Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Crit Care Med ; 44(8): e625-32, 2016 08.
Article in English | MEDLINE | ID: mdl-26963322

ABSTRACT

OBJECTIVES: Hepatic ischemia-reperfusion injury is a disease pattern that is associated with an acute inflammatory reaction. It is well known that neutrophils play an essential role in the early phase of hepatic ischemia-reperfusion injury and determine the extent of tissue damage. Hepatic ischemia-reperfusion injury can result in organ failure, which is linked to high mortality. Recent data indicate that the neuronal guidance receptor Plexin C1 is involved in the control of the acute inflammatory response and, as such, modulates the transmigration of neutrophils. Hence, we investigated the functional role of Plexin C1 in a mouse model of early hepatic ischemia-reperfusion injury. DESIGN: Animal study. SETTING: University experimental laboratory. SUBJECTS: Wild-type, PLXNC1 and chimeric mice. INTERVENTIONS: Hepatic ischemia-reperfusion injury or sham operation. MEASUREMENTS AND MAIN RESULTS: We found that the functional inhibition of Plexin C1 in wild-type mice treated with an anti-Plexin C1 antibody and a Semaphorin 7A peptide reduced hepatic ischemia-reperfusion injury, as measured by the levels of lactate dehydrogenase, aspartate, and alanine aminotransferase. This reduction in ischemia-reperfusion injury was accompanied by reduced numbers of neutrophils in ischemic hepatic tissue and reduced serum levels of inflammatory cytokines. Experiments using Plexin C1 receptor-deficient (PLXNC1) mice also demonstrated decreased hepatic ischemia-reperfusion injury. Studies of chimeric mice revealed that the hematopoietic Plexin C1 knockout is crucial for reducing the extent of hepatic ischemia-reperfusion injury. CONCLUSIONS: These results describe a role for Plexin C1 during ischemia-reperfusion injury, highlight the role of hematopoietic Plexin C1 in the development of hepatic ischemia-reperfusion injury, and suggest that Plexin C1 is a potential drug target.


Subject(s)
Cell Adhesion Molecules, Neuronal/agonists , Liver/physiopathology , Reperfusion Injury/prevention & control , Reperfusion Injury/physiopathology , Alanine Transaminase/metabolism , Animals , Antigens, CD/pharmacology , Aspartic Acid/metabolism , Disease Models, Animal , GPI-Linked Proteins/pharmacology , Inflammation Mediators/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Mice , Neutrophils/metabolism , Semaphorins/pharmacology
2.
Crit Care Med ; 42(9): e610-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25029243

ABSTRACT

OBJECTIVE: Liver ischemia and reperfusion injury is a common source of significant morbidity and mortality following liver transplantation, hemorrhagic shock, or major hepatic surgery. Based on studies showing a critical role for the neuronal guidance receptor neogenin (Neo1) outside the nervous system in mediating tissue adaption during acute inflammation, we hypothesized that Neo1 enhances hepatic ischemia and reperfusion injury. DESIGN: Animal study. SETTING: University-based experimental laboratory. SUBJECTS: Wid-type, neogenin deficient and chimeric mice. INTERVENTIONS: Neogenin expression was evaluated during inflammatory stimulation in vitro and during ischemia and reperfusion injury in vivo, intravital microscopy performed to study intravascular flow characteristics. The extent of liver injury was evaluated using histology, serum levels of lactate dehydrogenase, aspartate, and alanine aminotransferase. The functional role of Neo1 during liver IR was evaluated in mice with gene targeted repression of neogenin (Neo1-/-), bone marrow chimeric animals and controls. In addition, functional inhibition of neogenin was performed using antibody injection. MEASUREMENTS AND MAIN RESULTS: We observed an induction of Neo1 during inflammation in vitro and ischemia and reperfusion in vivo. Intravital microscopy demonstrated a decreased ability of Neo1 leukocytes to attach to endothelial vascular wall during inflammation. Subsequent studies in Neo1 mice showed attenuated serum levels of lactate dehydrogenase, aspartate, alanine, and proinflammatory cytokines during hepatic ischemia and reperfusion injury. This was associated with improved hepatic histology scores. Studies in chimeric animals demonstrated that the hematopoietic Neo1 expression to be crucial for the observed results. Treatment with an anti-Neo1 antibody resulted in a significant reduction of experimental hepatic ischemia and reperfusion injury, involving attenuated variable of lactate dehydrogenase, alanine, aspartate, and cytokine levels. CONCLUSIONS: These data provide a unique role for Neo1 in the development of hepatic ischemia and reperfusion injury and identified Neo1 as a potential target to prevent liver dysfunction in the future.


Subject(s)
Liver Diseases/epidemiology , Membrane Proteins/biosynthesis , Reperfusion Injury/prevention & control , Animals , Inflammation/physiopathology , Liver/physiopathology , Mice , Mice, Knockout , Neutrophils/metabolism
3.
Eur J Immunol ; 44(9): 2648-58, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24890788

ABSTRACT

Acute inflammation is the pathophysiological basis of important clinical conditions associated with organ failure. The initial inflammatory response is controlled by the chemokine system, yet recent data have indicated that the neuronal guidance cues are significantly involved in the orchestration of this process. Previous work has shown the proinflammatory capacity of the guidance cue semaphorin (Sema) 7a, but the role of one of its target receptors, the plexin C1 (PLXNC1) receptor is to date unknown. We report here that PLXNC1 is expressed outside the nervous system and induced during acute inflammation. PLXNC1(-/-) mice with C57BL/6 background demonstrated decreased inflammatory responses during zymosan A (ZyA)-induced peritonitis. Subsequent in vivo studies revealed altered rolling, adhesion, and transmigration properties of PLXNC1(-/-) leukocytes. Blockade of PLXNC1 was associated with attenuated chemotactic transendothelial migration properties in vitro. Studies in chimeric mice revealed that hematopoietic PLXNC1(-/-) animals demonstrated an attenuated inflammatory response. To probe the therapeutic potential of PLXNC1 we treated C57BL/6 WT mice with an anti-PLXNC1 antibody and a PLXNC1 binding peptide. Both of these interventions significantly dampened ZyA-induced peritonitis. These results implicate an important role of PLXNC1 during an acute inflammatory response and indicate PLXNC1 as a potential target for the control of conditions associated with acute inflammation.


Subject(s)
Nerve Tissue Proteins/immunology , Peritonitis/immunology , Receptors, Cell Surface/immunology , Acute Disease , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/pathology , Receptors, Cell Surface/genetics , Semaphorins/genetics , Semaphorins/immunology , Zymosan/toxicity
4.
BMC Cancer ; 13: 569, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24304513

ABSTRACT

BACKGROUND: Biomarkers allowing the characterization of malignancy and therapy response of oral squamous cell carcinomas (OSCC) or other types of carcinomas are still outstanding. The biochemical suicide molecule endonuclease DNaseX (DNaseI-like 1) has been used to identify the Apo10 protein epitope that marks tumor cells with abnormal apoptosis and proliferation. The transketolase-like protein 1 (TKTL1) represents the enzymatic basis for an anaerobic glucose metabolism even in the presence of oxygen (aerobic glycolysis/Warburg effect), which is concomitant with a more malignant phenotype due to invasive growth/metastasis and resistance to radical and apoptosis inducing therapies. METHODS: Expression of Apo10 and TKTL1 was analysed retrospectively in OSCC specimen (n = 161) by immunohistochemistry. Both markers represent independent markers for poor survival. Furthermore Apo10 and TKTL1 have been used prospectively for epitope detection in monocytes (EDIM)-blood test in patients with OSCC (n = 50), breast cancer (n = 48), prostate cancer (n = 115), and blood donors/controls (n = 74). RESULTS: Positive Apo10 and TKTL1 expression were associated with recurrence of the tumor. Multivariate analysis demonstrated Apo10 and TKTL1 expression as an independent prognostic factor for reduced tumor-specific survival. Apo10+/TKTL1+ subgroup showed the worst disease-free survival rate in OSCC.EDIM-Apo10 and EDIM-TKTL1 blood tests allowed a sensitive and specific detection of patients with OSCC, breast cancer and prostate cancer before surgery and in after care. A combined score of Apo10+/TKTL1+ led to a sensitivity of 95.8% and a specificity of 97.3% for the detection of carcinomas independent of the tumor entity. CONCLUSIONS: The combined detection of two independent fundamental biophysical processes by the two biomarkers Apo10 and TKTL1 allows a sensitive and specific detection of neoplasia in a noninvasive and cost-effective way. Further prospective trials are warranted to validate this new concept for the diagnosis of neoplasia and tumor recurrence.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , Deoxyribonuclease I/blood , Mouth Neoplasms/blood , Muscle Proteins/blood , Transketolase/blood , Antibodies, Monoclonal, Murine-Derived/chemistry , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Carcinoma, Squamous Cell/surgery , Case-Control Studies , Cell Line, Tumor , Deoxyribonuclease I/immunology , Disease-Free Survival , Female , Flow Cytometry , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Monocytes/metabolism , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Mouth Neoplasms/surgery , Multivariate Analysis , Muscle Proteins/immunology , Neck , Neoplasm Staging , Prognosis , ROC Curve , Retrospective Studies , Transketolase/immunology , Tumor Burden
5.
PLoS One ; 8(7): e69477, 2013.
Article in English | MEDLINE | ID: mdl-23936025

ABSTRACT

The UNC5 receptor family are chemorepulsive neuronal guidance receptors with additional functions outside the central nervous system. Previous studies have implicated that the UNC5B receptor influences the migration of leukocytes into sites of tissue inflammation. Given that this process is a critical step during the pathophysiology of myocardial ischemia followed by reperfusion (IR) we investigated the role of UNC5B during myocardial IR. In initial in-vitro experiments, the functional inhibition of UNC5B resulted in a significant reduction of chemotactic migration of neutrophils. In-vivo, using a model of acute myocardial ischemia in UNC5B(+/-) and wild type (WT) animals, we found a significant reduction of infarct sizes in UNC5B(+/-) animals. This was associated with significantly reduced levels of troponin-I and IL-6 in UNC5B(+/-) mice. The repression of UNC5B using siRNA and the functional inhibition of UNC5B significantly dampened the extent of myocardial IR injury. Following depletion of neutrophils, we were not able to observe any further reduction in infarct size through functional inhibition of UNC5B in WT and UNC5B(+/-) mice. In summary our studies demonstrate an important role for UNC5B during myocardial IR injury, and that UNC5B might be a potential therapeutic target to control reperfusion injury in the future.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Receptors, Cell Surface/metabolism , Animals , Cardiotonic Agents/metabolism , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Nervous System/metabolism , Netrin Receptors , Neutrophils/cytology , Transendothelial and Transepithelial Migration
7.
PLoS One ; 7(7): e41085, 2012.
Article in English | MEDLINE | ID: mdl-22848430

ABSTRACT

Recent evidence has demonstrated additional roles for the neuronal guidance protein receptor UNC5B outside the nervous system. Given the fact that ischemia reperfusion injury (IRI) of the liver is a common source of liver dysfunction and the role of UNC5B during an acute inflammatory response we investigated the role of UNC5B on acute hepatic IRI. We report here that UNC5B(+/-) mice display reduced hepatic IRI and neutrophil (PMN) infiltration compared to WT controls. This correlated with serum levels of lactate dehydrogenase (LDH), aspartate- (AST) and alanine- (ALT) aminotransferase, the presence of PMN within ischemic hepatic tissue, and serum levels of inflammatory cytokines. Moreover, injection of an anti-UNC5B antibody resulted in a significant reduction of hepatic IR injury. This was associated with reduced parameters of liver injury (LDH, ALT, AST) and accumulation of PMN within the injured hepatic tissue. In conclusion our studies demonstrate a significant role for UNC5B in the development of hepatic IRI and identified UNC5B as a potential drug target to prevent liver dysfunction in the future.


Subject(s)
Liver/metabolism , Receptors, Cell Surface/metabolism , Reperfusion Injury/metabolism , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , L-Lactate Dehydrogenase/metabolism , Liver/pathology , Liver/physiopathology , Mice , Mice, Knockout , Netrin Receptors , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Neutrophils/pathology , Receptors, Cell Surface/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
8.
PLoS One ; 7(3): e32145, 2012.
Article in English | MEDLINE | ID: mdl-22412855

ABSTRACT

Neuronal guidance proteins (NGP) were originally described in the context of axonal growth and migration. Yet recent work has demonstrated that NGPs also serve as guidance cues for immune competent cells. A crucial target receptor for NGPs during embryonic development is the neogenin receptor, however its role during acute inflammation is unknown. We report here that neogenin is abundantly expressed outside the nervous system and that animals with endogenous repression of neogenin (Neo1(-/-)) demonstrate attenuated changes of acute inflammation. Studies using functional inhibition of neogenin resulted in a significant attenuation of inflammatory peritonitis. In studies employing bone marrow chimeric animals we found the hematopoietic presence of Neo1(-/-) to be responsible for the attenuated inflammatory response. Taken together our studies suggest that the guidance receptor neogenin holds crucial importance for the propagation of an acute inflammatory response and further define mechanisms shared between the nervous and the immune system.


Subject(s)
Inflammation/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Cytokines/metabolism , Gene Expression , Inflammation/genetics , Inflammation Mediators/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Knockout , Organ Specificity/genetics , Peritonitis/genetics , Peritonitis/metabolism , Receptors, Cell Surface/genetics
10.
Inflammation ; 35(2): 566-73, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21607702

ABSTRACT

The endothelial barrier controls the passage of solutes from the vascular space. This is achieved through active reorganization of the actin cytoskeleton. A central cytoskeletal protein involved into this is vasodilator-stimulated phosphoprotein (VASP). However, the functional role of endothelial VASP during hypoxia has not been thoroughly elucidated. We determined endothelial VASP expression through real-time PCR (Rt-PCR), immunhistochemistry, and Western blot analysis during hypoxia. VASP promoter studies were performed using a PGL3 firefly luciferase containing plasmid. Following approval by the local authorities, VASP ( -/- ) mice and littermate controls were subjected to normobaric hypoxia (8% O(2), 92% N(2)) after intravenous injection of Evans blue dye. In in vitro studies, we found significant VASP repression in human microvascular and human umbilical vein endothelial cells through Rt-PCR, immunhistochemistry, and Western blot analysis. The VASP promoter construct demonstrated significant repression in response to hypoxia, which was abolished when the binding of hypoxia-inducible factor 1 alpha was excluded. Exposure of wild-type (WT) and VASP ( -/- ) animals to normobaric hypoxia for 4 h resulted in an increase in Evans blue tissue extravasation that was significantly increased in VASP ( -/- ) animals compared to WT controls. In summary, we demonstrate here that endothelial VASP holds significant importance for endothelial barrier properties during hypoxia.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Hypoxia , Endothelium, Vascular/physiology , Hypoxia/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Actin Cytoskeleton/physiology , Animals , Capillary Permeability , Cell Adhesion Molecules/genetics , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Phosphoproteins/genetics , Promoter Regions, Genetic
11.
FASEB J ; 26(4): 1549-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22198383

ABSTRACT

Lung injury is marked by a persistent self-propagating inflammation within the pulmonary tissue that is initiated by the migration of leukocytes into the alveolar space. Recent work has demonstrated that neuronal guidance proteins are involved into the orchestration of leukocyte migration. Neogenin is a crucial guidance receptor for axonal migration, yet its role during leukocyte migration and acute inflammation is to date unknown. Here, we report that neogenin influences neutrophil migration across endothelial HMEC-1 and alveolar A549 monolayers in vitro. In vivo, Neo1(-/-) mice demonstrated 59% reduced cell count, 41% reduced TNF-α, and 76% reduced IL-6 levels within the alveolar space during lung injury. In studies employing chimeric animals, the presence of Neo1(-/-) bone marrow was associated with a 42% reduction of cell count and reduced inflammatory changes within pulmonary tissue during lung injury. The functional inhibition of neogenin through antibody injection confirmed these results and the role of neogenin for the inflammatory changes within the alveolar space. Previously unappreciated, the guidance receptor neogenin has a significant effect on the orchestration of leukocyte migration and the control of acute inflammation.


Subject(s)
Lung Injury/metabolism , Membrane Proteins/metabolism , Pneumonia/metabolism , Animals , Cell Line , Cell Movement/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Interleukin-6/metabolism , Lung Injury/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Neutrophils/cytology , Neutrophils/physiology , Pneumonia/pathology , Respiration, Artificial/adverse effects , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 186(1): 549-55, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098235

ABSTRACT

Previous studies implicated the anti-inflammatory potential of the adenosine 2B receptor (A2BAR). A2BAR activation is achieved through adenosine, but this is limited by its very short t(1/2). To further define alternative adenosine signaling, we examined the role of netrin-1 during acute inflammatory peritonitis. In this article, we report that animals with endogenous repression of netrin-1 (Ntn1(+/-)) demonstrated increased cell count, increased peritoneal cytokine concentration, and pronounced histological changes compared with controls in a model of zymosan A peritonitis. Exogenous netrin-1 significantly decreased i.p. inflammatory changes. This effect was not present in animals with deletion of A2BAR (A2BAR(-/-)). A2BAR(-/-) animals demonstrated no change in cell count, i.p. cytokine concentration, or histology in response to netrin-1 injection. These data strengthen the role of netrin-1 as an immunomodulatory protein exerting its function in dependence of the A2BAR and further define alternative adenosine receptor signaling.


