Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 254
1.
J Reprod Immunol ; 153: 103692, 2022 09.
Article En | MEDLINE | ID: mdl-35970080

Indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO) are key enzymes for tryptophan degradation, regulating immune tolerance during pregnancy. The intrauterine renin-angiotensin system is also involved in the progression of a healthy pregnancy. Angiotensin(1-7) maintains the integrity of fetal membranes via counteracting the pro-inflammatory actions of Angiotensin II. No data are available on placental Angiotensin(1-7) co-expression with TDO. We aimed to characterize TDO mRNA expression and its localization in different areas of the placenta of physiological pregnancies delivered at term; its co-expression with Angiotensin(1-7) and its correlation with the plasma kynurenine/tryptophan (Kyn/Trp) ratio was investigated. This prospective observational study included a nonconsecutive series of 20 singleton uncomplicated pregnancies delivered vaginally. TDO mRNA was expressed in both maternal and fetal sides of the placentas and TDO protein also in the villi and it was co-expressed with IDO1 in almost half of the placental cells at these sites. The percentage of TDO+ and IDO1+ cells appeared to be influenced by maternal pre-gestational smoking and newborn weight. A strong correlation was found between the percentage of TDO+ and IDO1+ cells in the villi. TDO+ cells also expressed Angiotensin(1-7), with a higher percentage on the fetal side and in the villi compared to the maternal one. Kyn/Trp plasma ratio was not correlated with IDO and TDO expression nor with the patient's characteristics. Collectively, our data indicate that TDO is detectable in placental tissue and is co-expressed with IDO and with Angiotensin(1-7)+ on the fetal side and in the villi.


Angiotensin I , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase , Peptide Fragments , Placenta , Tryptophan Hydroxylase , Angiotensin I/genetics , Angiotensin I/immunology , Angiotensin II/immunology , Female , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Infant, Newborn , Kynurenine/analysis , Kynurenine/genetics , Kynurenine/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Placenta/enzymology , Placenta/immunology , Pregnancy , RNA, Messenger , Tryptophan/analysis , Tryptophan/genetics , Tryptophan/immunology , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/immunology , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/immunology
2.
Brain Res ; 1772: 147667, 2021 12 01.
Article En | MEDLINE | ID: mdl-34587500

INTRODUCTION: As several clinical trials have revealed that angiotensin-converting enzyme inhibitors and angiotensin II (Ang II) receptor blockers may be efficient in treating vascular dementia (VaD), the long-acting blockade of the renin-angiotensin system (RAS) would be useful considering the poor adherence of antihypertensive drugs. Accordingly, we continuously blocked RAS via vaccination and examined the effectiveness of the VaD model in rats. METHODS: Male Wistar rats were exposed to two-vessel occlusions (2VO) after three injections of Ang II peptide vaccine. The effects of the vaccine were evaluated in the novel object recognition test, brain RAS components, and markers for oligodendrocytes. RESULTS: In the vaccinated rats, anti-Ang II antibody titer level was increased in serum until Day 168, but not in cerebral parenchyma. Vaccinated rats showed better object recognition memory with inhibited demyelination in the corpus callosum and activation of astrocytes and microglia. Also, levels of BrdU/GSTπ-positive cells and the phosphorylation of cAMP response element binding protein was increased in vaccinated rats, indicating that the differentiation of oligodendrocyte progenitor cells to mature oligodendrocytes was accelerated. Vaccinated rats showed increased expression of fibroblast growth factor-2 (FGF2), which was observed in endothelial cells. Angiotensinogen mRNA was decreased at 7 days after 2VO but increased at 14 and 28 days. CONCLUSION: Ang II vaccine might have promoted oligodendrocyte differentiation and inhibited astrocytic and microglial activation by stimulating FGF2 signaling in the endothelial cells-oligodendrocyte/astrocyte/microglia coupling. These data indicate the feasibility of Ang II vaccine for preventing progression of vascular dementia.


