Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Sci Rep ; 13(1): 10943, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37414864

RESUMEN

The association between constipation and cardiovascular risk is unclear. This population-level matched cohort study compared the association of constipation with hypertension and incident cardiovascular events in 541,172 hospitalized patients aged ≥ 60 years. For each constipation admission, one exact age-matched non-constipated admission was randomly selected from all hospitalizations within 2 weeks to form the comparison cohort. The association of constipation with hypertension and cardiovascular events (myocardial infarction, angina, stroke and transient ischemic attack) were analysed using a series of binary logistic regressions adjusting for age, sex, cardiovascular risk factors, gastrointestinal disorders and sociological factors. Patients with constipation had a higher multivariate-adjusted risk for hypertension (odds ratio [OR], 1.96; 95% confidence interval [CI] 1.94-1.99; P < 0.001). Compared to patients with neither constipation nor hypertension, there was a higher multivariate-adjusted risk for cardiovascular events in patients with constipation alone (OR, 1.58; 95% CI 1.55-1.61; P < 0.001) or hypertension alone (OR, 6.12; 95% CI 5.99-6.26; P < 0.001). In patients with both constipation and hypertension, the risk for all cardiovascular events appeared to be additive (OR, 6.53; 95% CI 6.40-6.66; P < 0.001). In conclusion, among hospital patients aged 60 years or older, constipation is linked to an increased risk of hypertension and cardiovascular events. These findings suggest that interventions to address constipation may reduce cardiovascular risk in elderly patients.


Asunto(s)
Enfermedades Cardiovasculares , Hipertensión , Accidente Cerebrovascular , Anciano , Humanos , Australia/epidemiología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Estudios de Cohortes , Estreñimiento/complicaciones , Estreñimiento/epidemiología , Hipertensión/complicaciones , Hipertensión/epidemiología , Pacientes Internos , Factores de Riesgo , Accidente Cerebrovascular/complicaciones
2.
Front Cardiovasc Med ; 10: 1184982, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37332591

RESUMEN

Introduction: Depletion of mature B cells affords protection against experimental hypertension. However, whether B cell-mediated hypertension is dependent on differentiation into antibody-secreting cells (ASCs) remains unclear. Using the proteasome inhibitor, bortezomib, the present study tested the effect of ASC reduction on angiotensin II-induced hypertension. Methods: Male C57BL6/J mice were infused with angiotensin II (0.7 mg/kg/day; s.c.) for 28 days via osmotic minipump to induce hypertension. Normotensive control mice received saline infusion. Bortezomib (750 µg/kg) or vehicle (0.1% DMSO) was administered (i.v.) 3 days prior to minipump implantation, and twice weekly thereafter. Systolic blood pressure was measured weekly using tail-cuff plethysmography. Spleen and bone marrow B1 (CD19+B220-), B2 (B220+CD19+) and ASCs (CD138hiSca-1+Blimp-1+) were enumerated by flow cytometry. Serum immunoglobulins were quantified using a bead-based immunoassay. Results: Bortezomib treatment reduced splenic ASCs by ∼68% and ∼64% compared to vehicle treatment in normotensive (2.00 ± 0.30 vs. 0.64 ± 0.15 × 105 cells; n = 10-11) and hypertensive mice (0.52 ± 0.11 vs. 0.14 ± 0.02 × 105 cells; n = 9-11), respectively. Bone marrow ASCs were also reduced by bortezomib in both normotensive (4.75 ± 1.53 vs. 1.71 ± 0.41 × 103 cells; n = 9-11) and hypertensive mice (4.12 ± 0.82 vs. 0.89 ± 0.18 × 103 cells; n = 9-11). Consistent with ASC reductions, bortezomib reduced serum IgM and IgG2a in all mice. Despite these reductions in ASCs and antibody levels, bortezomib did not affect angiotensin II-induced hypertension over 28 days (vehicle: 182 ± 4 mmHg vs. bortezomib: 177 ± 7 mmHg; n = 9-11). Conclusion: Reductions in ASCs and circulating IgG2a and IgM did not ameliorate experimental hypertension, suggesting other immunoglobulin isotypes or B cell effector functions may promote angiotensin II-induced hypertension.

4.
Commun Biol ; 6(1): 265, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36914713

RESUMEN

Atherosclerotic plaque rupture leading to myocardial infarction is a major global health burden. Applying the tandem stenosis (TS) mouse model, which distinctively exhibits the characteristics of human plaque instability/rupture, we use quantitative proteomics to understand and directly compare unstable and stable atherosclerosis. Our data highlight the disparate natures and define unique protein signatures of unstable and stable atherosclerosis. Key proteins and pathway networks are identified such as the innate immune system, and neutrophil degranulation. The latter includes calprotectin S100A8/A9, which we validate in mouse and human unstable plaques, and we demonstrate the plaque-stabilizing effects of its inhibition. Overall, we provide critical insights into the unique proteomic landscape of unstable atherosclerosis (as distinct from stable atherosclerosis and vascular tissue). We further establish the TS model as a reliable preclinical tool for the discovery and testing of plaque-stabilizing drugs. Finally, we provide a knowledge resource defining unstable atherosclerosis that will facilitate the identification and validation of long-sought-after therapeutic targets and drugs for plaque stabilization.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Humanos , Animales , Ratones , Placa Aterosclerótica/tratamiento farmacológico , Proteómica , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/metabolismo , Modelos Animales de Enfermedad
5.
Expert Opin Ther Targets ; 27(3): 225-238, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36946552

RESUMEN

INTRODUCTION: Myocarditis is a severe lymphocyte-mediated inflammatory disorder of the heart, mostly caused by viruses and immune checkpoint inhibitors (ICIs). Recently, myocarditis as a rare adverse event of mRNA vaccines for SARS-CoV-2 has caused global attention. The clinical consequences of myocarditis can be very severe, but specific treatment options are lacking or not yet clinically proven. AREAS COVERED: This paper offers a brief overview of the biology of viruses that frequently cause myocarditis, focusing on mechanisms important for viral entry and replication following host infection. Current and new potential therapeutic targets/strategies especially for viral myocarditis are reviewed systematically. In particular, the immune system in myocarditis is dissected with respect to infective viral and non-infective, ICI-induced myocarditis. EXPERT OPINION: Vaccination is an excellent emerging preventative strategy for viral myocarditis, but most vaccines still require further development. Anti-viral treatments that inhibit viral replication need to be considered following viral infection in host myocardium, as lower viral load reduces inflammation severity. Understanding how the immune system continues to damage the heart even after viral clearance will define novel therapeutic targets/strategies. We propose that viral myocarditis can be best treated using a combination of antiviral agents and immunotherapies that control cytotoxic T cell activity.


Asunto(s)
COVID-19 , Miocarditis , Humanos , Miocarditis/terapia , Miocarditis/tratamiento farmacológico , Vacunas contra la COVID-19/efectos adversos , COVID-19/terapia , COVID-19/complicaciones , SARS-CoV-2 , Miocardio , Antivirales/uso terapéutico
6.
Front Immunol ; 13: 1040233, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36483558

RESUMEN

Aims: Cardiac fibrosis is central to heart failure (HF), especially HF with preserved ejection fraction (HFpEF), often caused by hypertension. Despite fibrosis causing diastolic dysfunction and impaired electrical conduction, responsible for arrhythmia-induced sudden cardiac death, the mechanisms are poorly defined and effective therapies are lacking. Here we show that crosstalk between cardiac cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is essential for development of non-ischemic hypertensive cardiac fibrosis. Methods and results: CD8 T cell depletion in hypertensive mice, strongly attenuated CF, reduced cardiac apoptosis and improved ventricular relaxation. Interaction between cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is highly dependent on the CD8+ T cells expressing the innate stress-sensing receptor NKG2D and stressed cardiomyocytes expressing the NKG2D activating ligand RAE-1. The interaction between NKG2D and RAE-1 results in CD8+ T cell activation, release of perforin, cardiomyocyte apoptosis, increased numbers of TGF-ß1 expressing macrophages and fibrosis. Deleting NKG2D or perforin from CD8+ T cells greatly attenuates these effects. Activation of the cytoplasmic DNA-STING-TBK1-IRF3 signaling pathway in overly stressed cardiomyocytes is responsible for elevating RAE-1 and MCP-1, a macrophage attracting chemokine. Inhibiting STING activation greatly attenuates cardiomyocyte RAE-1 expression, the cardiomyocyte apoptosis, TGF-ß1 and fibrosis. Conclusion: Our data highlight a novel pathway by which CD8 T cells contribute to an early triggering mechanism in CF development; preventing CD8+ T cell activation by inhibiting the cardiomyocyte RAE-1-CD8+ T cell-NKG2D axis holds promise for novel therapeutic strategies to limit hypertensive cardiac fibrosis.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión , Animales , Ratones , Factor de Crecimiento Transformador beta1 , Insuficiencia Cardíaca/etiología , Volumen Sistólico , Fibrosis , Linfocitos T
7.
Small ; 18(31): e2200967, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35710979

RESUMEN

Atherosclerosis is a major cause of mortality and morbidity worldwide. Left undiagnosed and untreated, atherosclerotic plaques can rupture and cause cardiovascular complications such as myocardial infarction and stroke. Atherosclerotic plaques are composed of lipids, including oxidized low-density lipoproteins and cholesterol crystals, and immune cells, including macrophages. 2-Hydroxypropyl-beta-cyclodextrin (CD) is FDA-approved for capturing, solubilizing, and delivering lipophilic drugs in humans. It is also known to dissolve cholesterol crystals and decrease atherosclerotic plaque size. However, its low retention time necessitates high dosages for successful therapy. This study reports CD delivery via air-trapped polybutylcyanoacrylate nanoparticles (with diameters of 388 ± 34 nm) loaded with CD (CDNPs). The multimodal contrast ability of these nanoparticles after being loaded with IR780 dye in mice is demonstrated using ultrasound and near-infrared imaging. It is shown that CDNPs enhance the cellular uptake of CD in murine cells. In an ApoE-/- mouse model of atherosclerosis, treatment with CDNPs significantly improves the anti-atherosclerotic efficacy of CD. Ultrasound triggering further improves CD uptake, highlighting that CDNPs can be used for ultrasound imaging and ultrasound-responsive CD delivery. Thus, CDNPs represent a theranostic nanocarrier for potential application in patients with atherosclerosis.


Asunto(s)
Aterosclerosis , Ciclodextrinas , Nanopartículas , Placa Aterosclerótica , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/tratamiento farmacológico , Colesterol , Humanos , Ratones , Imagen Multimodal , Nanopartículas/química , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/tratamiento farmacológico , Medicina de Precisión , Ultrasonografía
8.
Br J Pharmacol ; 179(5): 979-997, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33368184

RESUMEN

The rupture of unstable atherosclerotic plaques is the major cause of cardiovascular mortality and morbidity. Despite significant limitations in our understanding and ability to identify unstable plaque pathology and prevent plaque rupture, most atherosclerosis research utilises preclinical animal models exhibiting stable atherosclerosis. Here, we introduce the tandem stenosis (TS) mouse model that reflects plaque instability and rupture, as seen in patients. The TS model involves dual ligation of the right carotid artery, leading to locally predefined unstable atherosclerosis in hypercholesterolaemic mice. It exhibits key characteristics of human unstable plaques, including plaque rupture, luminal thrombosis, intraplaque haemorrhage, large necrotic cores, thin or ruptured fibrous caps and extensive immune cell accumulation. Altogether, the TS model represents an ideal preclinical tool for improving our understanding of human plaque instability and rupture, for the development of imaging technologies to identify unstable plaques, and for the development and testing of plaque-stabilising treatments for the prevention of atherosclerotic plaque rupture. LINKED ARTICLES: This article is part of a themed issue on Preclinical Models for Cardiovascular disease research (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.5/issuetoc.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/patología , Constricción Patológica , Modelos Animales de Enfermedad , Humanos , Ratones , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/patología
9.
Thromb Haemost ; 122(2): 196-207, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34619795

RESUMEN

BACKGROUND: 3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases. METHODS: The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease-angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE-/- model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation. RESULTS: Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers. CONCLUSION: Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Aneurisma de la Aorta Abdominal/prevención & control , Aterosclerosis/prevención & control , Factor XIIa/antagonistas & inhibidores , Placa Aterosclerótica/metabolismo , Animales , Aneurisma de la Aorta Abdominal/epidemiología , Apolipoproteínas E , Modelos Animales de Enfermedad , Inflamación , Masculino , Ratones
11.
Eur Heart J ; 42(9): 938-947, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33338208

RESUMEN

AIMS: Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS: We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION: Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.


Asunto(s)
Alarminas , Aterosclerosis , Linfocitos B , Infarto del Miocardio , Placa Aterosclerótica , Animales , Aterosclerosis/etiología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células Plasmáticas
13.
J Leukoc Biol ; 107(5): 713-715, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31608500

RESUMEN

Discussion on Tregs that have anti-inflammatory and anti-atherogenic properties as an ideal therapeutic target to reduce fatal cardiovascular deaths following major noncardiac surgery.


Asunto(s)
Aterosclerosis , Linfocitos T Reguladores , Humanos , Complicaciones Posoperatorias/etiología , Factores de Riesgo
16.
Front Immunol ; 10: 3046, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998318

RESUMEN

Interaction between B and CD4 T cells is crucial for their optimal responses in adaptive immunity. Immune responses augmented by their partnership promote chronic inflammation. Here we report that interaction between B and CD4 T cells augments their atherogenicity to promote lipid-induced atherosclerosis. Genetic deletion of the gene encoding immunoglobulin mu (µ) heavy chain (µMT) in ApoE-/- mice resulted in global loss of B cells including those in atherosclerotic plaques, undetectable immunoglobulins and impaired germinal center formation. Despite unaffected numbers in the circulation and peripheral lymph nodes, CD4 T cells were also reduced in spleens as were activated and memory CD4 T cells. In hyperlipidemic µMT-/- ApoE-/- mice, B cell deficiency decreased atherosclerotic lesions, accompanied by absence of immunoglobulins and reduced CD4 T cell accumulation in lesions. Adoptive transfer of B cells deficient in either MHCII or co-stimulatory molecule CD40, molecules required for B and CD4 T cell interaction, into B cell-deficient µMT-/- ApoE-/- mice failed to increase atherosclerosis. In contrast, wildtype B cells transferred into µMT-/- ApoE-/- mice increased atherosclerosis and increased CD4 T cells in lesions including activated and memory CD4 T cells. Transferred B cells also increased their expression of atherogenic cytokines IL-1ß, TGF-ß, MCP-1, M-CSF, and MIF, with partial restoration of germinal centers and plasma immunoglobulins. Our study demonstrates that interaction between B and CD4 T cells utilizing MHCII and CD40 is essential to augment their function to increase atherosclerosis in hyperlipidemic mice. These findings suggest that targeting B cell and CD4 T cell interaction may be a therapeutic strategy to limit atherosclerosis progression.


Asunto(s)
Aterosclerosis/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Animales , Apolipoproteínas E/inmunología , Citocinas/inmunología , Inmunoglobulinas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Aterosclerótica/inmunología
17.
J Am Heart Assoc ; 7(13)2018 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-29936416

RESUMEN

BACKGROUND: Peritoneal B1a cells attenuate atherosclerosis by secreting natural polyclonal immunoglobulin M (IgM). Regulatory B cells expressing T-cell immunoglobulin mucin domain-1 (TIM-1) expanded through TIM-1 ligation by anti-TIM-1 monoclonal antibody (RMT1-10) induces immune tolerance. METHODS AND RESULTS: We examined the capacity of RMT1-10 to expand peritoneal B1a cells to prevent atherosclerosis development and retard progression of established atherosclerosis. RMT1-10 treatment selectively doubled peritoneal B1a cells, tripled TIM-1+ B1a cells and increased TIM-1+IgM+interleukin (IL)-10+ by 3-fold and TIM-1+IgM+IL-10- B1a cells by 2.5-fold. Similar expansion of B1a B cells was observed in spleens. These effects reduced atherosclerotic lesion size, increased plasma IgM and lesion IgM deposits, and decreased oxidatively modified low-density lipoproteins in lesions. Lesion CD4+ and CD8+ T cells, macrophages and monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, expression of proinflammatory cytokines monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, IL1ß, apoptotic cell numbers and necrotic cores were also reduced. RMT1-10 treatment failed to expand peritoneal B1a cells and reduce atherosclerosis after splenectomy that reduces B1a cells, indicating that these effects are B1a cell-dependent. Apolipoprotein E-KO mice fed a high-fat diet for 6 weeks before treatment with RMT1-10 also increased TIM-1+IgM+IL-10+ and TIM-1+IgM+IL-10- B1a cells and IgM levels and attenuated progression of established atherosclerosis. CONCLUSIONS: RMT1-10 treatment attenuates atherosclerosis development and progression by selectively expanding IgM producing atheroprotective B1a cells. Antibody-based in vivo expansion of B1a cells could be an attractive approach for treating atherosclerosis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Linfocitos B Reguladores/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptor Celular 1 del Virus de la Hepatitis A/antagonistas & inhibidores , Inmunoglobulina M/metabolismo , Animales , Antiinflamatorios/farmacología , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apoptosis/efectos de los fármacos , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Receptor Celular 1 del Virus de la Hepatitis A/inmunología , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Inmunoglobulina M/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Necrosis , Placa Aterosclerótica , Transducción de Señal/efectos de los fármacos
18.
Arterioscler Thromb Vasc Biol ; 38(5): e71-e84, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29599140

RESUMEN

OBJECTIVE: B cells promote or protect development of atherosclerosis. In this study, we examined the role of MHCII (major histocompatibility II), CD40 (cluster of differentiation 40), and Blimp-1 (B-lymphocyte-induced maturation protein) expression by follicular B (FO B) cells in development of atherosclerosis together with the effects of IgG purified from atherosclerotic mice. APPROACH AND RESULTS: Using mixed chimeric Ldlr-/- mice whose B cells are deficient in MHCII or CD40, we demonstrate that these molecules are critical for the proatherogenic actions of FO B cells. During development of atherosclerosis, these deficiencies affected T-B cell interactions, germinal center B cells, plasma cells, and IgG. As FO B cells differentiating into plasma cells require Blimp-1, we also assessed its role in the development of atherosclerosis. Blimp-1-deficient B cells greatly attenuated atherosclerosis and immunoglobulin-including IgG production, preventing IgG accumulation in atherosclerotic lesions; Blimp-1 deletion also attenuated lesion proinflammatory cytokines, apoptotic cell numbers, and necrotic core. To determine the importance of IgG for atherosclerosis, we purified IgG from atherosclerotic mice. Their transfer but not IgG from nonatherosclerotic mice into Ldlr-/- mice whose B cells are Blimp-1-deficient increased atherosclerosis; transfer was associated with IgG accumulating in atherosclerotic lesions, increased lesion inflammatory cytokines, apoptotic cell numbers, and necrotic core size. CONCLUSIONS: The mechanism by which FO B cells promote atherosclerosis is highly dependent on their expression of MHCII, CD40, and Blimp-1. FO B cell differentiation into IgG-producing plasma cells also is critical for their proatherogenic actions. Targeting B-T cell interactions and pathogenic IgG may provide novel therapeutic strategies to prevent atherosclerosis and its adverse cardiovascular complications.


Asunto(s)
Aterosclerosis/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Células Plasmáticas/inmunología , Linfocitos T/inmunología , Animales , Apoptosis , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Linfocitos B/metabolismo , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Centro Germinal/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunoglobulina G/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Necrosis , Fenotipo , Placa Aterosclerótica , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Vías Secretoras , Transducción de Señal , Linfocitos T/metabolismo
19.
Theranostics ; 7(13): 3192-3206, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28900504

RESUMEN

One of the major hurdles in intravenous regenerative cell therapy is the low homing efficiency to the area where these cells are needed. To increase cell homing toward areas of myocardial damage, we developed a bispecific tandem single-chain antibody (Tand-scFvSca-1+GPIIb/IIIa) that binds with high affinity to activated platelets via the activated glycoprotein (GP)IIb/IIIa receptor, and to a subset of peripheral blood mononuclear cells (PBMC) which express the stem cell antigen-1 (Sca-1) receptor. Methods: The Tand-scFvSca-1+GPIIb/IIIa was engineered, characterized and tested in a mouse model of ischemia-reperfusion (IR) injury applying left coronary artery occlusion for 60 min. Fluorescence cell tracking, cell infiltration studies, echocardiographic and histological analyses were performed. Results: Treatment of mice undergoing myocardial infarction with targeted-PBMCs led to successful cell delivery to the ischemic-reperfused myocardium, followed by a significant decrease in infiltration of inflammatory cells. Homing of targeted-PBMCs as shown by fluorescence cell tracking ultimately decreased fibrosis, increased capillary density, and restored cardiac function 4 weeks after ischemia-reperfusion injury. Conclusion: Tand-scFvSca-1+GPIIb/IIIa is a promising candidate to enhance therapeutic cell delivery in order to promote myocardial regeneration and thereby preventing heart failure.


Asunto(s)
Plaquetas/metabolismo , Pruebas de Función Cardíaca , Leucocitos Mononucleares/trasplante , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/terapia , Animales , Células CHO , Adhesión Celular , Cricetinae , Cricetulus , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamación/patología , Leucocitos/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Neovascularización Fisiológica , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Anticuerpos de Cadena Única/metabolismo , Remodelación Ventricular
20.
Eur J Pharmacol ; 816: 67-75, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-28483458

RESUMEN

Cytotoxic lymphocytes (killer cells) play a critical role in host defence mechanisms, protecting against infections and in tumour surveillance. They can also exert detrimental effects in chronic inflammatory disorders and in autoimmune diseases. Tissue cell death and necrosis are prominent features of advanced atherosclerotic lesions including vulnerable/unstable lesions which are largely responsible for most heart attacks and strokes. Evidence for accumulation of killer cells in both human and mouse lesions together with their cytotoxic potential strongly suggest that these cells contribute to cell death and necrosis in lesions leading to vulnerable plaque development and potentially plaque rupture. Killer cells can be divided into two groups, adaptive and innate immune cells depending on whether they require antigen presentation for activation. Activated killer cells detect damaged or stressed cells and kill by cytotoxic mechanisms that include perforin, granzymes, TRAIL or FasL and in some cases TNF-α. In this review, we examine current knowledge on killer cells in atherosclerosis, including CD8 T cells, CD28- CD4 T cells, natural killer cells and γδ-T cells, mechanisms responsible for their activation, their migration to developing lesions and effector functions. We also discuss pharmacological strategies to prevent their deleterious vascular effects by preventing/limiting their cytotoxic effects within atherosclerotic lesions as well as potential immunomodulatory therapies that might better target lesion-resident killer cells, to minimise any compromise of the immune system, which could result in increased susceptibility to infections and reductions in tumour surveillance.


Asunto(s)
Aterosclerosis/inmunología , Células Asesinas Naturales , Animales , Aterosclerosis/tratamiento farmacológico , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Terapia Molecular Dirigida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...