Subject(s)
Down-Regulation/immunology , Inflammation Mediators/physiology , Nerve Growth Factors/physiology , Peritonitis/immunology , Peritonitis/prevention & control , Signal Transduction/immunology , Tumor Suppressor Proteins/physiology , Acute Disease , Animals , Caco-2 Cells , Disease Models, Animal , Humans , Immunologic Factors/biosynthesis , Immunologic Factors/physiology , Mice , Mice, Knockout , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/deficiency , Netrin-1 , Peritonitis/metabolism , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/physiology , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/deficiency
13.
PLoS One ; 6(12): e29494, 2011.
Article in English | MEDLINE | ID: mdl-22216296

ABSTRACT

Recent work has demonstrated that the formation of platelet neutrophil complexes (PNCs) affects inflammatory tissue injury. Vasodilator-stimulated phosphoprotein (VASP) is crucially involved into the control of PNC formation and myocardial reperfusion injury. Given the clinical importance of hepatic IR injury we pursued the role of VASP during hepatic ischemia followed by reperfusion. We report here that VASP(-/-) animals demonstrate reduced hepatic IR injury compared to wildtype (WT) controls. This correlated with serum levels of lactate dehydrogenase (LDH), aspartate (AST) and alanine (ALT) aminotransferase and the presence of PNCs within ischemic hepatic tissue and could be confirmed using repression of VASP through siRNA. In studies employing bone marrow chimeric mice we identified hematopoietic VASP to be of crucial importance for the extent of hepatic injury. Phosphorylation of VASP on Ser(153) through Prostaglandin E1 or on Ser(235) through atrial natriuretic peptide resulted in a significant reduction of hepatic IR injury. This was associated with a reduced presence of PNCs in ischemic hepatic tissue. Taken together, these studies identified VASP and VASP phosphorylation as crucial target for future hepatoprotective strategies.


Subject(s)
Cell Adhesion Molecules/metabolism , Liver/blood supply , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Reperfusion Injury/pathology , Alprostadil/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules/genetics , Flow Cytometry , Immunohistochemistry , Mice , Mice, Knockout , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation , Reperfusion Injury/metabolism
14.
Crit Care ; 14(5): R189, 2010.
Article in English | MEDLINE | ID: mdl-20969752

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) is an inflammatory disorder of pulmonary or extrapulmonary origin. We have previously demonstrated that netrin-1 dampens murine ALI, and in an attempt to advance this finding into future clinical practice we evaluated whether netrin-1 would reduce alveolar inflammation during porcine ALI. METHODS: This was a controlled in vivo experimental study in pigs. We induced ALI through lipoploysaccharide (LPS) infusion (50 µg/kg) for 2 hours. Following this, we exposed animals to either vehicle, intravenous netrin-1 (netrin-1 i.v.) or inhaled netrin-1 (netrin-1 inh.). Serum samples and bronchoalveolar lavage (BAL) were obtained to determine levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, interleukin-6 and interleukin-8 at baseline and 6 hours following treatment. Myeloperoxidase activity (MPO) and protein levels were determined in the BAL, and tissue samples were obtained for histological evaluation. Finally, animals were scanned with spiral CT. RESULTS: Following LPS infusion, animals developed acute pulmonary injury. Serum levels of TNF-α and IL-6 were significantly reduced in the netrin-1 i.v. group. BAL demonstrated significantly reduced cytokine levels 6 hours post-netrin-1 treatment (TNF-α: vehicle 633 ± 172 pg/ml, netrin-1 i.v. 84 ± 5 pg/ml, netrin-1 inh. 168 ± 74 pg/ml; both P < 0.05). MPO activity and protein content were significantly reduced in BAL samples from netrin-1-treated animals. Histological sections confirmed reduced inflammatory changes in the netrin-1-treated animals. Computed tomography corroborated reduced pulmonary damage in both netrin-1-treated groups. CONCLUSIONS: We conclude that treatment with the endogenous anti-inflammatory protein netrin-1 reduces pulmonary inflammation during the initial stages of ALI and should be pursued as a future therapeutic option.


Subject(s)
Acute Lung Injury/pathology , Nerve Growth Factors/physiology , Neurons/pathology , Pulmonary Alveoli/pathology , Tumor Suppressor Proteins/physiology , Acute Lung Injury/drug therapy , Animals , Cell Movement/drug effects , Cell Movement/physiology , Disease Models, Animal , Female , Inflammation/pathology , Inflammation/prevention & control , Nerve Growth Factors/administration & dosage , Netrin-1 , Neurons/drug effects , Pulmonary Alveoli/drug effects , Swine , Tumor Suppressor Proteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...