Dementia, Vascular/prevention & control , Immunotherapy/methods , Renin-Angiotensin System/immunology , Angiotensin II/immunology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antibodies/analysis , Brain/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Demyelinating Diseases/prevention & control , Male , Memory/physiology , Phosphorylation , Rats , Rats, Wistar , Recognition, Psychology , Vaccination , Vaccines, Subunit/therapeutic use
3.
PLoS One ; 16(9): e0257016, 2021.
Article En | MEDLINE | ID: mdl-34478478

BACKGROUND: Activation of the immune system is implicated in the Post-Acute Sequelae after SARS-CoV-2 infection (PASC) but the mechanisms remain unknown. Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II (Ang II) resulting in decreased activation of the AT1 receptor and decreased immune system activation. We hypothesized that autoantibodies against ACE2 may develop after SARS-CoV-2 infection, as anti-idiotypic antibodies to anti-spike protein antibodies. METHODS AND FINDINGS: We tested plasma or serum for ACE2 antibodies in 67 patients with known SARS-CoV-2 infection and 13 with no history of infection. None of the 13 patients without history of SARS-CoV-2 infection and 1 of the 20 outpatients that had a positive PCR test for SARS-CoV-2 had levels of ACE2 antibodies above the cutoff threshold. In contrast, 26/32 (81%) in the convalescent group and 14/15 (93%) of patients acutely hospitalized had detectable ACE2 antibodies. Plasma from patients with antibodies against ACE2 had less soluble ACE2 activity in plasma but similar amounts of ACE2 protein compared to patients without ACE2 antibodies. We measured the capacity of the samples to inhibit ACE2 enzyme activity. Addition of plasma from patients with ACE2 antibodies led to decreased activity of an exogenous preparation of ACE2 compared to patients that did not have antibodies. CONCLUSIONS: Many patients with a history of SARS-CoV-2 infection have antibodies specific for ACE2. Patients with ACE2 antibodies have lower activity of soluble ACE2 in plasma. Plasma from these patients also inhibits exogenous ACE2 activity. These findings are consistent with the hypothesis that ACE2 antibodies develop after SARS-CoV-2 infection and decrease ACE2 activity. This could lead to an increase in the abundance of Ang II, which causes a proinflammatory state that triggers symptoms of PASC.


Autoantibodies/blood , COVID-19/immunology , SARS-CoV-2/isolation & purification , Spike Glycoprotein, Coronavirus/blood , Angiotensin II/blood , Angiotensin II/immunology , Angiotensin-Converting Enzyme 2/genetics , Autoantibodies/immunology , Autoantibodies/isolation & purification , COVID-19/blood , COVID-19/virology , Female , Humans , Male , Peptidyl-Dipeptidase A/blood , Receptor, Angiotensin, Type 1/blood , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/immunology , Renin-Angiotensin System/genetics , Renin-Angiotensin System/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/isolation & purification
4.
Mol Cell Endocrinol ; 529: 111254, 2021 06 01.
Article En | MEDLINE | ID: mdl-33798633

The most classical view of the renin-angiotensin system (RAS) emphasizes its role as an endocrine regulator of sodium balance and blood pressure. However, it has long become clear that the RAS has pleiotropic actions that contribute to organ damage, including modulation of inflammation. Angiotensin II (Ang II) activates angiotensin type 1 receptors (AT1R) to promote an inflammatory response and organ damage. This represents the pathophysiological basis for the successful use of RAS blockers to prevent and treat kidney and heart disease. However, other RAS components could have a built-in capacity to brake proinflammatory responses. Angiotensin type 2 receptor (AT2R) activation can oppose AT1R actions, such as vasodilatation, but its involvement in modulation of inflammation has not been conclusively proven. Angiotensin-converting enzyme 2 (ACE2) can process Ang II to generate angiotensin-(1-7) (Ang-(1-7)), that activates the Mas receptor to exert predominantly anti-inflammatory responses depending on the context. We now review recent advances in the understanding of the interaction of the RAS with inflammation. Specific topics in which novel information became available recently include intracellular angiotensin receptors; AT1R posttranslational modifications by tissue transglutaminase (TG2) and anti-AT1R autoimmunity; RAS modulation of lymphoid vessels and T lymphocyte responses, especially of Th17 and Treg responses; interactions with toll-like receptors (TLRs), programmed necrosis, and regulation of epigenetic modulators (e.g. microRNAs and bromodomain and extraterminal domain (BET) proteins). We additionally discuss an often overlooked effect of the RAS on inflammation which is the downregulation of anti-inflammatory factors such as klotho, peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), transient receptor potential ankyrin 1 (TRPA1), SNF-related serine/threonine-protein kinase (SNRK), serine/threonine-protein phosphatase 6 catalytic subunit (Ppp6C) and n-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). Both transcription factors, such as nuclear factor κB (NF-κB), and epigenetic regulators, such as miRNAs are involved in downmodulation of anti-inflammatory responses. A detailed analysis of pathways and targets for downmodulation of anti-inflammatory responses constitutes a novel frontier in RAS research.


Angiotensin II/immunology , Angiotensin I/immunology , Inflammation/immunology , Peptide Fragments/immunology , Renin-Angiotensin System/immunology , Water-Electrolyte Balance/immunology , Angiotensin I/genetics , Angiotensin II/genetics , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , Autoimmunity , Blood Pressure/genetics , Blood Pressure/immunology , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Kidney/cytology , Kidney/immunology , Klotho Proteins/genetics , Klotho Proteins/immunology , Peptide Fragments/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/immunology , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/immunology , Renin-Angiotensin System/genetics , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Water-Electrolyte Balance/genetics
5.
J Hypertens ; 39(1): 181-189, 2021 01.
Article En | MEDLINE | ID: mdl-32667158

OBJECTIVES: We recently developed a novel peptide, AJP001, that possesses both a mouse T-cell epitope and adjuvant action. Direct conjugation to the antigen is useful for peptide vaccines without the addition of adjuvants. In this study, the efficacy of an angiotensin (Ang) II and AJP001-conjugated peptide vaccine (AJ-Ang II) was evaluated in mice. METHODS: The anti-Ang II antibody titer was measured in Balb/C mice following three injections of AJ-Ang II at 2-week intervals. SBP was measured during vaccination of Balb/C mice treated with Ang II infusion (1 µg/kg per min). RESULTS: AJ-Ang II treatment resulted in an increase in the anti-Ang II antibody titer in a dose-dependent manner without the addition of adjuvants. In the analysis of the humoral immune response, AJ-Ang II mainly elicited IgG1 antibodies and IL-4 and IL-10 production, as measured by an enzyme-linked immune absorbent spot assay, which suggests the induction of a Th2 response. Importantly, cotreatment with purified antibodies attenuated Ang II-induced extracellular signal-regulated kinase phosphorylation and nuclear factor (NF)-κB activation in cultured vascular smooth muscle cells. The SBP in immunized mice was significantly lower than that in nonimmunized mice (135.9 ±â€Š8.5 vs. 154.9 ±â€Š16.8 mmHg, P = 0.02). Furthermore, Ang II-induced perivascular fibrosis in the heart was significantly attenuated in immunized mice, which also exhibited decreased mRNA expression of collagen I/III and transforming growth factor-ß. CONCLUSION: AJ-Ang II may be a simple and useful therapeutic peptide vaccine without the addition of any adjuvants.


Angiotensin II , Epitopes, T-Lymphocyte/immunology , Vaccines, Subunit , Adjuvants, Immunologic , Angiotensin II/immunology , Animals , Cells, Cultured , Fibrosis , Heart , Mice , Myocytes, Smooth Muscle
6.
Adv Chronic Kidney Dis ; 27(5): 404-411, 2020 09.
Article En | MEDLINE | ID: mdl-33308506

Hypertension emerged from early reports as a potential risk factor for worse outcomes for persons with coronavirus disease 2019 (COVID-19). Among the putative links between hypertension and COVID-19 is a key counter-regulatory component of the renin-angiotensin system (RAS): angiotensin-converting enzyme 2 (ACE2). ACE2 facilitates entry of severe acute respiratory syndrome coronavirus 2, the virus responsible for COVID-19, into host cells. Because RAS inhibitors have been suggested to increase ACE2 expression, health-care providers and patients have grappled with the decision of whether to discontinue these medications during the COVID-19 pandemic. However, experimental models of analogous viral pneumonias suggest RAS inhibitors may exert protective effects against acute lung injury. We review how RAS and ACE2 biology may affect outcomes in COVID-19 through pulmonary and other systemic effects. In addition, we briefly detail the data for and against continuation of RAS inhibitors in persons with COVID-19 and summarize the current consensus recommendations from select specialty organizations.


Acute Lung Injury/metabolism , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , COVID-19/metabolism , Hypertension/drug therapy , Acute Lung Injury/epidemiology , Acute Lung Injury/immunology , Angiotensin I/immunology , Angiotensin I/metabolism , Angiotensin II/immunology , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2/immunology , COVID-19/epidemiology , COVID-19/immunology , Comorbidity , Humans , Hypertension/epidemiology , Hypertension/metabolism , JNK Mitogen-Activated Protein Kinases/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/immunology , Lung/metabolism , MAP Kinase Signaling System , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protective Factors , Receptors, Coronavirus/immunology , Receptors, Coronavirus/metabolism , Renin-Angiotensin System , Risk Factors , SARS-CoV-2 , Up-Regulation
7.
Hypertension ; 76(6): 1879-1888, 2020 12.
Article En | MEDLINE | ID: mdl-33040618

There is no proven medical therapy to inhibit the progression of abdominal aortic aneurysm (AAA) in the clinical setting. To develop a novel therapeutic approach for the treatment of AAA, we focused on vaccination targeting Ang II (angiotensin II) and assessed the effect of an Ang II peptide vaccine on the progression of AAA using a rat model. Ang II peptide was conjugated with KLH (keyhole limpet hemocyanin) carrier protein to induce a sufficient immune response. Male rats were subcutaneously immunized with Ang II-KLH with an adjuvant on days 0, 14, and 28. Aortic dilatation was induced by intraluminal incubation with elastase on day 35. Treatment with Ang II vaccine successfully induced the production of a high titer of anti-Ang II antibodies. Immunization with Ang II vaccine resulted in a significant reduction in expansion of the aortic diameter compared with control rats, without a blood pressure-lowering effect. Four weeks after operation, the increase in Ang II in the aneurysm wall was significantly inhibited by treatment with Ang II vaccine. Inhibition of Ang II action led to suppression of the inflammatory response in the AAA wall through attenuation of the NFκB (nuclear factor kappa B) and c-jun N-terminal kinase signaling cascades. Treatment with Ang II vaccine inhibited accumulation of macrophages in the AAA wall. In addition, expression of TNF-α (tumor necrosis factor alpha) and activation of MMP (matrix metalloproteinase)-2 and MMP-9 were also inhibited by treatment with Ang II vaccine, resulting in protection against the destruction of elastic fibers. This vaccine therapy could become a potent therapeutic option to treat patients with AAA.


Angiotensin II/immunology , Aortic Aneurysm, Abdominal/prevention & control , Disease Models, Animal , Vaccines, Subunit/administration & dosage , Animals , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Disease Progression , Hemocyanins/immunology , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/immunology , Vaccines, Subunit/immunology
8.
Nat Commun ; 11(1): 4549, 2020 09 11.
Article En | MEDLINE | ID: mdl-32917889

Arterial macrophages have different developmental origins, but the association of macrophage ontogeny with their phenotypes and functions in adulthood is still unclear. Here, we combine macrophage fate-mapping analysis with single-cell RNA sequencing to establish their cellular identity during homeostasis, and in response to angiotensin-II (AngII)-induced arterial inflammation. Yolk sac erythro-myeloid progenitors (EMP) contribute substantially to adventitial macrophages and give rise to a defined cluster of resident immune cells with homeostatic functions that is stable in adult mice, but declines in numbers during ageing and is not replenished by bone marrow (BM)-derived macrophages. In response to AngII inflammation, increase in adventitial macrophages is driven by recruitment of BM monocytes, while EMP-derived macrophages proliferate locally and provide a distinct transcriptional response that is linked to tissue regeneration. Our findings thus contribute to the understanding of macrophage heterogeneity, and associate macrophage ontogeny with distinct functions in health and disease.


Arteries/cytology , Arteritis/immunology , Cell Differentiation/physiology , Homeostasis/physiology , Macrophages/physiology , Aging/physiology , Angiotensin II/administration & dosage , Angiotensin II/immunology , Animals , Arteries/physiology , Bone Marrow/physiology , Bone Marrow Transplantation , Cell Lineage , Disease Models, Animal , Female , Hematopoietic Stem Cells/physiology , Humans , Male , Mice , Mice, Transgenic , RNA-Seq , Regeneration/physiology , Single-Cell Analysis , Transplantation Chimera
9.
Int Immunopharmacol ; 84: 106507, 2020 Jul.
Article En | MEDLINE | ID: mdl-32339920

The formation of an immunosuppressive microenvironment and up-regulation of PD-L1 protein are the main causes of tumor immune escape. Previous reports suggest that Angiotensin II (Ang II) can modulate the immune status of tumor microenvironment in non-small cell lung cancer (NSCLC), but the underlying mechanism remains not fully understood. Here we demonstrated that AngII treatment causes the reduction of intratumoral infiltrating CD4 T lymphocytes in tumor-bearing mice, increases the accumulation of immunosuppressive granulocytes and TAMs in tumor tissue, and upregulates the expression levels of immunosuppressive marker genes. In addition, AngII/AGTR1 axis triggers cell PD-L1 expression through a mechanism involving increases in PD-L1 mRNA stability by human antigen R (HuR), an AU-rich element (ARE)-binding protein. Collectively, AngII/AGTR1 signaling promotes the tumor immunosuppressive microenvironment by upregulating PD-L1 in NSCLC, the mechanism of which is largely accounted by HuR-mediated PD-L1 mRNA stabilization.


Angiotensin II/immunology , B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Immune Tolerance , Lung Neoplasms/immunology , Animals , B7-H1 Antigen/genetics , Cell Line , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Inbred C57BL , RNA, Messenger , Tumor Microenvironment/immunology
10.
J Pathol ; 247(5): 686-696, 2019 04.
Article En | MEDLINE | ID: mdl-30506724

Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Adaptive Immunity/physiology , Lung/growth & development , Angiotensin II/immunology , Angiotensin II/physiology , Animals , Bronchi/cytology , Fetal Development/immunology , Fetal Development/physiology , Humans , Lung/embryology , Lung/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Mice , Neovascularization, Physiologic/immunology , Neovascularization, Physiologic/physiology , Pneumonia/immunology , Pneumonia/physiopathology , Regeneration/immunology , Regeneration/physiology , Signal Transduction/immunology , Signal Transduction/physiology
11.
Front Immunol ; 10: 2885, 2019.
Article En | MEDLINE | ID: mdl-31921148

Patients with Type 2 Diabetes Mellitus (T2DM) suffer from a higher incidence and severity of pulmonary infections. This is likely due to immune impairment and structural abnormalities caused by T2DM-induced oxidative stress (OS) and chronic inflammation. Modulation of the Renin Angiotensin System (RAS) through blockade of the actions of angiotensin II (AII), or inducing the protective pathway, has the potential to reduce these pathological pathways. The effects of Angiotensin 1-7 [A(1-7)] and NorLeu3-A(1-7) [NorLeu], ligands of the protective RAS, on the innate immune response were evaluated in the db/db mouse model of T2DM. Only NorLeu treatment reduced the structural pathologies in the lung caused by T2DM. A decreased in bactericidal activity and phagocytosis in diabetic animals was also observed; both A(1-7) and NorLeu treatment restored these functions. Myeloid progenitor CFUs were reduced and neutrophil/progenitor OS was increased in saline-treated db/db mice, and was reversed by A(1-7) and NorLeu treatment. These results demonstrate the adverse effects of diabetes on factors that contribute to pulmonary infections and the therapeutic potential of protective RAS peptides. Overall, RAS-modification may be a viable therapeutic target to treat diabetic complications that are not addressed by glucose lowering drugs.


Angiotensin II/pharmacology , Angiotensin I/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/immunology , Immunity, Innate/drug effects , Peptide Fragments/pharmacology , Renin-Angiotensin System/drug effects , Angiotensin II/immunology , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Male , Mice , Mice, Transgenic , Renin-Angiotensin System/immunology
12.
Curr Hypertens Rep ; 20(3): 22, 2018 03 19.
Article En | MEDLINE | ID: mdl-29556794

PURPOSE OF REVIEW: Vaccines are commonly used as preventive methods, primarily against infectious diseases. The goal of our study is to develop the therapeutic vaccine for hypertension. RECENT FINDINGS: We and others recently reported that an angiotensin II (AngII) vaccine for hypertension successfully attenuated elevated blood pressures in an animal model without any immunogenic side effects. In this system, an immunogenic molecule (i.e., KLH) with adjuvants provides an antigen that supports the activation of helper T cells. In addition, pretreatment with the AngII vaccine exerts neuroprotective effects in a cerebral ischemia model and cardioprotective effects in a myocardial infarction model. In the early phase of clinical trial, the administration of an AngII vaccine (AngQb-Cyt006) successfully decreased blood pressure in hypertensive patients with the increase of anti-AngII antibody titer. Increasing the effectiveness of drug adherence interventions in the clinical setting may have a large impact on the health of the population, which can be improved by using successful therapeutic vaccines. In this review, we describe the concept of therapeutic vaccines for hypertension and future directions for therapeutic vaccines.


Blood Pressure/drug effects , Hypertension/therapy , Oligopeptides/pharmacology , Vaccines/therapeutic use , Angiotensin II/immunology , Angiotensin II/pharmacology , Animals , Blood Pressure/immunology , Disease Models, Animal , Humans , Hypertension/immunology
13.
Microbiol Immunol ; 61(12): 539-546, 2017 Dec.
Article En | MEDLINE | ID: mdl-29052263

Angiotensin II (ANG II) plays critical roles in modulation of circulatory homeostasis and activation of innate and adaptive immunity and has also been implicated in several mouse models of autoimmune disease. However, how ANG II regulates macrophages and is involved in development of experimental autoimmune myocarditis (EAM) remains unclear. Therefore, the present study aimed to address the above question and explore possible mechanisms. EAM was induced in BALB/c mice. ANG II was quantitated by ELISA and hematoxylin and eosin staining was employed to analyze pathological changes and macrophage infiltration. The chemotactic ability of ANG II was assessed by using a Transwell system. It was found that ANG II is up-regulated in serum and heart tissues of mice with EAM and that ANG II significantly drives monocyte/macrophage infiltration through the C-C chemokine receptor 2/5 (CCR2/5) axis. CCR2/5 antagonists and ANG II receptor inhibitor could all abrogate monocyte/macrophage infiltration and ameliorate development of EAM. Our results have firstly identified a novel function of ANG II: that it is a critical chemokine for monocyte/macrophage recruitment. Furthermore, our results indicate that ANG II is a potential candidate for treatment of inflammatory diseases.


Angiotensin II/immunology , Autoimmune Diseases/immunology , Macrophages/immunology , Monocytes/immunology , Myocarditis/immunology , Receptors, CCR2/immunology , Receptors, CCR5/immunology , Angiotensin II/genetics , Animals , Autoimmune Diseases/genetics , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred BALB C , Myocarditis/genetics , Receptors, CCR2/genetics , Receptors, CCR5/genetics
14.
Life Sci ; 191: 219-226, 2017 Dec 15.
Article En | MEDLINE | ID: mdl-29032115

AIM: Angiotensin II (AngII), a corpus cavernosum (CC) constrictor peptide, modulates Toll like receptor (TLR) expression, a key element of the innate immune system, contributing to impaired vascular function in pathological conditions. However, it is unknown whether TLR4 is involved in AngII-induced erectile dysfunction. In this study, we investigated whether TLR4 plays a role in cavernosal dysfunction caused by AngII upregulation. MATERIAL AND METHODS: Cavernosal smooth muscle cells (CSMC) from C57/BL6 mice were treated with AngII (0.1µM) or bacterial LPS (50ng/ml) for 12-24h and TLR4 expression was assessed. Mice were infused with AngII (90ng/min, 28days) and treated with anti-TLR4 antibody (0.1mg/daily, i.p.) for the last 14days of the treatment. CC tissue was used for functional studies and for Western blotting. Nitric Oxide Synthase (NOS) activity was measured by conversion of [3H]-l-arginine to [3H]-l-citrulline, systemic TNF-α levels by ELISA, and reactive oxygen species (ROS) by immunofluorescence. KEY FINDINGS: We report upregulation of TLR4 in CSMC following AngII or LPS stimulation. In AngII-infused mice, chronic treatment with anti-TLR4 antibody (28±2.1%) attenuates adrenergic CC contraction, which also ameliorates nitrergic (68.90±0.21 vs. 51.07±0.63, 8Hz, AngII-infused mice treated vs. non-treated). Decreased endothelial NOS expression, reduced NOS activity, and augmented levels of TNF-α, and ROS were found following AngII-infusion. These alterations were prevented, or at least decreased by anti-TLR4 antibody treatment. SIGNIFICANCE: Inhibition of TLR4 ameliorates AngII-impaired cavernosal relaxation, decreases TNF-α levels, and restores NO bioavailability, demonstrating that TLR4 partly mediates AngII-induced cavernosal dysfunction.


Angiotensin II/immunology , Erectile Dysfunction/immunology , Nitric Oxide/immunology , Toll-Like Receptor 4/immunology , Animals , Blood Pressure , Erectile Dysfunction/physiopathology , Male , Mice, Inbred C57BL , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type III/immunology , Penis/immunology , Penis/physiopathology , Tumor Necrosis Factor-alpha/immunology
15.
J Cardiol ; 70(3): 201-205, 2017 Sep.
Article En | MEDLINE | ID: mdl-28400080

Vaccines are primarily used worldwide as a preventive medicine for infectious diseases and have recently been applied to cancer. We and others have developed therapeutic vaccines designed for cardiovascular diseases that are notably different from previous vaccines. In the case of cancer vaccines, a specific protein in cancer cells is a target antigen, and the activation of cytotoxic T cells (CTL) is required to kill and remove the antigen-presenting cancer cells. Our therapeutic vaccines work against hypertension by targeting angiotensin II (Ang II) as the antigen, which is an endogenous hormone. Therapeutic vaccines must avoid CTL activation and induce the blocking antibodies for Ang II. The goal of our therapeutic vaccine for cardiovascular diseases is to induce the specific antibody response toward the target protein without inducing T-cell or antibody-mediated inflammation through the careful selection of the target antigen, carrier protein and adjuvants. The goal of our therapeutic vaccine is similar to that of antibody therapy. Recently, multiple antibody-based drugs have been developed for cancer, immune-related diseases, and dyslipidemia, which are efficient but expensive. If the effect of a therapeutic vaccine is nearly equivalent to antibody therapy as an alternative approach, the lower medical cost and improvement in drug adherence can be advantages of therapeutic vaccines. In this review, we will describe our concept of therapeutic vaccines for cardiovascular diseases and the future directions of therapeutic vaccines as novel antibody therapies.


Antibodies/therapeutic use , Cardiovascular Diseases/therapy , Vaccines/therapeutic use , Angiotensin II/immunology , Animals , Autoantigens/immunology , Cardiovascular Diseases/immunology , Humans
16.
Stroke ; 48(5): 1362-1368, 2017 05.
Article En | MEDLINE | ID: mdl-28364024

BACKGROUND AND PURPOSE: Medication nonadherence is one of major risk factors for the poor outcome in ischemic stroke. Vaccination is expected to solve such a problem because of its long-lasting effects, but its effect on ischemic brain damage is still unknown. Here, we focused on vaccination for renin-angiotensin system and examined the effects of angiotensin II (Ang II) peptide vaccine in permanent middle cerebral artery occlusion model in rats. METHODS: Male Wistar rats were exposed to permanent middle cerebral artery occlusion after 3× injections of Ang II peptide vaccine, and the serum or brain level of anti-Ang II antibody was examined. The effects of the vaccine were evaluated by differences in infarction volume, brain renin-angiotensin system components, and markers for neurodegeneration and oxidative stress. RESULTS: Ang II vaccination successfully produced anti-Ang II antibodies in serum without concomitant change in blood pressure. Sufficient production of serum anti-Ang II antibody led to reduction of infarct volume and induced the penetration of anti-Ang II antibody in ischemic hemisphere, with suppressed expression of Ang II type 1 receptor mRNA. Vaccinated rats with sufficient antibody production showed the reduction of Fluoro-Jade B-positive cells, spectrin fragmentation, 4-hydroxynonenal-positive cells, and Nox 2 mRNA expression. CONCLUSIONS: Our findings indicate that Ang II vaccination exerts neuroprotective and antioxidative effects in cerebral ischemia, with renin-angiotensin system blockade by penetration of anti-Ang II antibodies into ischemic brain lesion. Ang II peptide vaccination could be a promising approach to treat ischemic stroke.


Angiotensin II/immunology , Antibodies/immunology , Brain Ischemia/immunology , Brain Ischemia/prevention & control , Immunotherapy, Active/methods , Infarction, Middle Cerebral Artery/immunology , Oxidative Stress/immunology , Renin-Angiotensin System/immunology , Stroke/immunology , Stroke/prevention & control , Vaccines, Subunit/immunology , Animals , Antibodies/blood , Disease Models, Animal , Male , Rats , Rats, Wistar
17.
Sci Rep ; 7: 43920, 2017 03 07.
Article En | MEDLINE | ID: mdl-28266578

A peptide vaccine targeting angiotensin II (Ang II) was recently developed as a novel treatment for hypertension to resolve the problem of noncompliance with pharmacotherapy. Ang II plays a crucial role in the pathogenesis of cardiac remodeling after myocardial infarction (MI), which causes heart failure. In the present study, we examined whether the Ang II vaccine is effective in preventing heart failure. The injection of the Ang II vaccine in a rat model of MI attenuated cardiac dysfunction in association with an elevation in the serum anti-Ang II antibody titer. Furthermore, any detrimental effects of the Ang II vaccine were not observed in the rats that underwent sham operations. Treatment with immunized serum from Ang II vaccine-injected rats significantly suppressed post-MI cardiac dysfunction in MI rats and Ang II-induced remodeling-associated signaling in cardiac fibroblasts. Thus, our present study demonstrates that the Ang II vaccine may provide a promising novel therapeutic strategy for preventing heart failure.


Angiotensin II/metabolism , Heart Failure/prevention & control , Myocardial Infarction/complications , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vasoconstrictor Agents/antagonists & inhibitors , Angiotensin II/immunology , Animals , Disease Models, Animal , Rats , Treatment Outcome , Vasoconstrictor Agents/immunology
18.
Pharmacol Res ; 120: 88-96, 2017 Jun.
Article En | MEDLINE | ID: mdl-28330785

Hypertension is a multifactorial disease. Although a number of different underlying mechanisms have been learned from the various experimental models of the disease, hypertension still poses challenges for treatment. Angiotensin II plays an unquestionable role in blood pressure regulation acting through central and peripheral mechanisms. During hypertension, dysregulation of the Renin-Angiotensin System is associated with increased expression of pro-inflammatory cytokines and reactive oxygen species causing kidney damage, endothelial dysfunction, and increase in sympathetic activity, among other damages, eventually leading to decline in organ function. Recent studies have shown that these effects involve both the innate and the adaptive immune response. The contribution of adaptive immune responses involving different lymphocyte populations in various models of hypertension has been extensively studied. However, the involvement of the innate immunity mediating inflammation in hypertension is still not well understood. The innate and adaptive immune systems intimately interact with one another and are essential to an effectively functioning of the immune response; hence, the importance of a better understanding of the underlying mechanisms mediating innate immune system during hypertension. In this review, we aim to discuss mechanisms linking Angiotensin II and the innate immune system, in the pathogenesis of hypertension. The newest research investigating Angiotensin II triggering toll like receptor 4 activation in the kidney, vasculature and central nervous system contributing to hypertension will be discussed. Understanding the role of the innate immune system in the development of hypertension may bring to light new insights necessary to improve hypertension management.


Angiotensin II/immunology , Hypertension/immunology , Toll-Like Receptor 4/immunology , Adaptive Immunity , Animals , Brain/immunology , Brain/pathology , Humans , Hypertension/complications , Hypertension/pathology , Immunity, Innate , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Kidney/immunology , Kidney/pathology , Renin-Angiotensin System , Signal Transduction
19.
Free Radic Biol Med ; 109: 48-60, 2017 08.
Article En | MEDLINE | ID: mdl-28108279

Oxidative stress is a major hallmark of cardiovascular diseases although a causal link was so far not proven by large clinical trials. However, there is a close association between oxidative stress and inflammation and increasing evidence for a causal role of (low-grade) inflammation for the onset and progression of cardiovascular diseases, which may serve as the missing link between oxidative stress and cardiovascular morbidity and mortality. With the present review we would like to highlight the multiple redox regulated pathways in inflammation, discuss the sources of reactive oxygen and nitrogen species that are of interest for these processes and finally discuss the importance of angiotensin II (AT-II) as a trigger of cardiovascular inflammation and the initiation and progression of cardiovascular diseases.


Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Mitochondria/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Angiotensin II/genetics , Angiotensin II/immunology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Cardiovascular System/immunology , Cardiovascular System/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Traps/immunology , Extracellular Traps/metabolism , Gene Expression Regulation , Humans , Inflammation , Mitochondria/immunology , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Oxidation-Reduction , Oxidative Stress , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , Signal Transduction
20.
Inflammation ; 40(1): 154-165, 2017 Feb.
Article En | MEDLINE | ID: mdl-27807688

Angiotensin II (Ang II) dysregulation has been determined as cause or an effect of many diseases. The relationship between Ang II and reactive oxygen species (ROS), which are generated by enzymes in the nicotinamide adenine dinucleotide phosphate oxidase (NOX) family, has been the focus of many researchers for years. Inflammation in response to the activities of various NOXs with differing time-dependent characteristics was reported. It is still unclear how these factors interplay over the course of the inflammatory response and how signal transduction through mitogen-activated protein kinase (MAPK) pathways. Our study collected data on the effects of Ang II on human umbilical vascular endothelial cells (HUVECs) over a comprehensive time period. Our results demonstrated that NOXs had two time-dependent reactions in response to Ang II stimulation via MAPK pathways. First, ROS was produced only during the early inflammatory phase. NOX4 promoted more rapid generation of H2O2 via the JNK pathway than generation of O2·- via ERK1/2 and p38 pathways. During both the early and late phases of the inflammatory response, NOX4 activity was transduced through the JNK pathway, whereas NOX1 and NOX2 signals were transmitted via the ERK1/2 and p38 pathways. Signal transduction via ROS generation was more likely during the early phase of the inflammatory response, and increased cytokine levels were more likely induced by the late phase of the inflammatory response.


Angiotensin II/immunology , Inflammation/etiology , MAP Kinase Signaling System/immunology , NADPH Oxidases/metabolism , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells , Humans , NADPH Oxidases/immunology , Reactive Oxygen Species/metabolism , Time Factors
...