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











Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 743354, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34764959

RESUMEN

Macrophages are functionally plastic and can thus play different roles in various microenvironments. Testis is an immune privileged organ, and testicular macrophages (TMs) show special immunosuppressive phenotype and low response to various inflammatory stimuli. However, the underlying mechanism to maintain the immunosuppressive function of TMs remains unclear. S100A9, a small molecular Ca2+ binding protein, is associated with the immunosuppressive function of macrophages. However, no related research is available about S100A9 in mouse testis. In the present study, we explored the role of S100A9 in TMs. We found that S100A9 was expressed in TMs from postnatal to adulthood and contributed to maintaining the immunosuppressive phenotype of TMs, which is associated with the activation of PI3K/Akt pathway. S100A9 treatment promotes the polarization of bone marrow-derived macrophages from M0 to M2 in vitro. S100A9 was significantly increased in TMs following UPEC-infection and elevated S100A9 contributed to maintain the M2 polarization of TMs. Treatment with S100A9 and PI3K inhibitor decreased the proportion of M2-type TMs in control and UPEC-infected mouse. Our findings reveal a crucial role of S100A9 in maintaining the immunosuppressive function of TMs through the activation of PI3K/Akt pathway, and provide a reference for further understanding the mechanism of immunosuppressive function of TMs.


Asunto(s)
Calgranulina B/inmunología , Privilegio Inmunológico/inmunología , Macrófagos/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Testículo/inmunología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
2.
Front Immunol ; 12: 744324, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777357

RESUMEN

Maternal-fetal immune-tolerance occurs throughout the whole gestational trimester, thus a mother can accept a genetically distinct fetus without immunological aggressive behavior. HLA-G, one of the non-classical HLA class I molecules, is restricted-expression at extravillous trophoblast. It can concordantly interact with various kinds of receptors mounted on maternally immune cells residing in the uterus (e.g. CD4+ T cells, CD8+ T cells, natural killer cells, macrophages, and dendritic cells) for maintaining immune homeostasis of the maternal-fetus interface. HLA-G is widely regarded as the pivotal protective factor for successful pregnancies. In the past 20 years, researches associated with HLA-G have been continually published. Indeed, HLA-G plays a mysterious role in the mechanism of maternal-fetal immune-tolerance. It can also be ectopically expressed on tumor cells, infected sites and other pathologic microenvironments to confer a significant local tolerance. Understanding the characteristics of HLA-G in immunologic tolerance is not only beneficial for pathological pregnancy, but also helpful to the therapy of other immune-related diseases, such as organ transplant rejection, tumor migration, and autoimmune disease. In this review, we describe the biological properties of HLA-G, then summarize our understanding of the mechanisms of fetomaternal immunologic tolerance and the difference from transplant tolerance. Furthermore, we will discuss how HLA-G contributes to the tolerogenic microenvironment during pregnancy. Finally, we hope to find some new aspects of HLA-G in fundamental research or clinical application for the future.


Asunto(s)
Feto/inmunología , Antígenos HLA-G/inmunología , Privilegio Inmunológico/inmunología , Animales , Femenino , Humanos , Embarazo
3.
Cells ; 10(9)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34572009

RESUMEN

Cancer stem cells (CSCs) are broadly considered immature, multipotent, tumorigenic cells within the tumor mass, endowed with the ability to self-renew and escape immune control. All these features contribute to place CSCs at the pinnacle of tumor aggressiveness and (immune) therapy resistance. The immune privileged status of CSCs is induced and preserved by various mechanisms that directly affect them (e.g., the downregulation of the major histocompatibility complex class I) and indirectly are induced in the host immune cells (e.g., activation of immune suppressive cells). Therefore, deeper insights into the immuno-biology of CSCs are essential in our pursuit to find new therapeutic opportunities that eradicate cancer (stem) cells. Here, we review and discuss the ability of CSCs to evade the innate and adaptive immune system, as we offer a view of the immunotherapeutic strategies adopted to potentiate and address specific subsets of (engineered) immune cells against CSCs.


Asunto(s)
Privilegio Inmunológico/inmunología , Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Escape del Tumor/inmunología , Inmunidad Adaptativa/inmunología , Animales , Humanos , Inmunidad Innata/inmunología
4.
Front Immunol ; 12: 701935, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220866

RESUMEN

The eye and the brain have limited capacities for regeneration and as such, immune-mediated inflammation can produce devastating consequences in the form of neurodegenerative diseases of the central nervous system or blindness as a result of ocular inflammatory diseases such as uveitis. Accordingly, both the eye and the brain are designed to limit immune responses and inflammation - a condition known as "immune privilege". Immune privilege is sustained by physiological, anatomical, and regulatory processes that conspire to restrict both adaptive and innate immune responses.


Asunto(s)
Antígeno CD11c/inmunología , Córnea/inmunología , Células Dendríticas/inmunología , Privilegio Inmunológico/inmunología , Animales , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología
5.
Front Immunol ; 12: 673131, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054856

RESUMEN

Introduction: In pregnancy, the mother and fetus differ in HLA antigens, and yet the maternal immune system generally tolerates the fetus. KIR receptors expressed by maternal uterine NK cells at the maternal-fetal interface directly interact with HLA-C on extravillous trophoblast cells for optimal placental development. In this study, we aimed to determine whether there is a preferential selection for HLA compatibility and specific KIR/HLA-C combinations in uncomplicated and preeclamptic naturally conceived pregnancies compared to what would be expected by chance. Methods: Genotyping for maternal and fetal HLA-A, -B, -C, -DR, and -DQ, and maternal KIR was performed for 451 uncomplicated pregnancies and 77 pregnancies complicated with preeclampsia. The number of HLA antigen (mis)matches between mother and fetus was calculated and compared to expected values obtained by randomization of the HLA haplotype, inherited from the father, over the existing maternal haplotype of the fetuses. A similar methodology was executed for analysis of the KIR/HLA-C data (n=309). Results: In uncomplicated pregnancies, the degree of maternal-fetal HLA matching was not different than expected-by-chance values. In preeclamptic pregnancies, the degree of maternal-fetal HLA matching was different in observed compared to expected-by-chance values (p=0.012). More specifically, the degree of maternal-fetal matching of HLA-C was higher in the actual preeclamptic pregnancies than was expected-by-chance (p=0.007). Preeclamptic pregnancies showed an overall tendency towards higher maternal-fetal HLA compatibility, for total HLA matches (p=0.021), HLA class I (p=0.038) and HLA-C (p=0.025) compared to uncomplicated pregnancies. Conclusion: The data suggest that there is no preferential selection of maternal-fetal HLA compatibility in uncomplicated pregnancies. In contrast, increased total HLA, HLA class I and, especially, HLA-C compatibility is associated with preeclampsia, suggestive for a role of HLA mismatches in immune regulation leading to uncomplicated pregnancy.


Asunto(s)
Antígenos HLA/inmunología , Histocompatibilidad Materno-Fetal/inmunología , Preeclampsia/inmunología , Receptores KIR/inmunología , Adulto , Femenino , Humanos , Privilegio Inmunológico/inmunología , Embarazo
6.
Front Immunol ; 12: 641281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763083

RESUMEN

Background: Female Genital Tract (FGT) is an important micro-ecological area of human body. Microbiota in the lower reproductive tract may subsequently invade the uterine cavity during embryo implantation and produce immune responses. CBA/J×DBA/2 mating combination has been widely used as an abortion-prone mice model but whether microbiota existed in their uterine cavity remains unclear. In this context, the role of the microbial communities in immune response deserves attention. Objective: To investigate the relationship between the distribution of microbiota in the uterine cavity of CBA/J×DBA/2 abortion-prone mouse model and the immune imbalance of the maternal-fetal interface. Methods: In this study, female CBA/J mice were paired with male DBA/2 mice to develop an abortion-prone model (BA group), and with male BALB/c mice to build a standard pregnancy model (BC group). The non-pregnant female mice were served as the control group (C group). Uterine flushing fluid and sera were collected on day 13.5 of pregnancy. 16S rRNA sequencing technology was used to analyze the distribution of intrauterine microbiota. Phylogenetic Investigation of Communities were conducted to predict the microbiota functions by Reconstruction of Unobserved States (PICRUST) and Kyoto Encyclopedia of Genes and Genomes (KEGG). The serum IL 10, INF-γ, and TNF-α levels were examined using Enzyme-linked immunosorbent assay (ELISA) method. Results: All samples were detected with microbial communities. The α diversity (p = 0.00077) had significant differences among three groups. Proteobacteria was the most dominant phylum in C group (mean = 83.21%) and BA group (mean = 43.23%). Firmicutes was dominant in BC group (mean = 46.4%), as well as the second dominant one in C group (mean = 12.63%) and BA group (mean = 40.55%). Microbiota functions were associated with metabolism and immune response through the NOD-like receptor signaling pathway. The serum IL 10 level in BA group were significantly lower than that in BC group (10.14 ± 1.90 pg/ml, n = 8; vs. 19.03 ± 1.82 pg/ml, n = 10; p = 0.004). The serum TNF-α and INF-γ level in BA group were also significantly higher than that in BC group (523.1 ± 58.14 pg/ml, n = 8 vs. 310.3 ± 28.51 pg/ml, n = 10, p = 0.0029; 69.22 ± 5.38 pg/ml, n = 8 vs. 50.85 ± 2.45 pg/ml, n = 10, p = 0.0042). Conclusion: Microbial communities were colonized in uterine cavity of CBA/J mice both at non-pregnant stage and pregnant stage when mated with both BALB/c and DBA/2 male mice. The differentially abundant microbiome may be attributed to the immune tolerance through binding to the NOD-like receptor.


Asunto(s)
Aborto Espontáneo/inmunología , Aborto Espontáneo/microbiología , Útero/inmunología , Útero/microbiología , Animales , Modelos Animales de Enfermedad , Femenino , Privilegio Inmunológico/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Embarazo
7.
Front Immunol ; 12: 642392, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717198

RESUMEN

The success of pregnancy relies on the fine adjustment of the maternal immune system to tolerate the allogeneic fetus. Trophoblasts carrying paternal antigens are the only fetal-derived cells that come into direct contact with the maternal immune cells at the maternal-fetal interface. The crosstalk between trophoblasts and decidual immune cells (DICs) via cell-cell direct interaction and soluble factors such as chemokines and cytokines is a core event contributing to the unique immunotolerant microenvironment. Abnormal trophoblasts-DICs crosstalk can lead to dysregulated immune situations, which is well known to be a potential cause of a series of pregnancy complications including recurrent spontaneous abortion (RSA), which is the most common one. Immunotherapy has been applied to RSA. However, its development has been far less rapid or mature than that of cancer immunotherapy. Elucidating the mechanism of maternal-fetal immune tolerance, the theoretical basis for RSA immunotherapy, not only helps to understand the establishment and maintenance of normal pregnancy but also provides new therapeutic strategies and promotes the progress of immunotherapy against pregnancy-related diseases caused by disrupted immunotolerance. In this review, we focus on recent progress in the maternal-fetal immune tolerance mediated by trophoblasts-DICs crosstalk and clinical application of immunotherapy in RSA. Advancement in this area will further accelerate the basic research and clinical transformation of reproductive immunity and tumor immunity.


Asunto(s)
Decidua/inmunología , Privilegio Inmunológico/inmunología , Trofoblastos/inmunología , Aborto Habitual/inmunología , Aborto Habitual/terapia , Animales , Femenino , Humanos , Inmunoterapia/métodos , Embarazo
8.
Front Immunol ; 12: 758267, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046934

RESUMEN

Although the concepts related to fetal immune tolerance proposed by Sir Peter Medawar in the 1950s have not withstood the test of time, they revolutionized our current understanding of the immunity at the maternal-fetal interface. An important extension of the original Medawar paradigm is the investigation into the underlying mechanisms for adverse pregnancy outcomes, including recurrent spontaneous abortion, preterm birth, preeclampsia and gestational diabetes mellitus (GDM). Although a common pregnancy complication with systemic symptoms, GDM still lacks understanding of immunological perturbations associated with the pathological processes, particularly at the maternal-fetal interface. GDM has been characterized by low grade systemic inflammation that exacerbates maternal immune responses. In this regard, GDM may also entail mild autoimmune pathology by dysregulating circulating and uterine regulatory T cells (Tregs). The aim of this review article is to focus on maternal-fetal immunological tolerance phenomenon and discuss how local or systemic inflammation has been programmed in GDM. Specifically, this review addresses the following questions: Does the inflammatory or exhausted Treg population affecting the Th17:Treg ratio lead to the propensity of a pro-inflammatory environment? Do glycans and glycan-binding proteins (mainly galectins) contribute to the biology of immune responses in GDM? Our understanding of these important questions is still elementary as there are no well-defined animal models that mimic all the features of GDM or can be used to better understand the mechanistic underpinnings associated with this common pregnancy complication. In this review, we will leverage our preliminary studies and the literature to provide a conceptualized discussion on the immunobiology of GDM.


Asunto(s)
Diabetes Gestacional/inmunología , Privilegio Inmunológico/inmunología , Animales , Femenino , Humanos , Embarazo
9.
Exp Dermatol ; 30(4): 522-528, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33103270

RESUMEN

The skin forms a barrier that prevents dehydration and keeps us safe from pathogens. To ensure proper function, the skin possesses a myriad of stem cell populations that are essential for maintenance and repair upon damage. In order to protect, the skin is also an active immunological site, with abundant resident immune cells and strong recruitment of even more immune cells during wounding or infection. Such active and strong immunity makes the skin susceptible to a diverse spectrum of autoimmune diseases, such as vitiligo and alopecia areata. Conversely, despite constant immune surveillance, the skin is also a tissue where frequent malignancies occur, which suggests that immune evasion must also take place. Skin stem cells play a crucial role during both regeneration and tumorigenesis. How immune cells, and in particular T cells, interact with skin stem cells and the implications this crosstalk has in skin disease (both autoimmunity and cancer) is not fully understood. Uncovering the mechanisms governing immune-stem cells interactions in the skin is critical for the development of new therapeutic strategies to safeguard susceptible cells during autoimmunity and, conversely, to improve cancer immunotherapy. Here, I will discuss how distinct skin stem cell populations are attacked by, or conversely, cloaked from immune cells, and the implications their differences have in autoimmunity and cancer.


Asunto(s)
Autoinmunidad/inmunología , Privilegio Inmunológico/inmunología , Neoplasias Cutáneas/inmunología , Piel/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Humanos
10.
Int J Mol Sci ; 21(11)2020 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-32486493

RESUMEN

The eye is provided with immune protection against pathogens in a manner that greatly reduces the threat of inflammation-induced vision loss. Immune-mediated inflammation and allograft rejection are greatly reduced in the eye, a phenomenon called 'immune privilege'. Corneal tissue has inherent immune privilege properties with underlying three mechanisms: (1) anatomical, cellular, and molecular barriers in the cornea; (2) an immunosuppressive microenvironment; and (3) tolerance related to regulatory T cells and anterior chamber-associated immune deviation. This review describes the molecular mechanisms of the immunosuppressive microenvironment and regulatory T cells in the cornea that have been elucidated from animal models of ocular inflammation, especially those involving corneal transplantation, it also provides an update on immune checkpoint molecules in corneal and systemic immune regulation, and its relevance for dry eye associated with checkpoint inhibitor therapy.


Asunto(s)
Córnea/inmunología , Síndromes de Ojo Seco/inmunología , Privilegio Inmunológico/inmunología , Sistema Inmunológico , Animales , Cámara Anterior/inmunología , Antígeno B7-H1/metabolismo , Trasplante de Córnea , Proteína Ligando Fas/metabolismo , Rechazo de Injerto/inmunología , Humanos , Tolerancia Inmunológica , Factores Inmunológicos , Inmunosupresores/uso terapéutico , Inflamación , Ligandos , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Receptor fas/metabolismo
11.
Front Immunol ; 11: 619408, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33643300

RESUMEN

In mammals, the uterine mucosal immune system simultaneously recognizes and reacts to most bacteria as well as allogenic sperm mainly through the Toll-like receptors (TLR)2/4 signaling pathway. Here, we characterized the impact of pathogen-derived TLR2/4 ligands (peptidoglycan (PGN)/lipopolysaccharide (LPS)) on the immune crosstalk of sperm with the bovine endometrial epithelium. The real-time PCR analysis showed that the presence of low levels of PGN, but not LPS, blocked the sperm-induced inflammatory responses in bovine endometrial epithelial cells (BEECs) in vitro. Immunoblotting analysis revealed that PGN prevented the sperm-induced phosphorylation of JNK in BEECs. Activation or blockade of the TLR2 system in the endometrial epithelium verified that TLR2 signaling acts as a commonly-shared pathway for PGN and sperm recognition. The impairment of endometrial sperm recognition, induced by PGN, subsequently inhibited sperm phagocytosis by polymorphonuclear neutrophils (PMNs). Moreover, using an ex vivo endometrial explant that more closely resembles those in vivo conditions, showed that sperm provoked a mild and reversible endometrial tissue injury and triggered PMN recruitment into uterine glands, while PGN inhibited these events. Of note, PGN markedly increased the sperm attachment to uterine glands, and relatively so in the surface epithelium. However, addition of the anti-CD44 antibody into a PGN-sperm-explant co-culture completely blocked sperm attachment into glands and surface epithelia, indicating that the CD44 adhesion molecule is involved in the PGN-triggered sperm attachment to the endometrial epithelium. Together, these findings demonstrate that, the presence of PGN residues disrupts sperm immune recognition and prevents the physiological inflammation induced by sperm in the endometrial epithelium via the MyD88-dependent pathway of TLR2 signaling, possibly leading to impairment of uterine clearance and subsequent embryo receptivity.


Asunto(s)
Endometrio/inmunología , Privilegio Inmunológico/inmunología , Peptidoglicano/inmunología , Espermatozoides/inmunología , Receptor Toll-Like 2/inmunología , Animales , Bovinos , Femenino , Inmunidad Mucosa/inmunología , Lipopolisacáridos/inmunología , Masculino , Embarazo
12.
Br J Cancer ; 121(8): 666-678, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31523055

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) have innate abilities to resist even the harshest of therapies. To eradicate CSCs, parallels can be drawn from signalling modules that orchestrate pluripotency. Notch-Hedgehog hyperactivation are seen in CSCs, yet, not much is known about their conserved roles in tumour progression across cancers. METHODS: Employing a comparative approach involving 21 cancers, we uncovered clinically-relevant, pan-cancer drivers of Notch and Hedgehog. GISTIC datasets were used to evaluate copy number alterations. Receiver operating characteristic and Cox regression were employed for survival analyses. RESULTS: We identified a Notch-Hedgehog signature of 13 genes exhibiting high frequencies of somatic amplifications leading to transcript overexpression. The signature successfully predicted patients at risk of death in five cancers (n = 2278): glioma (P < 0.0001), clear cell renal cell (P = 0.0022), papillary renal cell (P = 0.00099), liver (P = 0.014) and stomach (P = 0.011). The signature was independent of other clinicopathological parameters and offered an additional resolution to stratify similarly-staged tumours. High-risk patients exhibited features of stemness and had more hypoxic tumours, suggesting that hypoxia may influence CSC behaviour. Notch-Hedgehog+ CSCs had an immune privileged phenotype associated with increased regulatory T cell function. CONCLUSION: This study will set the stage for exploring adjuvant therapy targeting the Notch-Hedgehog axis to help optimise therapeutic regimes leading to successful CSC elimination.


Asunto(s)
Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Linfocitos T Reguladores/inmunología , Escape del Tumor/genética , Hipoxia Tumoral/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Variaciones en el Número de Copia de ADN , Bases de Datos Genéticas , Progresión de la Enfermedad , Neoplasias Endometriales/genética , Neoplasias Endometriales/inmunología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Glioma/genética , Glioma/inmunología , Proteínas Hedgehog/genética , Humanos , Privilegio Inmunológico/inmunología , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Masculino , Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Receptores Notch/genética , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología
13.
J Neuroimmunol ; 333: 476964, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31112803

RESUMEN

It has been hypothesized that anterior chamber-associated immune deviation (ACAID) to neural antigens induced prior to central nervous system injury can inhibit self-reactivity and lessen secondary degeneration. This work evaluated the effect of ACAID induced to three neural tissue-derived extracts (whole extract, cytosolic extract, CE; or organelle-membrane extract) prior to optic nerve injury on retinal ganglion cell (RGC) survival. The results show that only ACAID to the CE increased RGC survival at 7 and14 days post-injury (dpi). This effect was achieved by retinal polarization towards an anti-inflammatory profile, driven by regulatory T cells and M2-type macrophages at 7 dpi.


Asunto(s)
Cámara Anterior/inmunología , Autoantígenos/inmunología , Privilegio Inmunológico/inmunología , Traumatismos del Nervio Óptico/inmunología , Retina/inmunología , Animales , Autoinmunidad , Citosol/inmunología , Femenino , Hipersensibilidad Tardía/inmunología , Macrófagos/inmunología , Compresión Nerviosa , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Ratas , Ratas Wistar , Células Ganglionares de la Retina/inmunología , Linfocitos T Reguladores/inmunología
14.
World Neurosurg ; 120: 302-315, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30196171

RESUMEN

Glioblastoma is a highly aggressive neoplasm with an extremely poor prognosis. Despite maximal gross resection and chemoradiotherapy, these grade IV astrocytomas consistently recur. Glioblastoma cells exhibit numerous pathogenic mechanisms to decrease tumor immunogenicity while promoting gliomagenesis, which manifests clinically as a median survival of less than 2 years and few long-term survivors. Recent clinical trials of vaccine-based immunotherapeutics against glioblastoma have demonstrated encouraging results in prolonging progression-free survival and overall survival. Several vaccine-based treatments have been trialed, such as peptide and heat-shock proteins, dendritic cell-based vaccines, and viral-based immunotherapy. In this literature review, we discuss the immunobiology of glioblastoma, significant current and completed vaccine-based immunotherapy clinical trials, and broad clinical challenges and future directions of glioblastoma vaccine-based immunotherapeutics.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia/métodos , Vacunas/uso terapéutico , Encéfalo/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Ensayos Clínicos como Asunto , Glioblastoma/inmunología , Glioblastoma/mortalidad , Humanos , Privilegio Inmunológico/inmunología , Tolerancia Inmunológica/inmunología , Inmunoterapia/tendencias , Células Madre Neoplásicas/inmunología , Supervivencia sin Progresión , Microambiente Tumoral/inmunología , Vacunas/inmunología
15.
Invest Ophthalmol Vis Sci ; 59(11): 4738-4747, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30267096

RESUMEN

Purpose: Severing of corneal nerves in preparation of corneal transplantation abolishes immune privilege of subsequent corneal transplants placed into either eye: a phenomenon termed sympathetic loss of immune privilege (SLIP). SLIP is due to the disabling of T regulatory cells (Tregs) by CD11c+ contrasuppressor (CS) cells. This study characterized the induction, function, and manipulation of CS cell activity and the effect of these cells on Tregs induced by anterior chamber-associated immune deviation (ACAID). Methods: CS cells were induced using a 2.0-mm trephine to score the corneal epithelium. CD11c+ CS cells were evaluated by adoptive transfer and by their capacity to disable CD8+ ACAID Tregs in local adoptive transfer (LAT) of suppression assays. CD11c+ cells were deleted from the ocular surface by subconjunctival injection of clodronate-containing liposomes. Results: CD11c+ CS cell were radiosenstive and long lived. As few as 1000 CS cells blocked the suppressive activity of previously generated CD8+ ACAID Tregs, indicating that CS cells act at the efferent arm of the immune response. Depletion of resident CD11c+ cells at the ocular surface prevented the generation of CS cells. Conclusions: Corneal nerve injury that occurs during keratoplasty converts ocular surface CD11c+ cells into CS cells that block CD8+ Tregs, which are induced by introducing antigens into the anterior chamber (i.e., ACAID Tregs). Depletion of CD11c+ cells at the ocular surface prevents the generation of CS cells and may be a useful strategy for preventing SLIP and enhancing the survival of second corneal transplants.


Asunto(s)
Cámara Anterior/inmunología , Antígeno CD11c/inmunología , Córnea/inervación , Privilegio Inmunológico/inmunología , Nervio Oftálmico/lesiones , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Supervivencia de Injerto , Hipersensibilidad Tardía/inmunología , Tolerancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
16.
Semin Oncol ; 45(3): 170-175, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30262396

RESUMEN

Despite their abilities to elicit immune responses, both syngeneic tumors and the half-mismatched placenta grow in the host, unlike a tissue allograft that is aggressively rejected. This is because of local and systemic factors that contribute to the immunologic privilege of tumors and the placenta. Checkpoint blockade immunotherapies subvert this privilege, with spectacularly beneficial outcomes in subsets of patients with certain types of cancer. A challenge for the community of scientists and clinicians is to replicate these successes in pregnant patients with cancer, without harm to the placenta. Here we compare and contrast the immunology of cancers and the placenta, and suggest that immunotherapy for pregnant patients with cancer may be a reasonable option, but that this should be explored systematically.


Asunto(s)
Privilegio Inmunológico/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Placenta/inmunología , Complicaciones Neoplásicas del Embarazo/terapia , Femenino , Antígenos HLA/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Placenta/metabolismo , Embarazo , Complicaciones Neoplásicas del Embarazo/inmunología , Complicaciones Neoplásicas del Embarazo/metabolismo , Linfocitos T/inmunología
17.
Brain Behav Immun ; 68: 169-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29061363

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is the most prevalent childhood mental disorders that often persists into adulthood. Moreover, methylphenidate (MPH) is the mainstay of medical treatment for this disorder. Yet, not much is known about the neurobiological impact of MPH on control versus ADHD conditions, which is crucial to simultaneously clarify the misuse/abuse versus therapeutic use of this psychostimulant. In the present study, we applied biochemical and behavioral approaches to broadly explore the early-life chronic exposure of two different doses of MPH (1.5 and 5 mg/kg/day) on control and ADHD rats (Wistar Kyoto and Spontaneously Hypertensive rats, respectively). We concluded that the higher dose of MPH promoted blood-brain barrier (BBB) permeability and elicited anxiety-like behavior in both control and ADHD animals. BBB dysfunction triggered by MPH was particularly prominent in control rats, which was characterized by a marked disruption of intercellular junctions, an increase of endothelial vesicles, and an upregulation of adhesion molecules concomitantly with the infiltration of peripheral immune cells into the prefrontal cortex. Moreover, both doses of MPH induced a robust neuroinflammatory and oxidative response in control rats. Curiously, in the ADHD model, the lower dose of MPH (1.5 mg/kg/day) had a beneficial effect since it balanced both immunity and behavior relative to vehicle animals. Overall, the contrasting effects of MPH observed between control and ADHD models support the importance of an appropriate MPH dose regimen for ADHD, and also suggest that MPH misuse negatively affects brain and behavior.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Privilegio Inmunológico/fisiología , Metilfenidato/farmacología , Animales , Ansiedad/metabolismo , Atención/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Privilegio Inmunológico/inmunología , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
18.
Z Rheumatol ; 76(8): 656-663, 2017 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-28710528

RESUMEN

The eye has all the mechanisms necessary for detection and processing (afferent immune reaction) as well as adequate initiation of an (efferent) immune response. Apart from the typical antigen-processing cells, locally present elements (e.g. glial cells and retinal pigment epithelium) can also be involved in the afferent reaction. For the efferent mechanisms a complex regulative system exists, which includes cellular and humoral responses and is essentially determined by surface molecules. In addition, the ocular environment is rich in immunosuppressive molecules that contribute to the regulation of immune cells. The adaptation of the anatomical and biochemical mechanisms for the creation of an immune-privileged microenvironment makes this sense organ unique. The purpose of this article is to highlight the specific features of the eye and to establish a reference to frequent ocular manifestations in rheumatic diseases.


Asunto(s)
Ojo/anatomía & histología , Ojo/inmunología , Inmunocompetencia/inmunología , Enfermedades Reumáticas/inmunología , Formación de Anticuerpos/inmunología , Presentación de Antígeno/inmunología , Enfermedades Autoinmunes/inmunología , Humanos , Privilegio Inmunológico/inmunología , Tolerancia Inmunológica/inmunología , Inmunidad Celular/inmunología , Queratoconjuntivitis Seca/inmunología , Neuroglía/inmunología , Epitelio Pigmentado de la Retina/inmunología
20.
Vestn Oftalmol ; 132(5): 117-124, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-28635736

RESUMEN

Transplantation of donated cornea is a radical and pathogenetically oriented measure of rehabilitation for patients with corneal blindness. Unlike other transplantation surgeries, keratoplasty is usually done without tissue typing and systemic immunosuppression. Even so, in the absence of risk factors, grafts can remain clear in as many as 90% of cases. The phenomenon is explained by immune privilege of the cornea. The hypothesis of immune privilege comprises several interrelated mechanisms that ensure long-lasting transparency of the graft. These include transfer limits of immunogenic stimuli to peripheral lymphoid tissues, deviation of the immune response, and neutralization of immune effector elements within the host-graft interface by immunosuppressive ocular environment. A change in any of the said components leads to loss of the immune-privilege status of the cornea, thus, significantly increasing the risk of graft rejection. We know several stages of the response activation cascade in graft rejection that can be influenced therapeutically, namely, antigen absorption, processing, and presentation as well as T-cell expansion and cellular inflow to the eye. The first two stages take place in the eye itself and are susceptible to instillation drugs and gene therapy. New highly selective medications aimed at specific signals from the immune cells and their pathways may be able to help restore the immune privilege of the cornea and improve the results of optical and reconstructive surgeries in high-risk patients.


Asunto(s)
Córnea/inmunología , Trasplante de Córnea/efectos adversos , Rechazo de Injerto , Privilegio Inmunológico/inmunología , Enfermedades de la Córnea/cirugía , Trasplante de Córnea/métodos , Rechazo de Injerto/inmunología , Rechazo de Injerto/fisiopatología , Humanos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA