Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Microbes Infect ; 26(5-6): 105343, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670216

RESUMEN

Hemozoin is a crystal synthesized by Plasmodium parasites during hemoglobin digestion in the erythrocytic stage. The hemozoin released when the parasites egress from the red blood cell, which is complexed with parasite DNA, is cleared from the circulation by circulating and tissue-resident monocytes and macrophages, respectively. Recently, we reported that intravenous administration of purified hemozoin complexed with Plasmodium berghei DNA (HzPbDNA) resulted in an innate immune response that blocked liver stage development of sporozoites that was dose-dependent and time-limited. Here, we further characterize the organismal, cellular, and molecular events associated with this protective innate response in the liver and report that a large proportion of the IV administered HzPbDNA localized to F4/80+ cells in the liver and that the rapid and strong protection against liver-stage development waned quickly such that by 1 week post-HzPbDNA treatment animals were fully susceptible to infection. RNAseq of the liver after IV administration of HzPbDNA demonstrated that the rapid and robust induction of genes associated with the acute phase response, innate immune activation, cellular recruitment, and IFN-γ signaling observed at day 1 was largely absent at day 7. RNAseq analysis implicated NK cells as the major cellular source of IFN-γ. In vivo cell depletion and IFN-γ neutralization experiments supported the hypothesis that tissue-resident macrophages and NK cells are major contributors to the protective response and the NK cell-derived IFN-γ is key to induction of the mechanisms that block sporozoite development in the liver. These findings advance our understanding of the innate immune responses that prevent liver stage malaria infection.


Asunto(s)
Hemoproteínas , Inmunidad Innata , Interferón gamma , Hígado , Malaria , Plasmodium berghei , Esporozoítos , Animales , Plasmodium berghei/inmunología , Esporozoítos/inmunología , Malaria/inmunología , Malaria/prevención & control , Malaria/parasitología , Hemoproteínas/inmunología , Ratones , Hígado/parasitología , Hígado/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Macrófagos/inmunología , Macrófagos/parasitología , ADN Protozoario/genética , Femenino
2.
Front Immunol ; 15: 1340373, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38375475

RESUMEN

Immune checkpoint inhibitors (ICIs) are specialized monoclonal antibodies (mAbs) that target immune checkpoints and their ligands, counteracting cancer cell-induced T-cell suppression. Approved ICIs like cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), its ligand PD-L1, and lymphocyte activation gene-3 (LAG-3) have improved cancer patient outcomes by enhancing anti-tumor responses. However, some patients are unresponsive, and others experience immune-related adverse events (irAEs), affecting organs like the lung, liver, intestine, skin and now the cardiovascular system. These cardiac irAEs include conditions like myocarditis, atherosclerosis, pericarditis, arrhythmias, and cardiomyopathy. Ongoing clinical trials investigate promising alternative co-inhibitory receptor targets, including T cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) and T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT). This review delves into the mechanisms of approved ICIs (CTLA-4, PD-1, PD-L1, and LAG-3) and upcoming options like Tim-3 and TIGIT. It explores the use of ICIs in cancer treatment, supported by both preclinical and clinical data. Additionally, it examines the mechanisms behind cardiac toxic irAEs, focusing on ICI-associated myocarditis and atherosclerosis. These insights are vital as ICIs continue to revolutionize cancer therapy, offering hope to patients, while also necessitating careful monitoring and management of potential side effects, including emerging cardiac complications.


Asunto(s)
Antineoplásicos Inmunológicos , Aterosclerosis , Sistema Cardiovascular , Miocarditis , Neoplasias , Humanos , Antígeno CTLA-4 , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Antígeno B7-H1/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A , Antineoplásicos Inmunológicos/efectos adversos , Receptor de Muerte Celular Programada 1/metabolismo , Miocarditis/etiología , Inmunoterapia/efectos adversos , Neoplasias/terapia , Anticuerpos Monoclonales/efectos adversos , Receptores Inmunológicos/uso terapéutico , Pulmón/patología , Sistema Cardiovascular/metabolismo , Aterosclerosis/etiología
3.
Arthritis Rheumatol ; 76(4): 553-565, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37997621

RESUMEN

OBJECTIVE: Arthritis associated with immune checkpoint inhibitor therapies highlights the importance of immune checkpoint expression for joint homeostasis. We investigated the role of programmed death ligand (PD-L) 1 in the synovium using a collagen-induced arthritis (CIA) mouse model. METHODS: We blocked PD-L1 using blocking antibodies during CIA and assessed the arthritis severity by clinical and histologic scoring. PD-L1 expression and the origin of synovial macrophages were investigated using flow cytometry and parabiosis. We used Cre-Lox mice to ascertain the protective role of PD-L1-expressing macrophages in arthritis. The immune profile of human and murine synovial PD-L1+ macrophages was determined by reverse transcriptase-polymerase chain reaction, flow cytometry, and single-cell RNA sequencing. RESULTS: Anti-PD-L1 antibody treatment during CIA worsened arthritis with increased immune cell infiltration compared with isotype control, supporting the regulatory role of PD-L1 in the joint. The main cells expressing PD-L1 in the synovium were macrophages. Using parabiosis, we showed that synovial PD-L1+ macrophages were both locally proliferating and partially replaced by the circulation. PD-L1+ macrophages had increased levels of MER proto-oncogene tyrosine kinase (MerTK) and interleukin (IL)-10 expression during acute CIA. Genetic depletion of PD-L1 on macrophages in LyzcrePD-L1fl/fl mice resulted in worsened CIA compared with controls. We found that human PD-L1+ macrophages in the synovium of healthy individuals and patients with rheumatoid arthritis express MerTK and IL-10. CONCLUSION: PD-L1+ macrophages with efferocytotic and anti-inflammatory characteristics protect the synovium from severe arthritis in the CIA mouse model. Tissue-protective, PD-L1-expressing macrophages are also present in the human synovium at homeostasis and during rheumatoid arthritis.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Humanos , Animales , Ratones , Antígeno B7-H1 , Tirosina Quinasa c-Mer/metabolismo , Membrana Sinovial/metabolismo , Modelos Animales de Enfermedad , Macrófagos
4.
iScience ; 26(10): 107990, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37829205

RESUMEN

Hypereosinophilic syndrome is a progressive disease with extensive eosinophilia that results in organ damage. Cardiac pathologies are the main reason for its high mortality rate. A better understanding of the mechanisms of eosinophil-mediated tissue damage would benefit therapeutic development. Here, we describe the cardiac pathologies that developed in a mouse model of hypereosinophilic syndrome. These IL-5 transgenic mice exhibited decreased left ventricular function at a young age which worsened with age. Mechanistically, we demonstrated infiltration of activated eosinophils into the heart tissue that led to an inflammatory environment. Gene expression signatures showed tissue damage as well as repair and remodeling processes. Cardiomyocytes from IL-5Tg mice exhibited significantly reduced contractility relative to wild type (WT) controls. This impairment may result from the inflammatory stress experienced by the cardiomyocytes and suggest that dysregulation of contractility and Ca2+ reuptake in cardiomyocytes contributes to cardiac dysfunction at the whole organ level in hypereosinophilic mice.

5.
Front Cardiovasc Med ; 10: 1227818, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37576111

RESUMEN

Introduction: Cardiac presentation of autoimmune sarcoidosis, known as cardiac sarcoidosis (CS), is a poorly understood disease with high mortality and low diagnosis rate. While CS is an immunological syndrome, little is known about how cardiac parenchymal and stromal cells mediate its pathogenesis. Moreover, while most current sarcoidosis research is based on research in pulmonary sarcoidosis (PS), it remains unclear how much both presentations of sarcoidosis overlap. To tackle these concerns, we leveraged publicly available sarcoidosis transcriptomic datasets. Methods: Two publicly available bronchoalveolar lavage single-cell RNA sequencing datasets were integrated to analyze PS relative to control. Additionally, two publicly available cardiac single-nucleus RNA sequencing datasets were integrated to analyze CS relative to control. Following integration, we ran cell-cell communication, transcription factor, and differential expression analyses on parenchymal, stromal, and immune subsets identified in our analysis. Results: Our analysis revealed that there was an expansion of stromal and immune cells in PS and CS. We also observed upregulation of Th17.1 and attenuated activation transcriptional profiles in the immune cells of CS and PS relative to control. Additionally, we found upregulation of pro-inflammatory and pro-fibrotic transcriptional profiles in the cardiac stromal cells of CS relative to control. We also found that cardiomyocytes exhibited upregulated cardiac stress and proliferation transcriptional profiles in CS relative to control. Conclusions: Our integrative transcriptomic analysis shows that despite tissue-specific differences, there are shared transcriptional trends between CS and PS. It also shows that stromal and parenchymal populations exhibit transcriptional trends that could explain their pathogenic role in CS.

6.
Int J Ophthalmol ; 15(2): 352-356, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186699

RESUMEN

AIM: To share the results of a national screening program for amblyopia in school children in the north of Jordan. METHODS: This is a prospective national screening study for amblyopia. The program rolls first and second-grade children (6 to 7 years old) in the north of Jordan. The eye examination included: best-corrected visual acuity, cover-uncover test, and cycloplegic retinoscopy. Monocular visual acuity was tested using an ETDRS visual acuity chart without correction. Moreover, children were tested with full cycloplegic refraction when the test criteria were met. Unilateral amblyopia was defined as a best-corrected visual acuity difference of 2 or more lines. In comparison, bilateral amblyopia was defined as a best-corrected visual acuity of 20/40 or worse in the best eye. RESULTS: The prevalence of amblyopia for the total sample tested (n=17 203) was 2.78% (n=479). The most common cause of amblyopia was hypermetropia (64.45%), followed by previous ocular surgeries (15.1%), myopia (10.43%), strabismus (9.39%), and congenital cataract (0.63%). CONCLUSION: This is the first and only study, identifing modifiable risk factors in Jordanian children with amblyopia. In their first couple of years of elementary education, many Jordanian children are affected by amblyopia and pass unnoticed. A more governmental effort is needed into screening programs to improve vision in the Jordanian population.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA