Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cancer Immunol Immunother ; 72(1): 151-168, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35776159

RESUMEN

The complex immunosuppressive nature of solid tumor microenvironments poses a significant challenge to generating efficacious and durable anticancer responses. Photoimmunotherapy is a cancer treatment strategy by which an antibody is conjugated with a non-toxic light-activatable dye. Following administration of the conjugate and binding to the target tumor, subsequent local laser illumination activates the dye, resulting in highly specific target cell membrane disruption. Here we demonstrate that photoimmunotherapy treatment elicited tumor necrosis, thus inducing immunogenic cell death characterized by the release of damage-associated molecular patterns (DAMPs). Photoimmunotherapy-killed tumor cells activated dendritic cells (DC), leading to the production of proinflammatory cytokines, T cell stimulation, priming antigen-specific T cells, and durable memory T cell responses, which led complete responder mice to effectively reject new tumors upon rechallenge. PD-1 blockade in combination with photoimmunotherapy enhanced overall anticancer efficacy, including against anti-PD-1-resistant tumors. The combination treatment also elicited abscopal anticancer activity, as observed by reduction of distal, non-illuminated tumors, further demonstrating the ability of photoimmunotherapy to harness local and peripheral T cell responses. With this work we therefore delineate the immune mechanisms of action for photoimmunotherapy and demonstrate the potential for cancer-targeted photoimmunotherapy to be combined with other immunotherapy approaches for augmented, durable anticancer efficacy. Moreover, we demonstrate responses utilizing various immunocompetent mouse models, as well as in vitro data from human cells, suggesting broad translational potential.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Animales , Ratones , Inmunoterapia/métodos , Fototerapia/métodos , Neoplasias/terapia , Modelos Animales de Enfermedad , Línea Celular Tumoral , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 115(41): 10410-10415, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254166

RESUMEN

Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal ß-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking ß-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1ß and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved ß-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunidad Innata/inmunología , Mediadores de Inflamación/metabolismo , Células Asesinas Naturales/patología , Lectinas/metabolismo , Piroptosis , Antígenos de Diferenciación Mielomonocítica/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Lectinas/genética
3.
EMBO J ; 34(22): 2775-88, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26459514

RESUMEN

The intracellular chaperone heat-shock protein 70 (Hsp70) can be secreted from cells, but its extracellular role is unclear, as the protein has been reported to both activate and suppress the innate immune response. Potential immunomodulatory receptors on myelomonocytic lineage cells that bind extracellular Hsp70 are not well defined. Siglecs are Ig-superfamily lectins on mammalian leukocytes that recognize sialic acid-bearing glycans and thereby modulate immune responses. Siglec-5 and Siglec-14, expressed on monocytes and neutrophils, share identical ligand-binding domains but have opposing signaling functions. Based on phylogenetic analyses of these receptors, we predicted that endogenous sialic acid-independent ligands should exist. An unbiased screen revealed Hsp70 as a ligand for Siglec-5 and Siglec-14. Hsp70 stimulation through Siglec-5 delivers an anti-inflammatory signal, while stimulation through Siglec-14 is pro-inflammatory. The functional consequences of this interaction are also addressed in relation to a SIGLEC14 polymorphism found in humans. Our results demonstrate that an endogenous non-sialic acid-bearing molecule can be either a danger-associated or self-associated signal through paired Siglecs, and may explain seemingly contradictory prior reports on extracellular Hsp70 action.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Lectinas/inmunología , Monocitos/inmunología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Línea Celular Tumoral , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lectinas/genética , Monocitos/patología , Receptores de Superficie Celular/genética , Transducción de Señal/genética
4.
J Biol Chem ; 287(15): 12195-203, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22371497

RESUMEN

O-Linked ß-N-acetylglucosamine, or O-GlcNAc, is a dynamic post-translational modification that cycles on and off serine and threonine residues of nucleocytoplasmic proteins. The O-GlcNAc modification shares a complex relationship with phosphorylation, as both modifications are capable of mutually inhibiting the occupation of each other on the same or nearby amino acid residue. In addition to diabetes, cancer, and neurodegenerative diseases, O-GlcNAc appears to play a significant role in cell growth and cell cycle progression, although the precise mechanisms are still not well understood. A recent study also found that all four core nucleosomal histones (H2A, H2B, H3, and H4) are modified with O-GlcNAc, although no specific sites on H3 were reported. Here, we describe that histone H3, a protein highly phosphorylated during mitosis, is modified with O-GlcNAc. Several biochemical assays were used to validate that H3 is modified with O-GlcNAc. Mass spectrometry analysis identified threonine 32 as a novel O-GlcNAc site. O-GlcNAc was detected at higher levels on H3 during interphase than mitosis, which inversely correlated with phosphorylation. Furthermore, increased O-GlcNAcylation was observed to reduce mitosis-specific phosphorylation at serine 10, serine 28, and threonine 32. Finally, inhibiting OGA, the enzyme responsible for removing O-GlcNAc, hindered the transition from G2 to M phase of the cell cycle, displaying a phenotype similar to preventing mitosis-specific phosphorylation on H3. Taken together, these data indicate that O-GlcNAcylation regulates mitosis-specific phosphorylations on H3, providing a mechanistic switch that orchestrates the G2-M transition of the cell cycle.


Asunto(s)
Acetilglucosamina/fisiología , Histonas/metabolismo , Mitosis , Procesamiento Proteico-Postraduccional , Acetilglucosamina/metabolismo , Secuencia de Aminoácidos , Fase G2 , Glicosilación , Células HeLa , Histonas/química , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fosforilación , Treonina/metabolismo
6.
J Innate Immun ; 12(4): 333-343, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31805552

RESUMEN

Pathogenic microorganisms are sensed by the inflammasome, resulting in the release of the pro-immune and proinflammatory cytokine interleukin-1ß (IL-1ß). In humans, the paired sialic acid-binding Ig-like lectin receptors Siglec-5 (inhibitory) and Siglec-14 (activating) have been shown to have reciprocal roles in regulating macrophage immune responses, but their interaction with IL-1ß signaling and the inflammasome has not been characterized. Here we show that in response to known inflammasome activators (ATP, nigericin) or the sialic acid-expressing human bacterial pathogen group B Streptococcus (GBS), the presence of Siglec-14 enhances, whereas Siglec-5 reduces, inflammasome activation and macrophage IL-1ß release. Human THP-1 macrophages stably transfected with Siglec-14 exhibited increased caspase-1 activation, IL-1ß release and pyroptosis after GBS infection, in a manner blocked by a specific inhibitor of nucleotide-binding domain leucine-rich repeat protein 3 (NLRP3), a protein involved in inflammasome assembly. Another leading pathogen, Streptococcus pneumoniae, lacks sialic acid but rather prominently expresses a sialidase, which cleaves sialic acid from macrophages, eliminating cis- interactions with the lectin receptor, thus attenuating Siglec-14 induced IL-1ß secretion. Vimentin, a cytoskeletal protein released during macrophage inflammatory activation is known to induce the inflammasome. We found that vimentin has increased interaction with Siglec-14 compared to Siglec-5, and this interaction heightened IL-1ß production by Siglec-14-expressing cells. Siglec-14 is absent from some humans because of a SIGLEC5/14 fusion polymorphism, and we found increased IL-1ß expression in primary macrophages from SIGLEC14+/+ individuals compared to those with the SIGLEC14-/+ and SIGLEC14-/- genotypes. Collectively, our results identify a new immunoregulatory role of Siglec-14 as a positive regulator of NLRP3 inflammasome activation.


Asunto(s)
Inflamasomas/inmunología , Lectinas/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Receptores de Superficie Celular/inmunología , Humanos , Inflamasomas/genética , Lectinas/genética , Macrófagos/microbiología , Macrófagos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores de Superficie Celular/genética , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/patología , Streptococcus agalactiae/inmunología , Células THP-1
7.
Evol Appl ; 12(2): 337-349, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30697344

RESUMEN

Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhea exclusively in humans and uses multiple strategies to infect, including acquisition of host sialic acids that cap and mask lipooligosaccharide termini, while restricting complement activation. We hypothesized that gonococci selectively target human anti-inflammatory sialic acid-recognizing Siglec receptors on innate immune cells to blunt host responses and that pro-inflammatory Siglecs and SIGLEC pseudogene polymorphisms represent host evolutionary adaptations to counteract this interaction. N. gonorrhoeae can indeed engage multiple human but not chimpanzee CD33rSiglecs expressed on innate immune cells and in the genitourinary tract--including Siglec-11 (inhibitory) and Siglec-16 (activating), which we detected for the first time on human cervical epithelium. Surprisingly, in addition to LOS sialic acid, we found that gonococcal porin (PorB) mediated binding to multiple Siglecs. PorB also bound preferentially to human Siglecs and not chimpanzee orthologs, modulating host immune reactions in a human-specific manner. Lastly, we studied the distribution of null SIGLEC polymorphisms in a Namibian cohort with a high prevalence of gonorrhea and found that uninfected women preferentially harbor functional SIGLEC16 alleles encoding an activating immune receptor. These results contribute to the understanding of the human specificity of N. gonorrhoeae and how it evolved to evade the human immune defense.

