Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
PLoS Pathog ; 19(5): e1011409, 2023 05.
Article in English | MEDLINE | ID: mdl-37200377

ABSTRACT

The hallmark of severe COVID-19 involves systemic cytokine storm and multi-organ injury including testicular inflammation, reduced testosterone, and germ cell depletion. The ACE2 receptor is also expressed in the resident testicular cells, however, SARS-CoV-2 infection and mechanisms of testicular injury are not fully understood. The testicular injury could be initiated by direct virus infection or exposure to systemic inflammatory mediators or viral antigens. We characterized SARS-CoV-2 infection in different human testicular 2D and 3D culture systems including primary Sertoli cells, Leydig cells, mixed seminiferous tubule cells (STC), and 3D human testicular organoids (HTO). Data shows that SARS-CoV-2 does not productively infect any testicular cell type. However, exposure of STC and HTO to inflammatory supernatant from infected airway epithelial cells and COVID-19 plasma decreased cell viability and resulted in the death of undifferentiated spermatogonia. Further, exposure to only SARS-CoV-2 Envelope protein caused inflammatory response and cytopathic effects dependent on TLR2, while Spike 1 or Nucleocapsid proteins did not. A similar trend was observed in the K18-hACE2 transgenic mice which demonstrated a disrupted tissue architecture with no evidence of virus replication in the testis that correlated with peak lung inflammation. Virus antigens including Spike 1 and Envelope proteins were also detected in the serum during the acute stage of the disease. Collectively, these data strongly suggest that testicular injury associated with SARS-CoV-2 infection is likely an indirect effect of exposure to systemic inflammation and/or SARS-CoV-2 antigens. Data also provide novel insights into the mechanism of testicular injury and could explain the clinical manifestation of testicular symptoms associated with severe COVID-19.


Subject(s)
COVID-19 , Male , Mice , Animals , Humans , COVID-19/metabolism , Testis , SARS-CoV-2 , Bystander Effect , Inflammation/metabolism , Mice, Transgenic
2.
Skin Pharmacol Physiol ; 36(1): 27-37, 2023.
Article in English | MEDLINE | ID: mdl-36693328

ABSTRACT

INTRODUCTION: Tumor necrosis factor (TNF)-α released after follicular injury such as that caused by plucking plays a role in the activation of hair regeneration. Microneedle (MN) treatment is applied to the scalp to increase permeability and facilitate the delivery of any number of compounds. Because the MN treatment causes injury to the epidermis, albeit minor, we reasoned that this treatment would lead to a temporary TNF-α surge and thereby promote hair regeneration. METHODS: To investigate the effects of MN-treatment-induced microinjury and TNF-α on hair growth, we used C57BL/6N mice which were divided into six experimental groups: three groups of 1) negative control (NC), 2) plucking positive control (PK), and 3) MN therapy system (MTS) mice; and three groups identical to above were treated with a TNF-α blocker for 3 weeks: 4) NCB, 5) PKB, and 6) MTSB group. RESULTS: After injury, TNF-α surge occurred on day 3 in the PK group and on day 6 in the MTS group. Wnt proteins and VEGF expression were markedly increased in the PK group on day 3 and on day 6 in the MTS group compared to the NC group. Following wound healing, only MTS and PK groups displayed thickened epidermis and longer HF length. Within the 2 weeks following treatment, the rate of hair growth was much slower in the injured mice treated with the TNF-α blocker. CONCLUSION: Our findings indicate that microinjury stimulates the wound-healing mechanism via TNF-α/Wnt/VEGF surge to induce hair growth, and that blocking TNF-α inhibits this growth process.


Subject(s)
Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , Mice, Inbred C57BL , Wound Healing , Hair/metabolism , Regeneration
3.
Clin Sci (Lond) ; 135(24): 2763-2780, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34854902

ABSTRACT

The aim of the present study was to evaluate the effect of Compound 21 (C21), a selective AT2R agonist, on the prevention of endothelial dysfunction, extracellular matrix (ECM) remodeling and arterial stiffness associated with diet-induced obesity (DIO). Five-week-old male C57BL/6J mice were fed a standard (Chow) or high-fat diet (HF) for 6 weeks. Half of the animals of each group were simultaneously treated with C21 (1 mg/kg/day, in the drinking water), generating four groups: Chow C, Chow C21, HF C, and HF C21. Vascular function and mechanical properties were determined in the abdominal aorta. To evaluate ECM remodeling, collagen deposition and TGF-ß1 concentrations were determined in the abdominal aorta and the activity of metalloproteinases (MMP) 2 and 9 was analyzed in the plasma. Abdominal aortas from HF C mice showed endothelial dysfunction as well as enhanced contractile but reduced relaxant responses to Ang II. This effect was abrogated with C21 treatment by preserving NO availability. A left-shift in the tension-stretch relationship, paralleled by an augmented ß-index (marker of intrinsic arterial stiffness), and enhanced collagen deposition and MMP-2/-9 activities were also detected in HF mice. However, when treated with C21, HF mice exhibited lower TGF-ß1 levels in abdominal aortas together with reduced MMP activities and collagen deposition compared with HF C mice. In conclusion, these data demonstrate that AT2R stimulation by C21 in obesity preserves NO availability and prevents unhealthy vascular remodeling, thus protecting the abdominal aorta in HF mice against the development of endothelial dysfunction, ECM remodeling and arterial stiffness.


Subject(s)
Aorta, Abdominal/drug effects , Diet, High-Fat/adverse effects , Imidazoles/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Vascular Stiffness/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Collagen/metabolism , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Mice, Inbred C57BL , Obesity/metabolism , Receptor, Angiotensin, Type 2/agonists , Transforming Growth Factor beta1/blood
4.
Clin Sci (Lond) ; 135(9): 1145-1163, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33899912

ABSTRACT

Compound 21 (C21), a selective agonist of angiotensin II type 2 receptor (AT2R), induces vasodilation through NO release. Since AT2R seems to be overexpressed in obesity, we hypothesize that C21 prevents the development of obesity-related vascular alterations. The main goal of the present study was to assess the effect of C21 on thoracic aorta endothelial function in a model of diet-induced obesity (DIO) and to elucidate the potential cross-talk among AT2R, Mas receptor (MasR) and/or bradykinin type 2 receptor (B2R) in this response. Five-week-old male C57BL6J mice were fed a standard (CHOW) or a high-fat diet (HF) for 6 weeks and treated daily with C21 (1 mg/kg p.o) or vehicle, generating four groups: CHOW-C, CHOW-C21, HF-C, HF-C21. Vascular reactivity experiments were performed in thoracic aorta rings. Human endothelial cells (HECs; EA.hy926) were used to elucidate the signaling pathways, both at receptor and intracellular levels. Arteries from HF mice exhibited increased contractions to Ang II than CHOW mice, effect that was prevented by C21. PD123177, A779 and HOE-140 (AT2R, Mas and B2R antagonists) significantly enhanced Ang II-induced contractions in CHOW but not in HF-C rings, suggesting a lack of functionality of those receptors in obesity. C21 prevented those alterations and favored the formation of AT2R/MasR and MasR/B2R heterodimers. HF mice also exhibited impaired relaxations to acetylcholine (ACh) due to a reduced NO availability. C21 preserved NO release through PKA/p-eNOS and AKT/p-eNOS signaling pathways. In conclusion, C21 favors the interaction among AT2R, MasR and B2R and prevents the development of obesity-induced endothelial dysfunction by stimulating NO release through PKA/p-eNOS and AKT/p-eNOS signaling pathways.


Subject(s)
Endothelium, Vascular/drug effects , Imidazoles/therapeutic use , Proto-Oncogene Proteins/metabolism , Receptor, Angiotensin, Type 2/agonists , Receptor, Bradykinin B2/metabolism , Receptors, G-Protein-Coupled/metabolism , Sulfonamides/therapeutic use , Thiophenes/therapeutic use , Vascular Diseases/prevention & control , Animals , Aorta, Thoracic/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat , Drug Evaluation, Preclinical , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Obesity/complications , Obesity/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thiophenes/pharmacology , Vascular Diseases/etiology , Vascular Diseases/metabolism
5.
Platelets ; 32(8): 1038-1042, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-33222575

ABSTRACT

There are approximately 38 million people globally living with Human immunodeficiency virus 1 (HIV-1) and given the tremendous success of combination antiretroviral therapy (cART) this has dramatically reduced mortality and morbidity with prevention benefits. However, HIV-1 persists during cART within the human body and re-appears upon cART interruption. This HIV-1 reservoir remains a barrier to cure with cellular sites of viral persistence not fully understood. In this study we provide evidence corroborating a recently published article in STM demonstrating the role of platelets as a novel cellular disseminator of HIV-1 particles in the setting of viral suppression. Using classical transmission electron microscopy with and without immunogold labeling, we visualize HIV-1 in both platelets and monocytes in cART suppressed HIV donors. Our study suggests that due to the close proximity of platelets and monocytes an alternative life cycle of HIV-1 cycling within monocytes and platelets without the need of active replication under cART occurs. Our findings are supported by the lack of detectable HIV-1 particles in platelets derived from HIV uninfected donors or the 'Berlin' patient suggesting that platelets may serve as an underappreciated hidden bearer for HIV-1 and should be considered in HIV remission studies and trials.


Subject(s)
Blood Platelets/metabolism , HIV Infections/blood , HIV-1/pathogenicity , Humans
6.
Pharm Biol ; 58(1): 400-409, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32420784

ABSTRACT

Context: Although Salvia plebeia (SP) R. Brown (Labiatae) is known to possess various biological activities, the effects of SP on hair growth have not been elucidated.Objective: To investigate the hair growth potential of SP extract by using human dermal papilla cells (hDPCs) and C57BL/6 mice.Materials and methods: The entire SP plant sample was ground into powder and extracted with 99.9% methyl alcohol. Various concentrations of SP extract were added to hDPCs to evaluate the proliferation, migration, and factors related to hair growth and cycling. Effect of topical SP administration on hair regrowth was tested in vivo in male C57BL/6 mice for 21 days.Results: SP extract significantly increased the proliferation of cultured hDPCs at doses of 15.6 and 31.3 µg/mL compared to control group by 123% and 132%, respectively. Expression of hepatocyte growth factor increased while the level of TGF-ß1 and SMAD2/3 decreased when treated with SP extract. At the molecular level, the extract activated Wnt/ß-catenin signalling by raising ß-catenin and phospho-GSK3ß expression. SP extract also exerted anti-apoptotic and proliferative effects in hDPCs by increasing the Bcl-2/Bax ratio and activating cell proliferation-related proteins, ERK and Akt. Finally, the extract caused an induction of the anagen phase leading to significantly enhanced hair growth in treated male mice.Discussion and conclusion: Our results indicate that SP extract has the capacity to activate hDPCs into a proliferative state to promote hair growth. Further research is necessary to determine the bioactive components and their mechanisms of action responsible for SP-related hair growth effect.


Subject(s)
Hair Follicle/drug effects , Hair Follicle/growth & development , Plant Extracts/pharmacology , Salvia , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Hair/cytology , Hair/drug effects , Hair/growth & development , Hair Follicle/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Plant Extracts/isolation & purification
7.
Cytokine ; 122: 154169, 2019 10.
Article in English | MEDLINE | ID: mdl-28988706

ABSTRACT

IL-37 is a member of the IL-1 family, but unlike most other members of this family of cytokines, it has wide-ranging anti-inflammatory properties. Initially shown to bind IL-18 binding protein and prevent IL-18-mediated inflammation, its known role has been expanded to include distinct pathways, both intracellular involving the transcription factor Smad3, and extracellular via binding to the orphan receptor IL-1R8. A number of recent publications investigating the role of IL-37 in atherosclerosis and ischemic heart disease have revealed promising therapeutic value of the cytokine. Although research concerning the role of IL-37 and its mechanism in atherosclerosis is relatively scant, there are a number of well-known atherosclerotic processes that this cytokine can mediate with the potential of modulating the disease progression itself. This review will probe in detail the effects of IL-37 on important pathological processes such as inflammation, dysregulated lipid metabolism, and apoptosis, by analyzing existing data as well as exploring the potential of this cytokine to influence these properties.


Subject(s)
Atherosclerosis/metabolism , Interleukin-1/metabolism , Interleukin-1/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Atherosclerosis/drug therapy , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Interleukin-1/genetics , Interleukin-18/metabolism , Lipid Metabolism Disorders/drug therapy , Macrophages/drug effects , Macrophages/metabolism , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad3 Protein/metabolism
8.
J Immunol ; 199(10): 3604-3613, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29030487

ABSTRACT

Atherosclerosis, the progressive buildup of plaque within arterial blood vessels, can lead to fatal downstream events, such as heart attack or stroke. A key event contributing to the development of atherosclerosis is the infiltration of monocytes and its associated inflammation, as well as the formation of lipid-laden macrophage foam cells within the vessel wall. IL-37 is recognized as an important anti-inflammatory cytokine expressed especially by immune cells. This study was undertaken to elucidate the role of macrophage-expressed IL-37 in reducing the production and effects of proinflammatory cytokines, preventing foam cell formation, and reducing the development of atherosclerosis. Expression of human IL-37 was achieved with a macrophage-specific overexpression system, using the CD68 promoter in mouse primary bone marrow-derived macrophages via retroviral transduction. Macrophage IL-37 expression in vitro resulted in decreased mRNA (e.g., IL-1B, IL-6, and IL-12) and secreted protein production (e.g., IL-6, M-CSF, and ICAM-1) of key inflammatory mediators. IL-37 expression also inhibited macrophage proliferation, apoptosis, and transmigration, as well as reduced lipid uptake, compared with controls in vitro. The in vivo effects of macrophage-expressed IL-37 were investigated through bone marrow transplantation of transduced hematopoietic stem cells into irradiated atherosclerosis-prone Ldlr-/- mice. After 10 wk on a high-fat/high-cholesterol diet, mice with IL-37-expressing macrophages showed reduced disease pathogenesis, which was demonstrated by significantly less arterial plaque development and systemic inflammation compared with control mice. The athero-protective effect of macrophage-expressed IL-37 has implications for development of future therapies to treat atherosclerosis, as well as other chronic inflammatory diseases.


Subject(s)
Atherosclerosis/immunology , Hyperlipidemias/immunology , Interleukin-1/metabolism , Macrophages/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Diet, High-Fat , Humans , Inflammation Mediators/metabolism , Interleukin-1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity
9.
Int J Mol Sci ; 20(10)2019 May 19.
Article in English | MEDLINE | ID: mdl-31109146

ABSTRACT

Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early "inflammatory" stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the "reparative/proliferative" phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient's post-MI diagnoses.


Subject(s)
Cardiovascular Diseases/immunology , Immunity, Innate , Macrophages/immunology , Myocardium/immunology , Animals , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Heart/physiopathology , Humans , Macrophages/pathology , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/cytology , Myocardium/pathology , Protective Factors , Ventricular Remodeling
10.
PLoS Genet ; 11(5): e1005155, 2015 May.
Article in English | MEDLINE | ID: mdl-26020946

ABSTRACT

Recent genome wide association studies have identified a number of genes that contribute to the risk for coronary heart disease. One such gene, TCF21, encodes a basic-helix-loop-helix transcription factor believed to serve a critical role in the development of epicardial progenitor cells that give rise to coronary artery smooth muscle cells (SMC) and cardiac fibroblasts. Using reporter gene and immunolocalization studies with mouse and human tissues we have found that vascular TCF21 expression in the adult is restricted primarily to adventitial cells associated with coronary arteries and also medial SMC in the proximal aorta of mouse. Genome wide RNA-Seq studies in human coronary artery SMC (HCASMC) with siRNA knockdown found a number of putative TCF21 downstream pathways identified by enrichment of terms related to CAD, including "vascular disease," "disorder of artery," and "occlusion of artery," as well as disease-related cellular functions including "cellular movement" and "cellular growth and proliferation." In vitro studies in HCASMC demonstrated that TCF21 expression promotes proliferation and migration and inhibits SMC lineage marker expression. Detailed in situ expression studies with reporter gene and lineage tracing revealed that vascular wall cells expressing Tcf21 before disease initiation migrate into vascular lesions of ApoE-/- and Ldlr-/- mice. While Tcf21 lineage traced cells are distributed throughout the early lesions, in mature lesions they contribute to the formation of a subcapsular layer of cells, and others become associated with the fibrous cap. The lineage traced fibrous cap cells activate expression of SMC markers and growth factor receptor genes. Taken together, these data suggest that TCF21 may have a role regulating the differentiation state of SMC precursor cells that migrate into vascular lesions and contribute to the fibrous cap and more broadly, in view of the association of this gene with human CAD, provide evidence that these processes may be a mechanism for CAD risk attributable to the vascular wall.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Coronary Artery Disease/genetics , Myocytes, Smooth Muscle/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Lineage/genetics , Coronary Artery Disease/pathology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Humans , Mice , Myoblasts/metabolism , Myoblasts/pathology , Myocytes, Smooth Muscle/metabolism , Stem Cells
11.
Arterioscler Thromb Vasc Biol ; 41(6): 1837-1838, 2021 06.
Article in English | MEDLINE | ID: mdl-34038166
12.
BMC Complement Altern Med ; 17(1): 109, 2017 Feb 13.
Article in English | MEDLINE | ID: mdl-28193226

ABSTRACT

BACKGROUND: Geranium sibiricum L. has been used as a medicinal plant to treat diarrhea, bacterial infection, and cancer in Bulgaria, Peru, and Korea. However, its hair growth-promoting effect was not investigated so far. This study examined the effects of Geranium sibiricum L. extract (GSE) on hair growth, using in vitro and in vivo models. METHODS: Antioxidant, proliferation and migration assay of GSE was performed with human dermal papilla cells (hDPCs). Hair-growth promoting effect was measured in animal model. Relative expression of interleukin-1, vascular endothelial growth factor, hepatocyte growth factor, and transforming growth factor beta 1 was determined by real time RT-PCR. Expression of Ki-67 and stem cell factor were analyzed by immunohistochemistry. RESULTS: GSE treatment proliferated and migrated human dermal papilla cells (hDPCs) more than treatment of 10 µM minoxidil. GSE significantly stimulated the expression of Ki-67 protein and the mRNA levels of hepatocyte growth factor and vascular endothelial growth factor in hDPCs. Topical application of 1,000 ppm GSE for 3 weeks promoted more significant hair growth on shaved C57BL/6 mice than did 5% minoxidil. The histological morphology of hair follicles demonstrated an active anagen phase with the induction of stem cell factor. GSE treatment significantly reduced the number of mast cells and the expression of transforming growth factor beta 1 in mouse skin tissues. CONCLUSIONS: These results demonstrated that GSE promotes hair growth in vitro and in vivo by regulating growth factors and the cellular response.


Subject(s)
Dermis/drug effects , Geranium , Hair/drug effects , Hepatocyte Growth Factor/metabolism , Plant Extracts/pharmacology , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Dermis/metabolism , Hair/growth & development , Hair Follicle/drug effects , Hepatocyte Growth Factor/genetics , Humans , Ki-67 Antigen/metabolism , Male , Mast Cells/metabolism , Mice, Inbred C57BL , RNA, Messenger/metabolism , Stem Cell Factor/metabolism , Vascular Endothelial Growth Factor A/genetics
13.
Basic Res Cardiol ; 111(1): 7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26667317

ABSTRACT

Recent advances in basic cardiovascular research as well as their translation into the clinical situation were the focus at the last "New Frontiers in Cardiovascular Research meeting". Major topics included the characterization of new targets and procedures in cardioprotection, deciphering new players and inflammatory mechanisms in ischemic heart disease as well as uncovering microRNAs and other biomarkers as versatile and possibly causal factors in cardiovascular pathogenesis. Although a number of pathological situations such as ischemia-reperfusion injury or atherosclerosis can be simulated and manipulated in diverse animal models, also to challenge new drugs for intervention, patient studies are the ultimate litmus test to obtain unequivocal information about the validity of biomedical concepts and their application in the clinics. Thus, the open and bidirectional exchange between bench and bedside is crucial to advance the field of ischemic heart disease with a particular emphasis of understanding long-lasting approaches in cardioprotection.


Subject(s)
Cardiovascular Diseases , Translational Research, Biomedical , Animals , Humans
14.
Basic Res Cardiol ; 111(6): 69, 2016 11.
Article in English | MEDLINE | ID: mdl-27743118

ABSTRACT

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.


Subject(s)
Cardiology/trends , Cardiovascular Diseases , Theranostic Nanomedicine/trends , Animals , Cardiology/methods , Humans
15.
Arterioscler Thromb Vasc Biol ; 35(2): 323-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25524771

ABSTRACT

OBJECTIVE: Macrophage foam cell formation is a key feature of atherosclerosis. Recent studies have shown that specific microRNAs (miRs) are regulated in modified low-density lipoprotein-treated macrophages, which can affect the cellular cholesterol homeostasis. Undertaking a genome-wide screen of miRs regulated in primary macrophages by modified low-density lipoprotein, miR-302a emerged as a potential candidate that may play a key role in macrophage cholesterol homeostasis. APPROACH AND RESULTS: The objective of this study was to assess the involvement of miR-302a in macrophage lipid homeostasis and if it can influence circulating lipid levels and atherosclerotic development when it is inhibited in a murine atherosclerosis model. We found that transfection of primary macrophages with either miR-302a or anti-miR-302a regulated the expression of ATP-binding cassette (ABC) transporter ABCA1 mRNA and protein. Luciferase reporter assays showed that miR-302a repressed the 3' untranslated regions (UTR) activity of mouse Abca1 by 48% and human ABCA1 by 45%. In addition, transfection of murine macrophages with miR-302a attenuated cholesterol efflux to apolipoprotein A-1 (apoA-1) by 38%. Long-term in vivo administration of anti-miR-302a to mice with low-density lipoprotein receptor deficiency (Ldlr(-/-)) fed an atherogenic diet led to an increase in ABCA1 in the liver and aorta as well as an increase in circulating plasma high-density lipoprotein levels by 35% compared with that of control mice. The anti-miR-302a-treated mice also displayed reduced atherosclerotic plaque size by ≈25% and a more stable plaque morphology with reduced signs of inflammation. CONCLUSIONS: These studies identify miR-302a as a novel modulator of cholesterol efflux and a potential therapeutic target for suppressing atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Diseases/blood , Atherosclerosis/blood , Cholesterol/blood , Macrophages/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoprotein A-I/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , COS Cells , Chlorocebus aethiops , Diet, High-Fat , Disease Models, Animal , Homeostasis , Humans , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Transfection
16.
Arterioscler Thromb Vasc Biol ; 35(2): 309-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25524774

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of cluster of differentiation 43 (CD43), an integral membrane glycoprotein with both proadhesive and antiadhesive activities, in atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor-deficient mice were lethally irradiated and reconstituted with either bone marrow from CD43(-/-) mice or from wild-type controls. We found that mice lacking the CD43 on their leukocytes had significantly less severe atherosclerosis and that, contrary to our expectation, macrophage infiltration into the vessel wall was not affected by the lack of CD43 in the leukocytes. However, we found that CD43 mediates cholesterol homeostasis in macrophages by facilitating cholesterol efflux. This resulted in a significant reduction in storage of cholesterol in the aorta of mice lacking CD43 in the leukocytes. CONCLUSIONS: CD43 may be an important mediator of macrophage lipid homeostasis, thereby affecting macrophage foam cell formation and ultimately atherosclerotic plaque development.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Leukocytes/metabolism , Leukosialin/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Down-Regulation , Leukocytes/immunology , Leukosialin/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
17.
J Immunol ; 192(2): 589-602, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337385

ABSTRACT

IL-32 is a multifaceted cytokine with a role in infections, autoimmune diseases, and cancer, and it exerts diverse functions, including aggravation of inflammation and inhibition of virus propagation. We previously identified IL-32 as a critical regulator of endothelial cell (EC) functions, and we now reveal that IL-32 also possesses angiogenic properties. The hyperproliferative ECs of human pulmonary arterial hypertension and glioblastoma multiforme exhibited a markedly increased abundance of IL-32, and, significantly, the cytokine colocalized with integrin αVß3. Vascular endothelial growth factor (VEGF) receptor blockade, which resulted in EC hyperproliferation, increased IL-32 three-fold. Small interfering RNA-mediated silencing of IL-32 negated the 58% proliferation of ECs that occurred within 24 h in scrambled-transfected controls. Reduction of IL-32 neither affected apoptosis (insignificant changes in Bak-1, Bcl-2, Bcl-xL, lactate dehydrogenase, annexin V, and propidium iodide) nor VEGF or TGF-ß levels, but siIL-32-transfected adult and neonatal ECs produced up to 61% less NO, IL-8, and matrix metalloproteinase-9, and up to 3-fold more activin A and endostatin. In coculture-based angiogenesis assays, IL-32γ dose-dependently increased tube formation up to 3-fold; an αVß3 inhibitor prevented this activity and reduced IL-32γ-induced IL-8 by 85%. In matrigel plugs loaded with IL-32γ, VEGF, or vehicle and injected into live mice, we observed the anticipated VEGF-induced increase in neocapillarization (8-fold versus vehicle), but unexpectedly, IL-32γ was equally angiogenic. A second signal such as IFN-γ was required to render cells responsive to exogenous IL-32γ; importantly, this was confirmed using a completely synthetic preparation of IL-32γ. In summary, we add angiogenic properties that are mediated by integrin αVß3 but VEGF-independent to the portfolio of IL-32, implicating a role for this versatile cytokine in pulmonary arterial hypertension and neoplastic diseases.


Subject(s)
Interleukins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Activins/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Endostatins/metabolism , Familial Primary Pulmonary Hypertension , Glioblastoma/embryology , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Integrin alphaVbeta3/metabolism , Interferon-gamma/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Nitrogen Oxides/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
Circulation ; 129(5): 598-606, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24201302

ABSTRACT

BACKGROUND: Atherosclerosis and vascular remodeling after injury are driven by inflammation and mononuclear cell infiltration. Extracellular RNA (eRNA) has recently been implicated to become enriched at sites of tissue damage and to act as a proinflammatory mediator. Here, we addressed the role of eRNA in high-fat diet-induced atherosclerosis and neointima formation after injury in atherosclerosis-prone mice. METHODS AND RESULTS: The presence of eRNA was revealed in atherosclerotic lesions from high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice in a time-progressive fashion. RNase activity in plasma increased within the first 2 weeks (44±9 versus 70±7 mU/mg protein; P=0.0012), followed by a decrease to levels below baseline after 4 weeks of high-fat diet (44±9 versus 12±2 mU/mg protein; P<0.0001). Exposure of bone marrow-derived macrophages to eRNA resulted in a concentration-dependent upregulation of the proinflammatory mediators tumor necrosis factor-α, arginase-2, interleukin-1ß, interleukin-6, and interferon-γ. In a model of accelerated atherosclerosis after arterial injury in apolipoprotein E-deficient (ApoE(-/-)) mice, treatment with RNase1 diminished the increased plasma level of eRNA evidenced after injury. Likewise, RNase1 administration reduced neointima formation in comparison with vehicle-treated ApoE(-/-) controls (25.0±6.2 versus 46.9±6.9×10(3) µm(2), P=0.0339) and was associated with a significant decrease in plaque macrophage content. Functionally, RNase1 treatment impaired monocyte arrest on activated smooth muscle cells under flow conditions in vitro and inhibited leukocyte recruitment to injured carotid arteries in vivo. CONCLUSIONS: Because eRNA is associated with atherosclerotic lesions and contributes to inflammation-dependent plaque progression in atherosclerosis-prone mice, its targeting with RNase1 may serve as a new treatment option against atherosclerosis.


Subject(s)
Extracellular Fluid/physiology , Plaque, Atherosclerotic/blood , RNA/physiology , Ribonucleases/physiology , Animals , Atherosclerosis/blood , Atherosclerosis/chemically induced , Atherosclerosis/drug therapy , Diet, High-Fat/adverse effects , Extracellular Fluid/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/drug therapy , RNA/blood , Ribonucleases/therapeutic use
20.
Am J Pathol ; 183(1): 313-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685110

ABSTRACT

Chitinase 1 (CHIT1) is secreted by activated macrophages. Chitinase activity is raised in atherosclerotic patient sera and is present in atherosclerotic plaque. However, the role of CHIT1 in atherosclerosis is unknown. Preliminary studies of atherosclerosis in cynomolgous monkeys revealed CHIT1 to be closely correlated with areas of macrophage infiltration. Thus, we investigated the effects of a chitinase inhibitor, allosamidin, on macrophage function in vitro and on atherosclerotic development in vivo. In RAW264.7 cells, allosamidin elevated monocyte chemoattractant protein 1 and tumor necrosis factor alpha expression, and increased activator protein 1 and nuclear factor-κB transcriptional activity. Although inducible nitric oxide synthase, IL-6, and IL-1ß expression were increased, Arg1 expression was decreased by chitinase inhibition, suggesting that suppression of CHIT1 activity polarizes macrophages into a M1 phenotype. Allosamidin decreased scavenger receptor AI, CD36, ABCA1, and ABCG1 expression which led to suppression of cholesterol uptake and apolipoprotein AI-mediated cholesterol efflux in macrophages. These effects were confirmed with CHIT1 siRNA transfection and CHIT1 plasmid transfection experiments in primary macrophages. Apolipoprotein E-deficient hyperlipidemic mice treated for 6 weeks with constant administration of allosamidin and fed an atherogenic diet showed aggravated atherosclerotic lesion formation. These data suggest that CHIT1 exerts protective effects against atherosclerosis by suppressing inflammatory responses and polarizing macrophages toward an M2 phenotype, and promoting lipid uptake and cholesterol efflux in macrophages.


Subject(s)
Acetylglucosamine/analogs & derivatives , Atherosclerosis/enzymology , Chitinases/antagonists & inhibitors , Enzyme Inhibitors/adverse effects , Macrophages/enzymology , Trisaccharides/adverse effects , Acetylglucosamine/adverse effects , Animals , Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Biomarkers/metabolism , Cell Line , Chitinases/metabolism , Cytokines/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL