposed to repeated inflammation every month, culminating in tissue breakdown and menstruation. Subsequently, the endometrium has a remarkable capacity for efficient repair and remodeling to enable implantation if fertilization takes place. Endometrial function is known to be governed by the ovarian hormones estradiol and progesterone. This review paper focuses on hormonal control of the cyclical tissue injury and repair that takes place in the local endometrial environment at the time of menstruation. Progesterone levels decline premenstrually as the corpus luteum regresses in the absence of pregnancy, and estradiol levels increase during the postmenstrual phase. The functional impact of these significant changes is discussed, including their immediate and downstream effects. Finally, we examine the contribution of aberrant endometrial function to the presentation of heavy menstrual bleeding and identify potential therapeutic targets for the treatment of this common gynecological problem.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- The University of Edinburgh's Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, UK
| | | |
Collapse
|
21
|
Abstract
Central and peripheral neurons as well as neuroendocrine cells express a variety of neurotransmitters/modulators that play critical roles in regulation of physiological systems. The synthesis of several neurotransmitters/modulators is regulated by O(2)-requiring rate-limiting enzymes. Consequently, hypoxia resulting from perturbations in O(2) homeostasis can affect neuronal functions by altering neurotransmitter synthesis. Two broad categories of hypoxia are frequently encountered: continuous hypoxia (CH) and intermittent hypoxia (IH). CH is often seen during high altitude sojourns, whereas IH is experienced in sleep-disordered breathing with recurrent apneas (i.e., brief, repetitive cessations of breathing). This article presents what is currently known on the effects of both forms of hypoxia on neurotransmitter levels and neurotransmitter synthesizing enzymes in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Ganesh K Kumar
- Institute for Integrative Physiology and Center for Systems Biology of O(2) Sensing, Biological Sciences Division, University of Chicago, Illinois, USA.
| |
Collapse
|
22
|
Marinoni E, Pacioni K, Sambuchini A, Moscarini M, Letizia C, DI Iorio R. Regulation by hypoxia of adrenomedullin output and expression in human trophoblast cells. Eur J Obstet Gynecol Reprod Biol 2010; 154:146-50. [PMID: 21094578 DOI: 10.1016/j.ejogrb.2010.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 09/06/2010] [Accepted: 10/13/2010] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Plasma adrenomedullin concentrations are increased in the fetal circulation in acute and chronic hypoxic conditions. The effect of hypoxia in regulating adrenomedullin synthesis and secretion was investigated in human placental trophoblast cells. STUDY DESIGN Human trophoblast cells obtained from term placentas (n = 7) were cultured in hypoxic condition (3% oxygen). Cytotrophoblast cells were cultured for up to 48 h and syncytiotrophoblasts for 2, 8 and 24 h. Changes in adrenomedullin output compared to normoxic conditions were measured by radioimmunoassay. Protein expression was evaluated with Western blot and immunocytochemistry. RESULTS Hypoxia induced a time-dependent increase in adrenomedullin output and protein expression by placental trophoblast cells. CONCLUSIONS Hypoxia regulates adrenomedullin secretion and expression by human placenta, thereby promoting increased adrenomedullin concentration in the fetal circulation in clinical circumstances characterized by reduced oxygen levels.
Collapse
Affiliation(s)
- Emanuela Marinoni
- Center for Scientific Research, San Pietro Hospital, Fatebenefratelli, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Yang J, Wu R, Zhou M, Wang P. Human adrenomedullin and its binding protein ameliorate sepsis-induced organ injury and mortality in jaundiced rats. Peptides 2010; 31:872-7. [PMID: 20132852 PMCID: PMC2854200 DOI: 10.1016/j.peptides.2010.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 01/21/2010] [Accepted: 01/22/2010] [Indexed: 01/11/2023]
Abstract
Sepsis is a serious complication for patients with obstructive jaundice. Although administration of adrenomedullin (AM) in combination with its binding protein (AMBP-1) is protective after injury, it remains unknown whether AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in the setting of biliary obstruction. The aim of this study is, therefore, to test the efficacy of human AM/AMBP-1 in a rat model of obstructive jaundice and polymicrobial sepsis. To study this, obstructive jaundice was induced in male adult rats (275-325g) by common bile duct ligation (BDL). One week after BDL, the rats were subjected to sepsis by cecal ligation and puncture (CLP). Plasma levels of AM and AMBP-1 were measured at 20h after CLP. In additional groups of BDL+CLP rats, human AM/AMBP-1 (24/80microg/kg body weight (BW)) or vehicle (i.e., human albumin) was administered intravenously at 5h after CLP. Blood and tissue samples were collected at 20h after CLP for various measurements. To determine the long-term effect of human AM/AMBP-1 after BDL+CLP, the gangrenous cecum was removed at 20h after CLP and 7-day survival was recorded. Our results showed that plasma levels of AM were significantly increased while AMBP-1 levels were markedly decreased after BDL+CLP (n=8, P<0.05). Administration of human AM/AMBP-1 attenuated tissue injury and inflammatory responses after BDL+CLP. Moreover, human AM/AMBP-1 significantly increased the survival rate from 21% (n=14) to 53% (n=15). Thus, human AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in jaundiced rats. Human AM/AMBP-1 can be further developed as a novel treatment for sepsis in jaundiced patients.
Collapse
Affiliation(s)
- Juntao Yang
- Department of Surgery, North Shore University Hospital and Long Island Jewish Medical Center, Manhasset, NY 11030, United States
| | | | | | | |
Collapse
|
24
|
Human adrenomedullin and its binding protein attenuate organ injury and reduce mortality after hepatic ischemia-reperfusion. Ann Surg 2009; 249:310-7. [PMID: 19212187 DOI: 10.1097/sla.0b013e3181961d43] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To determine whether administration of a vasoactive peptide, human adrenomedullin (AM), in combination with its binding protein (ie, AMBP-1), prevents or minimizes hepatic ischemia-reperfusion (I/R) injury. SUMMARY BACKGROUND DATA Hepatic I/R injury results from tissue hypoxia and subsequent inflammatory responses. Even though numerous pharmacological modalities and substances have been studied to reduce I/R-induced mortality, none have been entirely successful. We have shown that administration of AM/AMBP-1 produces significant beneficial effects under various pathophysiological conditions. However, it remains unknown if human AM/AMBP-1 has any protective effects on hepatic I/R-induced tissue damage and mortality. METHODS Seventy percent hepatic ischemia was induced in male adult rats by placing a microvascular clip across the hilum of the left and median lobes for 90 minutes. After removing the clip, human AM alone, human AMBP-1 alone, human AM in combination with human AMBP-1 or vehicle was administered intravenously over a period of 30 minutes. Blood and tissue samples were collected 4 hours after reperfusion for various measurements. In additional groups of animals, the nonischemic liver lobes were resected at the end of 90-minute ischemia. The animals were monitored for 7 days and survival was recorded. RESULTS After hepatic I/R, plasma levels of AM were significantly increased, whereas AMBP-1 levels were markedly decreased. Likewise, gene expression of AM in the liver was increased significantly, whereas AMBP-1 expression was markedly decreased. Administration of AM in combination with AMBP-1 immediately after the onset of reperfusion down-regulated inflammatory cytokines, decreased hepatic neutrophil infiltration, inhibited liver cell apoptosis and necrosis, and reduced liver injury and mortality in a rat model of hepatic I/R. On the other hand, administration of human AM alone or human AMBP-1 alone after hepatic I/R failed to produce significant protection. CONCLUSIONS Human AM/AMBP-1 may be a novel treatment to attenuate tissue injury after an episode of hepatic ischemia.
Collapse
|
25
|
Gazzolo D, Abella R, Marinoni E, di Iorio R, Li Volti G, Galvano F, Frigiola A, Temporini F, Moresco L, Colivicchi M, Sabatini M, Ricotti A, Strozzi MC, Crivelli S, Risso FM, Sannia A, Florio P. New markers of neonatal neurology. J Matern Fetal Neonatal Med 2009; 22 Suppl 3:57-61. [PMID: 19718579 DOI: 10.1080/14767050903181468] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hypoxia-ischemia (H-I) constitutes the main phenomenon responsible for brain-blood barrier permeability modifications leading to cerebral vascular auto-regulation loss in newborns. Hypotension, cerebral ischemia, and reperfusion are the main events involved in vascular auto-regulation loss leading to cell death and tissue damage. Reperfusion could be critical since organ damage, particularly of the brain, may be amplified during this period. An exaggerated activation of vasoactive agents, of calcium mediated effects could be responsible for reperfusion injury (R-I), which, in turns, leads to cerebral hemorrhage and damage. These phenomena represent a common repertoire in newborns complicated by perinatal acute or chronic hypoxia treated by risky procedures such as mechanical ventilation, nitric oxide supplementation, brain cooling, and extracorporeal membrane oxygenation (ECMO). Despite accurate monitoring, the post-insult period is crucial, as clinical symptoms and standard monitoring parameters may be silent at a time when brain damage is already occurring and the therapeutic window for pharmacological intervention is limited. Therefore, the measurement of circulating biochemical markers of brain damage, such as vasoactive agents and nervous tissue peptides is eagerly awaited in clinical practice to detect high risk newborns. The present review is aimed at investigating the role of biochemical markers such as adrenomedullin, a vasoactive peptide; S100B, a calcium binding protein, activin A, a glycoprotein, in the cascade of events leading to I-R injury in newborns complicated by perinatal asphyxia.
Collapse
Affiliation(s)
- Diego Gazzolo
- Department of Pediatrics and Neuroscience, G. Gaslini Children's Hospital University of Genoa, Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wu R, Dong W, Qiang X, Ji Y, Cui T, Yang J, Zhou M, Blau S, Marini CP, Ravikumar TS, Wang P. Human vasoactive hormone adrenomedullin and its binding protein rescue experimental animals from shock. Peptides 2008; 29:1223-30. [PMID: 18403050 PMCID: PMC2488201 DOI: 10.1016/j.peptides.2008.02.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 02/26/2008] [Accepted: 02/28/2008] [Indexed: 10/22/2022]
Abstract
We recently discovered that vascular responsiveness to adrenomedullin (AM), a vasoactive hormone, decreases after hemorrhage, which is markedly improved by the addition of its binding protein AMBP-1. One obstacle hampering the development of AM/AMBP-1 as resuscitation agents in trauma victims is the potential immunogenicity of rat proteins in humans. Although less potent than rat AM, human AM has been shown to increase organ perfusion in rats. We therefore hypothesized that administration of human AM/AMBP-1 improves organ function and survival after severe blood loss in rats. To test this, male Sprague-Dawley rats were bled to and maintained at an MAP of 40 mmHg for 90 min. They were then resuscitated with an equal volume of shed blood in the form of Ringer's lactate (i.e., low-volume resuscitation) over 60 min. At 15 min after the beginning of resuscitation, human AM/AMBP-1 (12/40 or 48/160 microg/kg BW) were administered intravenously over 45 min. Various pathophysiological parameters were measured 4h after resuscitation. In additional groups of animals, a 12-day survival study was conducted. Our result showed that tissue injury as evidenced by increased levels of transaminases, lactate, and creatinine, was present at 4h after hemorrhage and resuscitation. Moreover, pro-inflammatory cytokines TNF-alpha and IL-6 were also significantly elevated. Administration of AM/AMBP-1 markedly attenuated tissue injury, reduced cytokine levels, and improved the survival rate from 29% (vehicle) to 62% (low-dose) or 70% (high-dose). However, neither human AM alone nor human AMBP-1 alone prevented the significant increase in ALT, AST, lactate and creatinine at 4h after the completion of hemorrhage and resuscitation. Moreover, the half-life of human AM and human AMBP-1 in rats was 35.8 min and 1.68 h, respectively. Thus, administration of human AM/AMBP-1 may be a useful approach for attenuating organ injury, and reducing mortality after hemorrhagic shock.
Collapse
Affiliation(s)
- Rongqian Wu
- Department of Surgery, North Shore University Hospital and Long Island Jewish Medical Center, Manhasset, NY 11030, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Li Y, Jiang C, Wang X, Zhang Y, Shibahara S, Takahashi K. Adrenomedullin is a novel adipokine: adrenomedullin in adipocytes and adipose tissues. Peptides 2007; 28:1129-43. [PMID: 17433499 DOI: 10.1016/j.peptides.2007.03.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 03/01/2007] [Accepted: 03/01/2007] [Indexed: 01/23/2023]
Abstract
Adrenomedullin (AM) is a multifunctional regulatory peptide that is produced and secreted by various types of cells. The production and the secretion of AM have been demonstrated in cultured adipocytes and adipose tissues. Inflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and lipopolysaccharide are strong stimulators for AM expression in adipocytes. Furthermore, AM expression in the adipose tissue is increased in obesity, and plasma concentrations of AM are increased in obese subjects. One possible (patho)physiological role of AM secreted by adipose tissue may be actions against complications of the metabolic syndrome characterized by obesity, type 2 diabetic mellitus and hypertension, via its antioxidant and potent vasodilator effects. These findings indicate that AM is a new member of the adipokine family.
Collapse
Affiliation(s)
- Yin Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100083, PR China
| | | | | | | | | | | |
Collapse
|
28
|
Wu R, Dong W, Zhou M, Simms HH, Marini CP, Ravikumar TS, Wang P. Adrenomedullin and adrenomedullin binding protein-1 prevent metabolic acidosis after uncontrolled hemorrhage in rats. Crit Care Med 2007; 35:912-8. [PMID: 17255858 DOI: 10.1097/01.ccm.0000257327.61829.34] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Management of trauma victims with uncontrolled hemorrhage remains a major problem in combat casualty care at the far-forward battlefield setting. The neuroendocrine response to hemorrhage is to maintain perfusion to the heart and brain, often at the expense of other organ systems. Decreased organ perfusion after hemorrhagic shock is associated with metabolic acidosis, in which the up-regulated endothelin-1 plays an important role. We have recently shown that vascular responsiveness to adrenomedullin (AM), a newly discovered vasodilator peptide, is depressed after hemorrhage and resuscitation. Down-regulation of AM binding protein (AMBP-1) appears to be responsible for this hyporesponsiveness. We therefore hypothesized that administration of AM/AMBP-1 would prevent metabolic acidosis after uncontrolled hemorrhage via down-regulation of endothelin-1. DESIGN Prospective, controlled, and randomized animal study. SETTING A research institute laboratory. SUBJECTS Male Sprague-Dawley rats (275-325 g). INTERVENTIONS A rat model of uncontrolled hemorrhage with an extremely low volume of fluid resuscitation was used to mimic the combat situation. MEASUREMENTS AND MAIN RESULTS Both lumbar veins of male adult rats were isolated and severed at the junction to the vena cava. The abdomen was kept open but covered with a saline wet gauze for 45 mins and then closed in layers. The animals received 1 mL of normal saline (vehicle) with or without AM (12 microg/kg of body weight) and AMBP-1 (40 microg/kg of body weight) over 45 mins. Various variables were measured at 4 hrs after resuscitation. The bleed-out volumes in the vehicle group and the AM/ AMBP-1 treatment group were 6.78 +/- 0.19 and 6.81 +/- 0.25 mL/rat, respectively. The results indicate that AM/AMBP-1 administration prevented metabolic acidosis, mitigated organ injury, down-regulated preproendothelin-1 gene expression, and decreased plasma levels of endothelin-1 after hemorrhage. CONCLUSIONS AM/AMBP-1 may provide a novel approach for the treatment of uncontrolled hemorrhage. The beneficial effect of AM/AMBP-1 is associated with down-regulation of endothelin-1.
Collapse
Affiliation(s)
- Rongqian Wu
- Department of Surgery, North Shore University Hospital and Long Island Jewish Medical Center, Manhasset, NY, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Abe M, Sata M, Suzuki E, Takeda R, Takahashi M, Nishimatsu H, Nagata D, Kangawa K, Matsuo H, Nagai R, Hirata Y. Effects of adrenomedullin on acute ischaemia-induced collateral development and mobilization of bone-marrow-derived cells. Clin Sci (Lond) 2006; 111:381-7. [PMID: 16922679 DOI: 10.1042/cs20060137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adrenomedullin exerts not only vasodilatory effects, but also angiogenic effects. In the present study, we investigated the effects of adrenomedullin on collateral formation and circulating bone-marrow-derived cells after acute tissue ischaemia. Bone marrow of 8–10-week-old female C57BL/6J mice was replaced with that from GFP (green fluorescent protein) transgenic mice (GFP mice). At 8 weeks after transplantation, hindlimb ischaemia was induced by resecting the right femoral artery and a plasmid expressing human adrenomedullin (50 μg) was injected into the ischaemic muscle, followed by in vivo electroporation on a weekly basis. Overexpression of adrenomedullin significantly enhanced the blood flow recovery compared with controls (blood flow ratio, 1.0±0.2 compared with 0.6±0.3 respectively, at week 4; P<0.05) and increased capillary density in the ischaemic leg as determined by anti-CD31 immunostaining of the ischaemic muscle (567±40 compared with 338±65 capillaries/mm2 respectively, at week 5; P<0.05). There were more GFP-positive cells in the thigh muscle of the mice injected with adrenomedullin than in that of the control mice (29.6±4.5 compared with 16.5±3.3 capillaries/mm2 respectively, at week 5; P<0.05). We repeated the same experiments using LacZ-knock-in mice instead of GFP mice, and obtained similar results. These findings suggest that adrenomedullin may augment ischaemia-induced collateral formation with some effects on circulating bone-marrow-derived cells.
Collapse
Affiliation(s)
- Minami Abe
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nagata D, Takahashi M, Sawai K, Tagami T, Usui T, Shimatsu A, Hirata Y, Naruse M. Molecular mechanism of the inhibitory effect of aldosterone on endothelial NO synthase activity. Hypertension 2006; 48:165-71. [PMID: 16754797 DOI: 10.1161/01.hyp.0000226054.53527.bb] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the proinflammatory and profibrotic actions of aldosterone (Aldo) on the vasculature have been reported, the effects and molecular mechanisms of Aldo on endothelial function are yet to be determined. We investigated how Aldo regulates endothelial NO synthase (eNOS) function in human umbilical vein endothelial cells (HUVECs). HUVECs were incubated for 16 hours with Aldo 10(-7) mol/L. The concentration of reactive oxygen species was estimated by measuring 2',7'-dichlorodihydrofluorescein diacetate chemiluminescence. Signal transduction was estimated by Western immunoblots. Real-time RT-PCR was performed to measure expression of transcripts of endogenous GTP cyclohydrolase-1 and components of reduced nicotinamide-adenine dinucleotide phosphate oxidase. To eliminate the possible effect of the glucocorticoid receptor (GR) and to emphasize the role of mineralocorticoid receptor, we used GR small interfering RNA and knocked down GR expression in several experiments. NO output was estimated by intracellular cGMP concentration. Reactive oxygen species production increased significantly in Aldo-treated HUVECs but was abolished by pretreatment with eplerenone. Transcripts of p47(phox) were increased by Aldo treatment. Vascular endothelial growth factor-induced eNOS Ser 1177 but not Akt Ser 473 phosphorylation levels were reduced significantly by pretreatment with Aldo. Pretreatment with either eplerenone or okadaic acid restored phosphorylation levels of eNOS Ser 1177 in Aldo-treated cells, suggesting that protein phosphatase 2A was upregulated by Aldo via mineralocorticoid receptor. The decrease in NO output caused by Aldo pretreatment was reversed significantly by 5,6,7,8-tetrahydrobiopterin, GTP cyclohydrolase-1 overexpression, or p47(phox) knockdown. These results suggest that Aldo inhibits eNOS function through bimodal mechanisms of 5,6,7,8-tetrahydrobiopterin deficiency and protein phosphatase 2A activation.
Collapse
Affiliation(s)
- Daisuke Nagata
- Division of Endocrinology, National Hospital Organization Kyoto Medical Center, Research Institute, Kyoto, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Shi Y, Yoshihara F, Nakahama H, Ichimaru N, Yazawa K, Sada M, Goto R, Kawano Y, Moriyama T, Takahara S, Okuyama A, Kangawa K. A novel immunosuppressant FTY720 ameliorates proteinuria and alterations of intrarenal adrenomedullin in rats with autoimmune glomerulonephritis. ACTA ACUST UNITED AC 2005; 127:233-8. [PMID: 15680492 DOI: 10.1016/j.regpep.2004.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Accepted: 12/10/2004] [Indexed: 12/19/2022]
Abstract
FTY720 has been originally developed as a new immunosuppressive agent, which prolongs graft survival in organ transplantation. Adrenomedullin (AM) participates in the regulation of sodium homeostasis and has renoprotective effects. The possible involvement of renal AM in the pathophysiology of glomerulonephritis (GN) and the effect of FTY720 has been evaluated in rats. HgCl2 (1 mg/kg body weight) was inoculated subcutaneously 3 times/week for a total of 2 weeks. FTY720 (3 or 10 mg/kg) was inoculated subcutaneously daily. The proteinuria, urinary N-acetyl-beta-D-glucosaminidase (NAG) excretion and serum total cholesterol levels were increased and serum albumin level was reduced in rats with HgCl2-induced GN compared with controls. FTY720 reduced proteinuria (3 mg/kg: -25%; 10 mg/kg: -41%), urinary NAG excretion (-11%; -52%) and total cholesterol level (-21%; -55%) in a dose-dependent manner. Renal AM level and its mRNA expression were increased in rats with GN compared with controls (Peptide Cortex: +69%; Medulla: +82%; mRNA Cortex: +25%). Interestingly, FTY720 additionally increased these levels (Peptide Cortex: +38%; Medulla: +39%; mRNA Cortex: +20%). Renal AM levels correlated with urinary NAG excretion and creatinine clearance. These results suggest that FTY720 suppresses the renal damage in rats with GN and renal AM may participate in the pathophysiology of GN and the renoprotective effects of FTY720.
Collapse
Affiliation(s)
- Yi Shi
- Department of Urology, Osaka University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Julián M, Cacho M, García MA, Martín-Santamaría S, de Pascual-Teresa B, Ramos A, Martínez A, Cuttitta F. Adrenomedullin: a new target for the design of small molecule modulators with promising pharmacological activities. Eur J Med Chem 2005; 40:737-50. [PMID: 15927308 DOI: 10.1016/j.ejmech.2004.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2004] [Revised: 10/26/2004] [Accepted: 10/27/2004] [Indexed: 10/25/2022]
Abstract
Adrenomedullin (AM) is a 52-amino acid peptide with a pluripotential activity. AM is expressed in many tissues throughout the body, and plays a critical role in several diseases such as cancer, diabetes, cardiovascular and renal disorders, among others. While AM is a protective agent against cardiovascular disorders, it behaves as a stimulating factor in other pathologies such as cancer and diabetes. Therefore, AM is a new and promising target for the development of molecules which, through their ability to regulate AM levels, could be used in the treatment of these pathologies.
Collapse
Affiliation(s)
- Miguel Julián
- Departamento de Química, Facultad de Farmacia, Universidad San Pablo CEU, Urbanización Montepríncipe, 28668 Boadilla del Monte, Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Nishimatsu H, Suzuki E, Satonaka H, Takeda R, Omata M, Fujita T, Nagai R, Kitamura T, Hirata Y. Endothelial dysfunction and hypercontractility of vascular myocytes are ameliorated by fluvastatin in obese Zucker rats. Am J Physiol Heart Circ Physiol 2004; 288:H1770-6. [PMID: 15550522 DOI: 10.1152/ajpheart.00751.2004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
To study the mechanisms of vascular dysfunction in diabetes mellitus, we examined the responses of the aorta to adrenomedullin (AM) and ANG II in obese Zucker (OZ), lean Zucker (LZ), and OZ rats administered fluvastatin (OZ + Flu). AM-induced endothelium-dependent vasorelaxation was impaired in OZ rats compared with LZ rats, and fluvastatin restored AM-induced, endothelium-dependent vasorelaxation (%Deltatension at 10(-7) mol/l AM; LZ, -85.1 +/- 3.1%; OZ, -50.7 +/- 2.5%; OZ + Flu, -75.6 +/- 2.7%). Expression of endothelial nitric oxide synthase (eNOS) and Akt phosphorylation in response to AM (10(-7) mol/l) were also diminished in OZ rats. Fluvastatin restored the eNOS expression and Akt phosphorylation [eNOS expression (relative intensity): LZ, 2.3 +/- 0.4; OZ, 1.0 +/- 0.2; OZ + Flu, 1.8 +/- 0.3; Akt phosphorylation (relative intensity): LZ, 2.3 +/- 0.2; OZ, 1.0 +/- 0.3; OZ + Flu, 1.9 +/- 0.2]. ANG II-induced vasoconstriction was enhanced in the aortic rings of OZ rats compared with LZ rats, and this enhanced vasoconstriction was partially normalized by fluvastatin and was abolished when the aorta of OZ rats was preincubated with the Rho kinase inhibitor Y-27632. GTPgammaS-induced contraction of permeabilized aortic smooth muscle cells, which is an indicator of the Rho-dependent Ca(2+) sensitization of contraction, was enhanced in OZ rats compared with LZ rats, and this enhanced contraction was suppressed in OZ + Flu rats. These results suggested that endothelium-dependent vasorelaxation was impaired, Ca(2+) sensitization of contraction was augmented in blood vessels of OZ rats and that fluvastatin restored vascular function by activating the Akt-dependent pathway and inhibiting the Rho-dependent pathway.
Collapse
MESH Headings
- Adrenomedullin
- Angiotensin II/pharmacology
- Animals
- Aorta/drug effects
- Aorta/physiology
- Calcium/metabolism
- Diabetic Angiopathies/drug therapy
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Fatty Acids, Monounsaturated/pharmacology
- Fluvastatin
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Indoles/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type III
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/physiopathology
- Peptides/pharmacology
- Phosphorylation/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Rats
- Rats, Zucker
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
- Vasoconstrictor Agents/pharmacology
- Vasodilation/drug effects
- Vasodilation/physiology
- Vasodilator Agents/pharmacology
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Hiroaki Nishimatsu
- Department of Urology, Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Suzuki Y, Horio T, Nonogi H, Hayashi T, Kitamura K, Eto T, Kangawa K, Kawano Y. Adrenomedullin as a sensitive marker for coronary and peripheral arterial complications in patients with atherosclerotic risks. Peptides 2004; 25:1321-6. [PMID: 15350700 DOI: 10.1016/j.peptides.2004.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Revised: 04/23/2004] [Accepted: 04/23/2004] [Indexed: 11/26/2022]
Abstract
Plasma adrenomedullin (AM) levels are elevated in various pathological states including cardiovascular and inflammatory diseases. The present study investigated whether an increased AM level is a marker of vascular complications in patients with atherosclerotic risks. In 114 patients with cardiovascular risks and/or diseases including ischemic heart disease (IHD) and peripheral arterial disease (PAD), plasma AM concentration and other inflammatory markers such as high sensitive C-reactive protein (CRP) and interleukin (IL)-6 were examined. The plasma AM level was not altered by the absence or presence of each of four major risk factors, i.e., hypertension, diabetes mellitus, hyperlipidemia, and smoking and its level was not significantly correlated with blood pressure, plasma glucose, or serum lipid levels. The patients with IHD had a significantly higher concentration of plasma AM than those without IHD. The AM level in subjects with PAD was also increased significantly compared with those without PAD. The plasma AM was strongly correlated with inflammatory parameters such as CRP and IL-6. Among AM, CRP, and IL-6, however, only AM was an independent predictor for both IHD and PAD by multiple logistic regression analysis. Our findings suggest the possibility that plasma AM is a novel sensitive marker for the presence of vascular lesions in patients with atherosclerotic risks.
Collapse
Affiliation(s)
- Yoshihiko Suzuki
- Department of Medicine, National Cardiovascular Center, 5-7-1, Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Nagata D, Takeda R, Sata M, Satonaka H, Suzuki E, Nagano T, Hirata Y. AMP-activated protein kinase inhibits angiotensin II-stimulated vascular smooth muscle cell proliferation. Circulation 2004; 110:444-51. [PMID: 15262850 DOI: 10.1161/01.cir.0000136025.96811.76] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is a stress-activated protein kinase that works as a metabolic sensor of cellular ATP levels. Here, we investigated whether AMPK signaling has a role in the regulation of the angiotensin II (Ang II)-induced proliferation signal in rat vascular smooth muscle cells (VSMCs). METHODS AND RESULTS Aminoimidazole-4-carboxamide-1-beta-ribofuranoside (AICAR) activated AMPK in rat VSMCs and inhibited Ang II-induced extracellular signal-regulated kinase 1/2 phosphorylation but not that of p38 MAPK or Akt/PKB. Although Ang II activated AMPK, this activation was significantly inhibited by catalase, N-acetylcysteine, and diphenyleneiodonium chloride, an NADPH oxidase inhibitor. Moreover, the observation that AMPK was activated by H2O2 suggests that AMPK is redox sensitive. The Ang II type 1 receptor antagonist valsartan but not the Ang II type 2 receptor antagonist PD123319 significantly inhibited Ang II-induced AMPK activation, suggesting that Ang II-induced AMPK activation was Ang II type 1 receptor dependent. Whereas 3H-thymidine incorporation by VSMCs treated with Ang II was significantly inhibited when the cells were pretreated with 1 mmol/L AICAR, the inhibition of AMPK by dominant-negative AMPK overexpression augmented Ang II-induced cell proliferation. Subcutaneous injection of AICAR (1 mg/g body weight per day) for 2 weeks suppressed neointimal formation after transluminal mechanical injury of the rat femoral artery. CONCLUSIONS Our findings indicate that Ang II-induced AMPK activation is synchronized with extracellular signal-regulated kinase signaling and that AMPK works as an inhibitor of the Ang II proliferative pathway. AMPK signaling might serve as a new therapeutic target of vascular remodeling in cardiovascular diseases.
Collapse
Affiliation(s)
- Daisuke Nagata
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Sandner P, Hofbauer KH, Tinel H, Kurtz A, Thiesson HC, Ottosen PD, Walter S, Skøtt O, Jensen BL. Expression of adrenomedullin in hypoxic and ischemic rat kidneys and human kidneys with arterial stenosis. Am J Physiol Regul Integr Comp Physiol 2004; 286:R942-51. [PMID: 14715486 DOI: 10.1152/ajpregu.00274.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate regional aspects of hypoxic regulation of adrenomedullin (AM) in kidneys, we mapped the distribution of AM in the rat kidney after hypoxia (normobaric hypoxic hypoxia, carbon monoxide, and CoCl2 for 6 h), anemia (hematocrit lowered by bleeding) and after global transient ischemia for 1 h (unilateral renal artery occlusion and reperfusion for 6 and 24 h) and segmental infarct (6 and 24 h). AM expression and localization was determined in normal human kidneys and in kidneys with arterial stenosis. Hypoxia stimulated AM mRNA expression significantly in rat inner medulla (CO 13 times, 8% O2 6 times, and CoCl2 8 times), followed by the outer medulla and cortex. AM mRNA level was significantly elevated in response to anemia and occlusion-reperfusion. Immunoreactive AM was associated with the thin limbs of Henle's loop, distal convoluted tubule, collecting ducts, papilla surface epithelium, and urothelium. AM labeling was prominent in the inner medulla after CO and in the outer medulla after occlusion-reperfusion. The infarct border zone was strongly labeled for AM. In cultured inner medullary collecting duct cells, AM mRNA was significantly increased by hypoxia. AM mRNA was equally distributed in human kidney and AM was localized as in the rat kidney. In human kidneys with artery stenosis, AM mRNA was not significantly enhanced compared with controls, but AM immunoreactivity was observed in tubules, vessels, and glomerular cells. In summary, AM expression was increased in the rat kidney in response to hypoxic and ischemic hypoxia in keeping with oxygen gradients. AM was widely distributed in the human kidney with arterial stenosis. AM may play a significant role to counteract hypoxia in the kidney.
Collapse
Affiliation(s)
- Peter Sandner
- Dept. of Physiology and Pharmacology, University of Southern Denmark, Winsløwparken 21, No. 3, DK-5000 Odense, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Xu Q, Konta T, Nakayama K, Furusu A, Moreno-Manzano V, Lucio-Cazana J, Ishikawa Y, Fine LG, Yao J, Kitamura M. Cellular defense against H2O2-induced apoptosis via MAP kinase-MKP-1 pathway. Free Radic Biol Med 2004; 36:985-93. [PMID: 15059639 DOI: 10.1016/j.freeradbiomed.2004.01.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Revised: 12/01/2003] [Accepted: 01/15/2004] [Indexed: 11/21/2022]
Abstract
Mitogen-activated protein (MAP) kinase phosphatase-1 (MKP-1) is an oxidative stress-inducible gene. In this study, we investigated signaling pathways involved in oxidative stress-induced MKP-1 expression and its role in apoptosis of rat mesangial cells. Northern and Western blot analyses showed that H(2)O(2) induced expression of MKP-1 mRNA and protein in a dose-dependent manner, without affecting the stability of the transcript. H(2)O(2) induced phosphorylation of extracellular signal-regulated kinase, p38 MAP kinase, and c-Jun N-terminal kinase and consequently activated activator protein 1 (AP-1). Selective inhibitors of individual MAP kinases or a dominant-negative mutant of c-jun significantly suppressed the expression of MKP-1 by H(2)O(2). Inhibition of MKP-1 by a protein tyrosine phosphatase inhibitor (vanadate) enhanced H(2)O(2)-triggered apoptosis. Consistently, transfection with a wild-type MKP-1, but not its catalytically inactive mutant MKP-1CS, attenuated H(2)O(2)-induced apoptosis. These data elucidate, for the first time, that induction of MKP-1 by H(2)O(2) is mediated by the MAP kinase-AP-1 pathway and that the induced MKP-1 is involved in cellular defense against oxidative stress-induced apoptosis of mesangial cells.
Collapse
Affiliation(s)
- Qihe Xu
- Department of Medicine, Royal Free and University College Medical School, University College London, London, England, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nagata D, Mogi M, Walsh K. AMP-activated protein kinase (AMPK) signaling in endothelial cells is essential for angiogenesis in response to hypoxic stress. J Biol Chem 2003; 278:31000-6. [PMID: 12788940 DOI: 10.1074/jbc.m300643200] [Citation(s) in RCA: 289] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a stress-activated protein kinase that is regulated by hypoxia and other cellular stresses that result in diminished cellular ATP levels. Here, we investigated whether AMPK signaling in endothelial cells has a role in regulating angiogenesis. Hypoxia induced the activating phosphorylation of AMPK in human umbilical vein endothelial cells (HUVECs), and AMPK activation was required for the maintenance of pro-angiogenic Akt signaling under these conditions. Suppression of AMPK signaling inhibited both HUVEC migration to VEGF and in vitro differentiation into tube-like structures in hypoxic, but not normoxic cultures. Dominant-negative AMPK also inhibited in vivo angiogenesis in Matrigel plugs that were implanted subcutaneously in mice. These data identify AMPK signaling as a new regulator of angiogenesis that is specifically required for endothelial cell migration and differentiation under conditions of hypoxia. As such, endothelial AMPK signaling may be a critical determinant of blood vessel recruitment to tissues that are subjected to ischemic stress.
Collapse
Affiliation(s)
- Daisuke Nagata
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
39
|
Sarica K, Sari I, Balat A, Erbağci A, Yurtseven C, Yağci F, Karakök M. Evaluation of adrenomedullin levels in renal parenchyma subjected to extracorporeal shockwave lithotripsy. UROLOGICAL RESEARCH 2003; 31:267-71. [PMID: 12830337 DOI: 10.1007/s00240-003-0323-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2002] [Accepted: 03/25/2003] [Indexed: 11/26/2022]
Abstract
Despite its safety and efficacy, the traumatic effects of high-energy shock waves (HESW) on renal morphology and function during long-term follow-up have yet to be elucidated. Although the main target of shock waves is the stone located in the kidney, the surrounding tissue and other organs are also subjected to trauma during this procedure. In contrast to renal blood flow evaluation after shock wave treatment, ischemic development, causing varying degrees of damage at the tissue level, has not been well evaluated. The renoprotective peptide adrenomedullin (AM) is a potent vasorelaxing, natriuretic and cell growth modulating peptide, which is thought to act as an autocrine/paracrine regulator in renal glomeruli and tubules. In this experimental study, renal parenchymal AM levels were assessed in an attempt to evaluate the effect of HESW on the tissue levels of this peptide, which may be responsible for the regulation of ischemia induced by extracorporeal shock wave lithotripsy(ESWL), in a rabbit model. Thirty white New Zealand rabbits, each weighing 3-5 kg were used. The animals were divided into three main groups, and varying numbers of shock waves (1000, 1500, 2000) were applied under fluoroscopic localization to the same kidney of all animals. Ketamine HCl anesthesia was administered (15-20 mg/kg) and all of the procedures were performed with a Multimed 2000 lithotriptor. Untreated contralateral kidneys were evaluated as controls. Following HESW application, the treated and untreated kidneys of each animal were removed through bilateral flank incisions under ketamine HCl anesthesia after 24 h and 7 days, respectively. Tissue AM levels were assessed with immunohistochemistry. During the early follow-up period (24 h), both treated and untreated kidneys showed a moderate to high degree of AM positivity. The number of tubules stained with AM increased as the number of shock waves increased and the expression of this protein became evident, possibly due to a higher degree of tissue damage. Additionally, a limited degree of AM positivity was noted in the contralateral kidneys although this was not as evident as the positivity seen in the treated kidneys. Assessment of tissue AM levels during late follow-up (7 days) in both kidneys demonstrated a moderate or limited degree of positivity in the treated kidneys. Limited or no positivity could be demonstrated in the contralateral kidneys at this time. Taking the certain traumatic effects of HESW, which causes transient ischemia during ESWL, into account, we conclude that the application of HESW results in a transient decrease in renal perfusion, causing ischemic injury in treated as well as in contralateral (untreated) kidneys. This ischemic event lasts for a short time and seemed to be dose- and time-dependent. Increased tissue levels of AM appear to be a potential defence against ESWL induced ischemia.
Collapse
Affiliation(s)
- Kemal Sarica
- Department of Urology, Sahinbey Medical Center, University of Gaziantep, Medical School, 27070 Kolejtepe/Gaziantep, Turkey.
| | | | | | | | | | | | | |
Collapse
|
40
|
Nikitenko LL, Smith DM, Bicknell R, Rees MCP. Transcriptional regulation of the CRLR gene in human microvascular endothelial cells by hypoxia. FASEB J 2003; 17:1499-501. [PMID: 12824306 DOI: 10.1096/fj.02-0993fje] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Adrenomedullin is a 52 amino acid peptide that shows a remarkable range of effects on the vasculature that include inter alia, vasodilatation, regulation of permeability, inhibition of endothelial cell apoptosis, and promotion of angiogenesis. Recently the G-protein coupled receptor (GPCR) calcitonin receptor-like receptor (CRLR), and receptor activity modifying proteins (RAMPs) have become recognized as integral components of the adrenomedullin signaling system. However, mechanisms of regulation of CRLR expression are still largely unknown. This is in part due to lack of information on the gene promoter. In this study we have determined the transcriptional start of human CRLR cDNA by 5'-RACE and cloned the proximal 5'-flanking region of the gene by PCR. The 2318 bp genomic fragment contains the basal promoter of human CRLR, including potential TATA-boxes and several GC boxes. Regulatory elements binding known transcription factors, such as Sp-1, Pit-1, glucocorticoid receptor, and hypoxia-inducible factor-1 alpha (HIF-1alpha) were also identified. When cloned into reporter gene vectors, the genomic fragment showed significant promoter activity, indicating that the 5'-flanking region isolated by PCR contains the gene promoter of human CRLR. Of significance is that the cloned promoter fragments were activated by hypoxia when transfected in primary microvascular endothelial cells. Site-directed mutagenesis of the consensus hypoxia-response element (HRE) in the 5'-flanking region abolished such a response. We also demonstrated by semi-quantitative RT-PCR that transcription of the gene is activated by hypoxia in microvascular endothelial cells. In contrast, expression of RAMPs 1, 2, and 3 was unaffected by low oxygen tension. We conclude that simultaneous transcriptional up-regulation of CRLR and its ligand adrenomedullin in endothelial cells could lead to a potent survival loop and therefore might play a significant role in vascular responses to hypoxia and ischemia.
Collapse
Affiliation(s)
- Leonid L Nikitenko
- NDOG, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | | | | | | |
Collapse
|
41
|
Abe M, Sata M, Nishimatsu H, Nagata D, Suzuki E, Terauchi Y, Kadowaki T, Minamino N, Kangawa K, Matsuo H, Hirata Y, Nagai R. Adrenomedullin augments collateral development in response to acute ischemia. Biochem Biophys Res Commun 2003; 306:10-5. [PMID: 12788059 DOI: 10.1016/s0006-291x(03)00903-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Expression of adrenomedullin, discovered as a vasodilatory peptide, is markedly up-regulated under pathological conditions such as tissue ischemia and inflammation, which are associated with neovascularization. Here, we tested the hypothesis that overly expressed adrenomedullin may augment collateral flow to ischemic tissues. We induced hindlimb ischemia in wild-type mice and injected a naked plasmid expressing human adrenomedullin or an empty vector into the ischemic muscle, followed by in vivo electroporation. Adrenomedullin markedly enhanced blood flow recovery as determined by Laser Doppler imaging. The mice treated with an empty vector suffered frequent autoamputation of the ischemic toe, which was completely prevented by adrenomedullin. Anti-CD31 immunostaining revealed that adrenomedullin significantly increased capillary density. The angiogenic effect of adrenomedullin was abrogated in endothelial nitric oxide synthase (eNOS)-deficient mice. These results indicate that adrenomedullin may promote collateral growth in response to ischemia through activation of eNOS.
Collapse
Affiliation(s)
- Minami Abe
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, 113-8655, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
López J, Martínez A. Cell and molecular biology of the multifunctional peptide, adrenomedullin. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 221:1-92. [PMID: 12455746 DOI: 10.1016/s0074-7696(02)21010-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adrenomedullin (AM) is a recently discovered regulatory peptide involved in many functions including vasodilatation, electrolyte balance, neurotransmission, growth, and hormone secretion regulation, among others. This 52-amino acid peptide is expressed by specific cell types in many organs throughout the body. A complex receptor system has been described for AM; it requires at least the presence of a seven-transmembrane-domain G-protein-coupled receptor, a single-transmembrane-domain receptor activity modifying protein, and a receptor component protein needed to establish the connection with the downstream signal transduction pathway, which usually involves cyclicAMP. In addition, a serum-binding protein regulates the biological actions of AM, frequently by increasing AM functional attributes. Changes in levels of circulating AM correlate with several critical diseases, including cardiovascular and renal disorders, sepsis, cancer, and diabetes. Whether AM is a causal agent, a protective reaction, or just a marker for these diseases is currently under investigation. New technologies seeking to elevate and/or reduce AM levels are being investigated as potential therapeutic avenues.
Collapse
Affiliation(s)
- José López
- Cell Biology Unit, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | | |
Collapse
|
43
|
Xu Q, Konta T, Furusu A, Nakayama K, Lucio-Cazana J, Fine LG, Kitamura M. Transcriptional induction of mitogen-activated protein kinase phosphatase 1 by retinoids. Selective roles of nuclear receptors and contribution to the antiapoptotic effect. J Biol Chem 2002; 277:41693-700. [PMID: 12186877 DOI: 10.1074/jbc.m207095200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
All-trans-retinoic acid (t-RA) inhibits hydrogen peroxide (H(2)O(2))-induced apoptosis by inhibiting the c-Jun N-terminal kinase (JNK)-activator protein 1 (AP-1) pathway. In this report, we examined the involvement of mitogen-activated protein kinase phosphatase 1 (MKP-1) in suppression of JNK and the antiapoptotic effect of t-RA and the roles of nuclear receptors in the regulation of MKP-1 by t-RA. We found that not only t-RA, but also a selective agonist of retinoic acid receptor (RAR), a selective agonist of retinoid X receptor (RXR), and a pan-agonist of RAR and RXR all induced MKP-1 at the transcriptional level. Activation of RAR was required for all of these triggering effects, but activation of RXR was required only for the RXR agonist-induced MKP-1 expression. Among the three RAR subtypes, RARalpha and RARgamma, but not RARbeta, mediated the t-RA-induced MKP-1 expression. The antiapoptotic effect of t-RA on H(2)O(2)-induced apoptosis in several cell types was correlated with the inducibility of MKP-1 by t-RA. Inhibition of MKP-1 by vanadate enhanced JNK phosphorylation and attenuated the antiapoptotic effect of t-RA. Furthermore, overexpression of MKP-1 inhibited H(2)O(2)-induced JNK phosphorylation and apoptosis. To our knowledge, this is the first to demonstrate that 1) MKP-1 is inducible by retinoids at the transcriptional level, 2) RXR and individual RAR subtypes have different roles in this process, and 3) the induced MKP-1 plays a significant role in mediating both JNK inhibition and the antiapoptotic effect of t-RA in oxidative stress.
Collapse
Affiliation(s)
- Qihe Xu
- Department of Medicine, Royal Free and University College Medical School, University College London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
44
|
Kanozawa K, Shimosawa T, Nagasawa R, Matsuda A, Kato H, Matsumura O, Mitarai T, Isoda K, Fujita T. Mature form of adrenomedullin is a useful marker to evaluate blood volume in hemodialysis patients. Am J Kidney Dis 2002; 40:794-801. [PMID: 12324915 DOI: 10.1053/ajkd.2002.35691] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We investigated the pathophysiological significance of the mature form of adrenomedullin (AM) in hemodialysis (HD) patients. Thirty-nine HD patients were enrolled and divided into two groups: those undergoing ultrafiltration (UF) during an HD session, group I; and those not undergoing UF, group II. We measured mature AM, atrial natriuretic peptide (ANP), endothelin-1, nitric oxide, cyclic guanosine 3',5'-monophosphate, and catecholamine levels at 1-hour intervals during HD sessions. On-line optical measurement of hematocrit was used to estimate change in blood volume during HD. In group II, blood volume did not change significantly during HD, nor did plasma mature AM concentrations estimated at the beginning and end of the HD treatment (3.0 +/- 0.3 and 2.8 +/- 0.2 fmol/mL, respectively). However, blood volume decreased significantly in group I patients (-7.3% +/- 0.6%), as did plasma mature AM concentrations (from 4.4 +/- 0.3 to 3.1 +/- 0.3 fmol/mL; P < 0.01). In contrast to mature AM, plasma ANP concentrations declined in both groups (from 193 +/- 32 to 87 +/- 14 pg/mL in group I and 67 +/- 12 to 46 +/- 8 pg/mL in group II). We conclude that mature AM is a useful marker to evaluate circulating blood volume in HD patients. Circulating blood volume may regulate the conversion of AM from the inactive to the mature form.
Collapse
Affiliation(s)
- Koichi Kanozawa
- Department of Internal Medicine, Saitama Medical Center, Saitama Medical School, Kawagoe, Saitama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Suzuki E, Nishimatsu H, Nagata D, Satonaka H, Goto A, Omata M, Fujita T, Nagai R, Hirata Y. Constitutive activation of proto-oncogen protein p21 induces cell cycle arrest in the G1 phase in contact-inhibited vascular endothelial cells. Hypertens Res 2002; 25:773-8. [PMID: 12452332 DOI: 10.1291/hypres.25.773] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In an attempt to find a strategy to modulate the proliferation of vascular endothelial cells, we examined whether constitutive activation of proto-oncogen protein p21 (Ras) induced the reentry of confluent human umbilical vascular endothelial cells (HUVECs) into the S phase. When an adenovirus construct expressing a constitutively active Ras mutant (Ad/RasG12V) was infected into HUVECs, their morphology changed strikingly and they appeared to be transformed. However, Ad/RasG12V-infected HUVECs did not enter the S phase, as determined by assessing 3H-thymidine incorporation. In accordance with the above results, the expression of cyclin A both at the transcript and protein levels did not increase in Ad/RasG12V-infected HUVECs relative to that in control cells, although the expression of cyclin D1 was induced in Ad/RasG12V-infected cells. Interestingly, the expression of the cyclin-dependent kinase (CDK) inhibitor p21cip1 was remarkably increased while that of p27kip1 did not decrease in Ad/RasG12V-infected HUVECs. Furthermore, CDK2 activity was not induced in Ad/RasG12V-infected HUVECs. These results suggested that the constitutive activation of Ras promoted the reentry of confluent HUVECs in the G0 phase into the G1 phase, but not into the S phase. The results also indicated that the constitutive activation of Ras might have induced the persistent expression of p21cip1 and p27kip1, and that this induction of p21cip1 and p27kip1 expression possibly caused the cell cycle arrest at the G1 phase.
Collapse
Affiliation(s)
- Etsu Suzuki
- Department of Intemal Medicine, Faculty of Medicine. University of Tokyo, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Suzuki E, Nishimatsu H, Satonaka H, Walsh K, Goto A, Omata M, Fujita T, Nagai R, Hirata Y. Angiotensin II induces myocyte enhancer factor 2- and calcineurin/nuclear factor of activated T cell-dependent transcriptional activation in vascular myocytes. Circ Res 2002; 90:1004-11. [PMID: 12016267 DOI: 10.1161/01.res.0000017629.70769.cc] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is well known that angiotensin II (Ang II) is implicated in the phenotypic modulation and hypertrophy of vascular smooth muscle cells (VSMCs). To study the mechanisms by which Ang II contributes to the pathological changes of VSMCs, we examined whether Ang II stimulated myocyte enhancer factor 2 (MEF2)- and calcineurin/nuclear factor of activated T cell (NFAT)-dependent transcriptional activation of genes in VSMCs. Ang II increased the DNA binding activity of MEF2A and its expression at the protein level. Ang II induced c-jun promoter activity, and this increase was inhibited by dominant-negative mutants of MEF2A and mitogen-activated protein kinase kinase 6 but not by calcineurin inhibitors. Ang II stimulated NFAT DNA binding activity and NFAT-dependent gene transcription, and these effects of Ang II were inhibited by calcineurin inhibitors. Furthermore, Ang II induced the promoter activity of the nonmuscle-type myosin heavy chain B gene, which we used as a marker of the dedifferentiated state of VSMCs, and this increase was inhibited by calcineurin inhibitors but not by the dominant-negative mutants of MEF2A or mitogen-activated protein kinase kinase 6. Finally, Ang II increased protein synthesis, and this increase was inhibited by infection with an adenovirus construct that expresses the dominant-negative mutant of MEF2A but not by calcineurin inhibitors. These results suggest that Ang II stimulates the MEF2- and calcineurin/NFAT-dependent pathways and that these pathways have distinct roles in VSMCs.
Collapse
Affiliation(s)
- Etsu Suzuki
- Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cuttitta F, Pío R, Garayoa M, Zudaire E, Julián M, Elsasser TH, Montuenga LM, Martínez A. Adrenomedullin functions as an important tumor survival factor in human carcinogenesis. Microsc Res Tech 2002; 57:110-9. [PMID: 11921362 DOI: 10.1002/jemt.10059] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Adrenomedullin (AM) is a pluripotent regulatory peptide initially isolated from a human pheochromocytoma (adrenal tumor) and subsequently shown to play a critical role in cancer cell division, tumor neovascularization, and circumvention of programmed cell death, thus it is an important tumor cell survival factor underlying human carcinogenesis. A variety of neural and epithelial cancers have been shown to produce abundant amounts of AM. Recent findings have implicated elevation of serum AM with the onset of malignant expression. In addition, patients with tumors producing high levels of this peptide have a poor prognostic clinical outcome. Given that most human epithelial cancers display a microenvironment of reduced oxygen tension, it is interesting to note that AM and several of its receptors are upregulated during hypoxic insult. The existence of such a regulatory pathway has been implicated as the basis for the overexpression of AM/AM-R in human malignancies, thereby generating a subsequent autocrine/paracrine growth advantage for the tumor cell. Furthermore, AM has been implicated as a potential immune suppressor substance, inhibiting macrophage function and acting as a newly identified negative regulator of the complement cascade, protective properties which may help cancer cells to circumvent immune surveillance. Hence, AM's traditional participation in normal physiology (cited elsewhere in this issue) can be extended to a primary player in human carcinogenesis and may have clinical relevance as a biological target for the intervention of tumor progression.
Collapse
Affiliation(s)
- Frank Cuttitta
- Cell and Cancer Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Adrenomedullin (AM) was originally identified in the extracts of human pheochromocytoma tissue, but this peptide is now known to be synthesized and secreted from many kinds of cells in the body, including vascular smooth muscle cells, endothelial cells, fibroblasts, cardiac myocytes, epithelial cells, and cancer cells. In this review, we summarize AM-secreting and AM gene-expressing cells in addition to the regulation of secretion and gene expression of AM. Although the data are still limited to deduce the general features of AM gene expression, synthesis, and secretion, AM is assumed to be classified into the new class of biologically active peptides, which is mainly expressed and secreted from non-endocrine type cells by the stimulation with inflammation-related substances. It is also interesting that serious physiological conditions such as inflammation or hypoxia potently stimulate AM expression and release, suggesting its unique physiological function distinct from other known biologically active peptides.
Collapse
Affiliation(s)
- Naoto Minamino
- National Cardiovascular Center Research Institute, Fujishirodai, Suita, Osaka 565-8565, Japan.
| | | | | |
Collapse
|
49
|
Yoshihara F, Horio T, Nishikimi T, Matsuo H, Kangawa K. Possible involvement of oxidative stress in hypoxia-induced adrenomedullin secretion in cultured rat cardiomyocytes. Eur J Pharmacol 2002; 436:1-6. [PMID: 11834240 DOI: 10.1016/s0014-2999(01)01462-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although hypoxia induces adrenomedullin gene expression in cultured rat cardiac myocytes, it is still unknown whether oxidative stress is involved in the hypoxia-induced adrenomedullin production. We investigated whether oxidative stress might participate in hypoxia-induced adrenomedullin secretion and whether adrenomedullin might have a protective effect on damaged myocytes. Hypoxia increased adrenomedullin secretion and its gene expression in cardiac myocytes, but not in nonmyocytes. Furthermore, oxidative stress (hydrogen peroxide) also increased adrenomedullin secretion from myocytes. N-acetyl-L-cysteine, a free radical scavenger, completely inhibited the stimulation of adrenomedullin secretion by hydrogen peroxide, and this agent reduced the stimulation of adrenomedullin secretion by hypoxia. Lactate dehydrogenase leakage, a marker of cell injury, was significantly increased with the exposure to hydrogen peroxide and adrenomedullin significantly reduced this leakage. These findings suggest that an oxidative stress may be involved, in part, in the increased adrenomedullin secretion from cardiac myocytes under hypoxic condition. Adrenomedullin secreted from myocytes may play a cell protective role in an autocrine manner.
Collapse
Affiliation(s)
- Fumiki Yoshihara
- Research Institute, National Cardiovascular Center, 5-7-1 Fujishirodai, Osaka 565-8565, Suita, Japan.
| | | | | | | | | |
Collapse
|
50
|
Totsune K, Takahashi K, Mackenzie HS, Arihara Z, Satoh F, Sone M, Murakami O, Ito S, Brenner BM, Mouri T. Adrenomedullin and its receptor complexes in remnant kidneys of rats with renal mass ablation: decreased expression of calcitonin receptor-like receptor and receptor-activity modifying protein-3. Peptides 2001; 22:1933-7. [PMID: 11754984 DOI: 10.1016/s0196-9781(01)00526-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Adrenomedullin (AM) has vasodilator and diuretic actions, similarly to natriuretic peptides. AM receptor complexes are composed of calcitonin receptor-like receptor (CRLR) and receptor-activity modifying protein-2 (RAMP2), or CRLR and RAMP3. We aimed to know whether gene expression of AM and AM receptor complexes are regulated in kidneys under pathophysiological conditions. Expression of AM, RAMP2, RAMP3 and CRLR mRNA was studied in the remnant kidney of rats with renal mass ablation using competitive quantitative RT-PCR techniques. Partial cloning was performed to determine the rat RAMP3 nucleotide sequence. In normal rat kidneys, expression levels of RAMP2, RAMP3, CRLR and AM mRNAs were 26.5 +/- 1.9 mmol/mole of GAPDH, 7.7 +/- 0.9 mmol/mole of GAPDH, 3.6 +/- 0.2 mmol/mole of GAPDH and 0.57 +/- 0.03 mmol/mole of GAPDH (mean +/- SE, n = 6), respectively. RAMP3 mRNA levels decreased significantly to about 50% and about 70% of control (sham-operated rats) 4 days and 14 days after 5/6 nephrectomy, respectively. CRLR mRNA levels also decreased significantly to about 30% and about 43% of control. Sodium intake restriction had no significant effects on the RAMP3 and CRLR gene expression. On the other hand, RAMP2 mRNA expression in the kidney was suppressed by sodium intake restriction regardless of nephrectomy, while RAMP2 levels in the remnant kidney were not significantly changed by 5/6 nephrectomy. Neither 5/6 nephrectomy or sodium intake restriction had any significant effects on the AM gene expression in the kidney. The present study showed that expression of mRNAs encoding AM, RAMP2, RAMP3 and CRLR were differentially regulated in remnant kidneys of rats with renal mass ablation.
Collapse
Affiliation(s)
- K Totsune
- Second Department of Internal Medicine, Tohoku University School of Medicine, 980-8574, Sendai, Miyagi, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|