8.
Tuberculosis (Edinb) ; 104: 38-45, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28454648

RESUMEN

Humans exposed to Mycobacterium tuberculosis (Mtb) have variable susceptibility to tuberculosis (TB) and its outcomes. Siglec-5 and Siglec-14 are members of the sialic-acid binding lectin family that regulate immune responses to pathogens through inhibitory (Siglec-5) and activating (Siglec-14) domains. The SIGLEC14 coding sequence is deleted in a high proportion of individuals, placing a SIGLEC5-like gene under the expression of the SIGLEC14 promoter (the SIGLEC14 null allele) and causing expression of a Siglec-5 like protein in monocytes and macrophages. We hypothesized that the SIGLEC14 null allele was associated with Mtb replication in monocytes, T-cell responses to the BCG vaccine, and clinical susceptibility to TB. The SIGLEC14 null allele was associated with protection from TB meningitis in Vietnamese adults but not with pediatric TB in South Africa. The null allele was associated with increased IL-2 and IL-17 production following ex-vivo BCG stimulation of blood from 10 week-old South African infants vaccinated with BCG at birth. Mtb replication was increased in THP-1 cells overexpressing either Siglec-5 or Siglec-14 relative to controls. To our knowledge, this is the first study to demonstrate an association between SIGLEC expression and clinical TB, Mtb replication, or BCG-specific T-cell cytokines.


Asunto(s)
Vacuna BCG/administración & dosificación , Lectinas/genética , Mycobacterium tuberculosis/inmunología , Receptores de Superficie Celular/genética , Tuberculosis Meníngea/genética , Tuberculosis Meníngea/prevención & control , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/prevención & control , Vacunación , Inmunidad Adaptativa , Adolescente , Adulto , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/inmunología , Vacuna BCG/inmunología , Estudios de Casos y Controles , Preescolar , Citocinas/inmunología , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno , Humanos , Lactante , Recién Nacido , Lectinas/inmunología , Masculino , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium tuberculosis/crecimiento & desarrollo , Fenotipo , Estudios Prospectivos , Receptores de Superficie Celular/inmunología , Sudáfrica , Linfocitos T/inmunología , Linfocitos T/microbiología , Células THP-1 , Factores de Tiempo , Resultado del Tratamiento , Tuberculosis Meníngea/inmunología , Tuberculosis Meníngea/microbiología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Vietnam
9.
J Exp Med ; 211(6): 1231-42, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24799499

RESUMEN

Group B Streptococcus (GBS) causes invasive infections in human newborns. We recently showed that the GBS ß-protein attenuates innate immune responses by binding to sialic acid-binding immunoglobulin-like lectin 5 (Siglec-5), an inhibitory receptor on phagocytes. Interestingly, neutrophils and monocytes also express Siglec-14, which has a ligand-binding domain almost identical to Siglec-5 but signals via an activating motif, raising the possibility that these are paired Siglec receptors that balance immune responses to pathogens. Here we show that ß-protein-expressing GBS binds to both Siglec-5 and Siglec-14 on neutrophils and that the latter engagement counteracts pathogen-induced host immune suppression by activating p38 mitogen-activated protein kinase (MAPK) and AKT signaling pathways. Siglec-14 is absent from some humans because of a SIGLEC14-null polymorphism, and homozygous SIGLEC14-null neutrophils are more susceptible to GBS immune subversion. Finally, we report an unexpected human-specific expression of Siglec-5 and Siglec-14 on amniotic epithelium, the site of initial contact of invading GBS with the fetus. GBS amnion immune activation was likewise influenced by the SIGLEC14-null polymorphism. We provide initial evidence that the polymorphism could influence the risk of prematurity among human fetuses of mothers colonized with GBS. This first functionally proven example of a paired receptor system in the Siglec family has multiple implications for regulation of host immunity.


Asunto(s)
Amnios/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Lectinas/inmunología , Neutrófilos/inmunología , Receptores de Superficie Celular/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Adulto , Amnios/metabolismo , Amnios/microbiología , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Genotipo , Interacciones Huésped-Patógeno/inmunología , Humanos , Recién Nacido , Lectinas/genética , Lectinas/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Fosforilación , Polimorfismo Genético , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/genética , Streptococcus agalactiae/fisiología
10.
Am J Physiol Gastrointest Liver Physiol ; 292(1): G215-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16920699

RESUMEN

Following massive small bowel resection (SBR) in mice, there are sustained increases in crypt depth and villus height, resulting in enhanced mucosal surface area. The early mechanisms responsible for resetting and sustaining this increase are presently not understood. We hypothesized that expansion of secretory lineages is an early and sustained component of the adaptive response. This was assessed in the ileum by quantitative morphometry at 12 h, 36 h, 7 days, and 28 days and by quantitative RT-PCR of marker mRNAs for proliferation and differentiated goblet, Paneth cell, and enterocyte genes at 12 h after 50% SBR or sham operation. As predicted, SBR elicited increases of both crypt and villus epithelial cells, which were sustained though the 28 days of the experiment. Significant increases in the overall number and percentage of both Paneth and goblet cells within intestinal epithelium occurred by 12 h and were sustained up to 28 days after SBR. The increases of goblet cells after SBR were initially observed within villi at 12 h, with marked increases occurring in crypts at 36 h and 7 days. Consistent with this finding, qRT-PCR demonstrated significant increases in the expression of mRNAs associated with proliferation (c-myc) and differentiated goblet cells (Tff3, Muc2) and Paneth cells (lysozyme), whereas mRNA associated with differentiated enterocytes (sucrase-isomaltase) remained unchanged. From these data, we speculate that early expansion of intestinal secretory lineages within the epithelium of the ileum occurs following SBR, possibly serving to amplify the signal responsible for initiating and sustaining intestinal adaptation.


Asunto(s)
Mucosa Intestinal/cirugía , Intestino Delgado/cirugía , Anastomosis Quirúrgica , Animales , Cartilla de ADN , Íleon/anatomía & histología , Íleon/fisiología , Íleon/cirugía , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Intestino Delgado/metabolismo , Intestino Delgado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvellosidades/fisiología , Microvellosidades/ultraestructura , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Complejo Sacarasa-Isomaltasa/genética
11.
Am J Physiol Gastrointest Liver Physiol ; 293(5): G1013-22, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17855764

RESUMEN

Sustained increases in mucosal surface area occur in remaining bowel following massive intestinal loss. The mechanisms responsible for expanding and perpetuating this response are not presently understood. We hypothesized that an increase in the number of intestinal stem cells (ISC) occurs following intestinal resection and is an important component of the adaptive response in mice. This was assessed in the jejunum of mice 2-3 days, 4-5 days, 6-7 days, 2 wk, 6 wk, and 16 wk following ileocecal resection (ICR) or sham operation. Changes in ISC following ICR compared with sham resulted in increased crypt fission and were assayed by 1) putative ISC population (SP) by flow cytometry, 2) Musashi-1 immunohistochemistry, and 3) bromodeoxyuridine (BrdU) label retention. Observed early increases in crypt depth and villus height were not sustained 16 wk following operation. In contrast, long-term increases in intestinal caliber and overall number of crypts per circumference appear to account for the enhanced mucosal surface area following ICR. Flow cytometry demonstrated that significant increases in SP cells occur within 2-3 days following resection. By 7 days, ICR resulted in marked increases in crypt fission and Musashi-1 immunohistochemistry staining. Separate label-retention studies confirmed a 20-fold increase in BrdU incorporation 6 wk following ICR, confirming an overall increase in the number of ISC. These studies support that expansion of ISC occurs following ICR, leading to an overall increase number of crypts through a process of fission and intestinal dilation. Understanding the mechanism expanding ISCs may provide important insight into management of intestinal failure.


Asunto(s)
Ciego/cirugía , Íleon/cirugía , Mucosa Intestinal/fisiología , Células Madre/citología , Células Madre/fisiología , Aclimatación , Animales , División Celular , Mucosa Intestinal/citología , Yeyuno/citología , Yeyuno/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA