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.
Cell Stem Cell ; 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38663406

RESUMEN

Due to the limitations of autologous chimeric antigen receptor (CAR)-T cells, alternative sources of cellular immunotherapy, including CAR macrophages, are emerging for solid tumors. Human induced pluripotent stem cells (iPSCs) offer an unlimited source for immune cell generation. Here, we develop human iPSC-derived CAR macrophages targeting prostate stem cell antigen (PSCA) (CAR-iMacs), which express membrane-bound interleukin (IL)-15 and truncated epidermal growth factor receptor (EGFR) for immune cell activation and a suicide switch, respectively. These allogeneic CAR-iMacs exhibit strong antitumor activity against human pancreatic solid tumors in vitro and in vivo, leading to reduced tumor burden and improved survival in a pancreatic cancer mouse model. CAR-iMacs appear safe and do not exhibit signs of cytokine release syndrome or other in vivo toxicities. We optimized the cryopreservation of CAR-iMac progenitors that remain functional upon thawing, providing an off-the-shelf, allogeneic cell product that can be developed into CAR-iMacs. Overall, our preclinical data strongly support the potential clinical translation of this human iPSC-derived platform for solid tumors, including pancreatic cancer.

2.
Chin Med J (Engl) ; 137(5): 533-546, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38321811

RESUMEN

ABSTRACT: Lung cancer remains the most common cause of cancer death. Given the continued research into new drugs and combination therapies, outcomes in lung cancer have been improved, and clinical benefits have been expanded to a broader patient population. However, the overall cure and survival rates for lung cancer patients remain low, especially in metastatic cases. Among the available lung cancer treatment options, such as surgery, radiation therapy, chemotherapy, targeted therapies, and alternative therapies, immunotherapy has shown to be the most promising. The exponential progress in immuno-oncology research and recent advancements made in the field of immunotherapy will further increase the survival and quality of life for lung cancer patients. Substantial progress has been made in targeted therapies using tyrosine kinase inhibitors and monoclonal antibody immune checkpoint inhibitors with many US Food And Drug Administration (FDA)-approved drugs targeting the programmed cell death ligand-1 protein (e.g., durvalumab, atezolizumab), the programmed cell death-1 receptor (e.g., nivolumab, pembrolizumab), and cytotoxic T-lymphocyte-associated antigen 4 (e.g., tremelimumab, ipilimumab). Cytokines, cancer vaccines, adoptive T cell therapies, and Natural killer cell mono- and combinational therapies are rapidly being studied, yet to date, there are currently none that are FDA-approved for the treatment of lung cancer. In this review, we discuss the current lung cancer therapies with an emphasis on immunotherapy, including the challenges for future research and clinical applications.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/terapia , Calidad de Vida , Nivolumab , Inmunoterapia , Terapia Combinada , Antígeno B7-H1/metabolismo
3.
Cancer Treat Res ; 190: 49-94, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38112999

RESUMEN

Cancer immunotherapy, which modulates immune responses against tumors using immune-checkpoint inhibitors or adoptive cell transfer, has emerged as a novel and promising therapy for tumors. However, only a minority of patients demonstrate durable responses, while the majority of patients are resistant to immunotherapy. The immune system can paradoxically constrain and promote tumor development and progression. This process is referred to as cancer immunoediting. The mechanisms of resistance to immunotherapy seem to be that cancer cells undergo immunoediting to evade recognition and elimination by the immune system. RNA modifications, specifically N6-methyladenosine (m6A) methylation, have emerged as a key regulator of various post-transcriptional gene regulatory processes, such as RNA export, splicing, stability, and degradation, which play unappreciated roles in various physiological and pathological processes, including immune system development and cancer pathogenesis. Therefore, a deeper understanding of the mechanisms by which RNA modifications impact the cancer immunoediting process can provide insight into the mechanisms of resistance to immunotherapies and the strategies that can be used to overcome such resistance. In this chapter, we briefly introduce the background of cancer immunoediting and immunotherapy. We also review and discuss the roles and mechanisms of RNA m6A modifications in fine-tuning the innate and adaptive immune responses, as well as in regulating tumor escape from immunosurveillance. Finally, we summarize the current strategies targeting m6A regulators for cancer immunotherapy.


Asunto(s)
Neoplasias , ARN , Humanos , Neoplasias/genética , Neoplasias/terapia , Inmunoterapia , Inmunoterapia Adoptiva
4.
Sci Immunol ; 8(81): eabn7993, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36897958

RESUMEN

Spliced X-box-binding protein 1 (XBP1s) is an essential transcription factor downstream of interleukin-15 (IL-15) and AKT signaling, which controls cell survival and effector functions of human natural killer (NK) cells. However, the precise mechanisms, especially the downstream targets of XBP1s, remain unknown. In this study, by using XBP1 conditional knockout mice, we found that XBP1s is critical for IL-15-mediated NK cell survival but not proliferation in vitro and in vivo. Mechanistically, XBP1s regulates homeostatic NK cell survival by targeting PIM-2, a critical anti-apoptotic gene, which in turn stabilizes XBP1s protein by phosphorylating it at Thr58. In addition, XBP1s enhances the effector functions and antitumor immunity of NK cells by recruiting T-bet to the promoter region of Ifng. Collectively, our findings identify a previously unknown mechanism by which IL-15-XBP1s signaling regulates the survival and effector functions of NK cells.


Asunto(s)
Interleucina-15 , Proteínas Serina-Treonina Quinasas , Proteína 1 de Unión a la X-Box , Animales , Humanos , Ratones , Proteínas de Unión al ADN/genética , Retroalimentación , Células Asesinas Naturales/metabolismo , Ratones Noqueados , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
5.
Nat Immunol ; 24(2): 255-266, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36658237

RESUMEN

Despite tumor-associated macrophages (TAMs) playing a key role in shaping the tumor microenvironment (TME), the mechanisms by which TAMs influence the TME and contribute to cancer progression remain unclear. Here, we show that the N6-methyladenosine reader YTHDF2 regulates the antitumor functions of TAMs. YTHDF2 deficiency in TAMs suppressed tumor growth by reprogramming TAMs toward an antitumoral phenotype and increasing their antigen cross-presentation ability, which in turn enhanced CD8+ T cell-mediated antitumor immunity. YTHDF2 deficiency facilitated the reprogramming of TAMs by targeting interferon-γ-STAT1 signaling. The expression of YTHDF2 in TAMs was regulated by interleukin-10-STAT3 signaling. Selectively targeting YTHDF2 in TAMs using a Toll-like receptor 9 agonist-conjugated small interfering RNA reprogrammed TAMs toward an antitumoral phenotype, restrained tumor growth and enhanced the efficacy of PD-L1 antibody therapy. Collectively, our findings describe the role of YTHDF2 in orchestrating TAMs and suggest that YTHDF2 inhibition is an effective approach to enhance cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Macrófagos , Macrófagos Asociados a Tumores , Neoplasias/metabolismo , Inmunoterapia , Microambiente Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Nat Commun ; 13(1): 234, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017515

RESUMEN

Environmental enteric dysfunction is associated with malnutrition as well as infant growth stunting and has been classically defined by villous blunting, decreased crypt-to-villus ratio, and inflammation in the small intestine. Here, we characterized environmental enteric dysfunction among infant rhesus macaques that are naturally exposed to enteric pathogens commonly linked to human growth stunting. Remarkably, despite villous atrophy and histological abnormalities observed in the small intestine, poor growth trajectories and low serum tryptophan levels were correlated with increased histopathology in the large intestine. This work provides insight into the mechanisms underlying this disease and indicates that the large intestine may be an important target for therapeutic intervention.


Asunto(s)
Intestino Grueso/patología , Intestino Delgado/patología , Macaca mulatta/crecimiento & desarrollo , Animales , Duodeno/patología , Femenino , Tracto Gastrointestinal , Expresión Génica , Trastornos del Crecimiento/patología , Humanos , Íleon/patología , Inflamación , Enfermedades Intestinales , Mucosa Intestinal , Yeyuno/patología , Masculino , Desnutrición
7.
J Virol ; 96(1): e0096421, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34668775

RESUMEN

A comprehensive analysis and characterization of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection model that mimics non-severe and severe coronavirus disease 2019 (COVID-19) in humans is warranted for understating the virus and developing preventive and therapeutic agents. Here, we characterized the K18-hACE2 mouse model expressing human (h)ACE2 in mice, controlled by the human keratin 18 (K18) promoter, in the epithelia, including airway epithelial cells where SARS-CoV-2 infections typically start. We found that intranasal inoculation with higher viral doses (2 × 103 and 2 × 104 PFU) of SARS-CoV-2 caused lethality of all mice and severe damage of various organs, including lung, liver, and kidney, while lower doses (2 × 101 and 2 × 102 PFU) led to less severe tissue damage and some mice recovered from the infection. In this hACE2 mouse model, SARS-CoV-2 infection damaged multiple tissues, with a dose-dependent effect in most tissues. Similar damage was observed in postmortem samples from COVID-19 patients. Finally, the mice that recovered from infection with a low dose of virus survived rechallenge with a high dose of virus. Compared to other existing models, the K18-hACE2 model seems to be the most sensitive COVID-19 model reported to date. Our work expands the information available about this model to include analysis of multiple infectious doses and various tissues with comparison to human postmortem samples from COVID-19 patients. In conclusion, the K18-hACE2 mouse model recapitulates both severe and non-severe COVID-19 in humans being dose-dependent and can provide insight into disease progression and the efficacy of therapeutics for preventing or treating COVID-19. IMPORTANCE The pandemic of coronavirus disease 2019 (COVID-19) has reached nearly 240 million cases, caused nearly 5 million deaths worldwide as of October 2021, and has raised an urgent need for the development of novel drugs and therapeutics to prevent the spread and pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To achieve this goal, an animal model that recapitulates the features of human COVID-19 disease progress and pathogenesis is greatly needed. In this study, we have comprehensively characterized a mouse model of SARS-CoV-2 infection using K18-hACE2 transgenic mice. We infected the mice with low and high doses of SARS-CoV-2 to study the pathogenesis and survival in response to different infection patterns. Moreover, we compared the pathogenesis of the K18-hACE2 transgenic mice with that of the COVID-19 patients to show that this model could be a useful tool for the development of antiviral drugs and therapeutics.


Asunto(s)
COVID-19/patología , Modelos Animales de Enfermedad , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/inmunología , COVID-19/mortalidad , COVID-19/virología , Humanos , Sueros Inmunes/inmunología , Queratina-18/genética , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Reinfección/inmunología , Reinfección/mortalidad , Reinfección/patología , Reinfección/virología , SARS-CoV-2/inmunología , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Cancer Immunol Res ; 9(10): 1229-1241, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34326137

RESUMEN

Trogocytosis is a fast, cell-cell contact-dependent uptake of membrane patches and associated molecules by one cell from another. Here, we report our investigation of trogocytosis of TYRO3, a cell membrane protein, from tumor target cells to natural killer (NK) cells and the associated functional consequences for NK cells. We found that although NK cells did not express endogenous TYRO3 on the cell surface, activated NK cells rapidly acquired TYRO3 from tumor cells via trogocytosis in vitro and in vivo. NK cells that acquired TYRO3, which we termed TYRO3+ NK cells, had significantly enhanced cytotoxicity and IFNγ production as well as higher expression of some activated surface markers compared with TYRO3- NK cells. Furthermore, the activation status of NK cells and TYRO3 expression levels on donor cells, either endogenous or ectopic, positively correlated with trogocytosis levels. When the antigen-presenting cell (APC) K562 leukemia cell line, a feeder cell line to expand NK cells, overexpressed TYRO3, TYRO3 was transferred to NK cells via trogocytosis, which improved NK-cell proliferation ex vivo. This provides a strategy to manufacture NK cells or their engineered counterparts, such as chimeric antigen receptor NK cells, for the treatment of cancer or infectious diseases.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Trogocitosis , Animales , Proliferación Celular , Humanos , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Ratones , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Quiméricos de Antígenos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Exp Med ; 218(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34160549

RESUMEN

N 6-methyladenosine (m6A) is the most prevalent posttranscriptional modification on RNA. NK cells are the predominant innate lymphoid cells that mediate antiviral and antitumor immunity. However, whether and how m6A modifications affect NK cell immunity remain unknown. Here, we discover that YTHDF2, a well-known m6A reader, is upregulated in NK cells upon activation by cytokines, tumors, and cytomegalovirus infection. Ythdf2 deficiency in NK cells impairs NK cell antitumor and antiviral activity in vivo. YTHDF2 maintains NK cell homeostasis and terminal maturation, correlating with modulating NK cell trafficking and regulating Eomes, respectively. YTHDF2 promotes NK cell effector function and is required for IL-15-mediated NK cell survival and proliferation by forming a STAT5-YTHDF2 positive feedback loop. Transcriptome-wide screening identifies Tardbp to be involved in cell proliferation or survival as a YTHDF2-binding target in NK cells. Collectively, we elucidate the biological roles of m6A modifications in NK cells and highlight a new direction to harness NK cell antitumor immunity.


Asunto(s)
Adenosina/análogos & derivados , Antivirales/inmunología , Inmunidad , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , ARN/metabolismo , Adenosina/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Homeostasis , Interleucina-15/metabolismo , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/metabolismo , Transcriptoma/genética
10.
Cancer Res ; 81(13): 3635-3648, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34006525

RESUMEN

IL15 is a pleiotropic cytokine with multiple roles that improve immune responses to tumor cells. Oncolytic viruses (OV) specifically lyse tumors and activate immune responses. Systemic administration of IL15 or its complex with the IL15Rα and chimeric antigen receptor (CAR) natural killer (NK) cells are currently being tested in the clinic. Here, we generated a herpes simplex 1-based OV-expressing human IL15/IL15Rα sushi domain fusion protein (named OV-IL15C), as well as off-the-shelf EGFR-CAR NK cells, and studied their monotherapy and combination efficacy in vitro and in multiple glioblastoma (GBM) mouse models. In vitro, soluble IL15/IL15Rα complex was secreted from OV-IL15C-infected GBM cells, which promoted GBM cytotoxicity and improved survival of NK and CD8+ T cells. Frozen, readily available off-the-shelf EGFR-CAR NK cells showed enhanced killing of tumor cells compared with empty vector-transduced NK cells. In vivo, OV-IL15C significantly inhibited tumor growth and prolonged survival of GBM-bearing mice in the presence of CD8+ T cells compared with parental OV. OV-IL15C plus EGFR-CAR NK cells synergistically suppressed tumor growth and significantly improved survival compared with either monotherapy, correlating with increased intracranial infiltration and activation of NK and CD8+ T cells and elevated persistence of CAR NK cells in an immunocompetent model. Collectively, OV-IL15C and off-the-shelf EGFR-CAR NK cells represent promising therapeutic strategies for GBM treatment to improve the clinical management of this devastating disease. SIGNIFICANCE: The combination of an oncolytic virus expressing the IL15/IL15Rα complex and frozen, ready-to-use EGFR-CAR NK cells elicits strong antitumor responses in glioblastoma.


Asunto(s)
Glioblastoma/terapia , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Virus Oncolíticos/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , Apoptosis , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Terapia Combinada , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Viroterapia Oncolítica/métodos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Immunol ; 206(4): 677-685, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33419766

RESUMEN

The E3 ubiquitin ligase Cbl-b has been characterized as an intracellular checkpoint in T cells; however, the function of Cbl-b in primary human NK cells, an innate immune anti-tumor effector cell, is not well defined. In this study, we show that the expression of Cbl-b is significantly upregulated in primary human NK cells activated by IL-15, IL-2, and the human NK cell-sensitive tumor cell line K562 that lacks MHC class I expression. Pretreatment with JAK or AKT inhibitors prior to IL-15 stimulation reversed Cbl-b upregulation. Downregulation of Cbl-b resulted in significant increases in granzyme B and perforin expression, IFN-γ production, and cytotoxic activity against tumor cells. Collectively, we demonstrate upregulation of Cbl-b and its inhibitory effects in IL-15/IL-2/K562-activated human NK cells, suggesting that Cbl-b plays a negative feedback role in human NK cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Punto de Control Inmunitario/metabolismo , Células Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Citotoxicidad Inmunológica , Granzimas/genética , Granzimas/metabolismo , Humanos , Proteínas de Punto de Control Inmunitario/genética , Inmunidad Innata , Interferón gamma/metabolismo , Interleucina-15/metabolismo , Interleucina-2/metabolismo , Células K562 , Activación de Linfocitos , Perforina/genética , Perforina/metabolismo , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal , Regulación hacia Arriba
12.
Genome Biol ; 20(1): 173, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31451108

RESUMEN

BACKGROUND: Diarrhea is the second leading cause of death in children under 5 years of age. Enhanced understanding of causal pathways, pathogenesis, and sequelae of diarrhea is urgently needed. Although the gut microbiota is believed to play a role in susceptibility to diarrheal diseases, our understanding of this association remains incomplete. Infant rhesus macaques (Macaca mulatta) are susceptible to diarrhea making them an ideal model to address this question. RESULTS: The maturation of the infant rhesus macaque gut microbiome throughout the first 8 months of life occurs in a similar pattern as that described for human infants. Moreover, the microbiome of the captive reared infant rhesus macaque more closely resembles that of human infants in the developing world than in the western world. Importantly, prior to disease onset, the gut microbiome of infants that later develop diarrhea is enriched in pathways of immunomodulatory metabolite synthesis, while those of infants that remain asymptomatic are enriched in pathways for short-chain fatty acid production. We identify Prevotella strains that are more abundant at 1 month in infants that later develop diarrhea. At 8 months, the microbiomes of animals that experience diarrhea show increased abundance of Campylobacter and a reduction in Helicobacter macacae. CONCLUSION: The composition of the microbial community could provide a phenotypic marker of an infant's susceptibility to diarrheal disease. Given the significant physiological and immunological similarities between human and nonhuman primates, these findings provide potential markers of susceptibility to diarrhea that could be modulated to improve infant health, especially in the developing world.


Asunto(s)
Diarrea/microbiología , Microbioma Gastrointestinal , Envejecimiento , Animales , Animales Recién Nacidos , Antibacterianos/uso terapéutico , Bacterias/genética , Biomarcadores/metabolismo , Portador Sano/microbiología , Niño , Preescolar , Países Desarrollados , Países en Desarrollo , Diarrea/tratamiento farmacológico , Susceptibilidad a Enfermedades , Femenino , Genoma Bacteriano , Humanos , Lactante , Macaca mulatta , Masculino , Metagenómica , Filogenia
13.
FASEB J ; 33(6): 7767-7777, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30897342

RESUMEN

Chronic heavy alcohol consumption, also referred to as chronic heavy drinking (CHD), results in intestinal injury characterized by increased permeability, dysbiosis, nutrient malabsorption, potentially higher susceptibility to infection, and increased risk of colorectal cancer. However, our understanding of the mechanisms by which CHD results in intestinal damage remains incomplete. Here, we investigated the impact of chronic drinking on transcriptional and functional responses of lamina propria leukocytes (LPLs) isolated from the 4 major gut sections. Although no significant differences were detected between LPLs isolated from the ethanol and control groups at resting state within each major gut section, our analysis uncovered key regional differences in composition and function of LPLs independent of alcohol consumption. However, in response to phorbol myristate acetate and ionomycin, duodenal LPLs from ethanol-drinking animals generated a dampened response, whereas jejunal and ileal LPLs from ethanol-drinking animals produced a heightened response. Transcriptional responses following stimulation were pronounced in ileal and duodenal LPLs from the ethanol-drinking group but less evident in jejunal and colonic LPLs compared with controls, suggesting a more significant impact of alcohol on these gut regions. The altered intestinal LPL function detected in our study reveals remarkable region specificity and novel insight into potential mechanisms of intestinal injury associated with CHD.-Barr, T., Lewis, S. A., Sureshchandra, S., Doratt, B., Grant, K. A., Messaoudi, I. Chronic ethanol consumption alters lamina propria leukocyte response to stimulation in a region-dependent manner.


Asunto(s)
Alcoholismo/patología , Etanol/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Leucocitos/efectos de los fármacos , Animales , Etanol/farmacología , Perfilación de la Expresión Génica , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Macaca mulatta , Masculino , Transcripción Genética
14.
Front Physiol ; 9: 1089, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30131724

RESUMEN

Pre-pregnancy (pregravid) obesity has been linked to several adverse health outcomes for both mother and offspring. Complications during pregnancy include increased risk for gestational diabetes, hypertension, preeclampsia, placental abruption, and difficulties during delivery. Several studies suggest that these negative outcomes are mediated by heightened systemic inflammation as well as changes in placental development and function. However, the molecular mechanisms by which pregravid obesity affects these processes are poorly understood. In this study, we aimed to address this question by carrying out a comprehensive analysis of the systemic maternal immune system coupled with placental gene expression and microbial profiling at term delivery (11 lean and 14 obese). Specifically, we examined the impact of pregravid obesity on circulating cytokines, chemokine, adipokines, and growth factors using multiplex Luminex assay. Innate and adaptive immune cell frequencies and their cytokine production in response to stimuli were measured using flow cytometry. Finally, changes in placental transcriptome and microbiome were profiled using RNA- and 16S-sequencing, respectively. Pregravid obesity is characterized by insulin and leptin resistance, high levels of circulating inflammatory markers IL-6 and CRP, in addition to chemokine IL-8 (p < 0.01). Moreover, pregravid obesity was associated with lower frequency of naïve CD4+ T-cells (p < 0.05), increased frequency of memory CD4+ T-cells (p < 0.01), and a shift towards Th2 cytokine production (p = 0.05). Myeloid cells from the obese cohort produced higher levels of pro-inflammatory cytokines but lower levels of chemokines following TLR stimulation (p < 0.05). Lastly, pregravid obesity is associated with increased abundance of Bacteroides and changes in the expression of genes important for nutrient transport and immunity (FDR < 0.05). Collectively, these data indicate that pregravid obesity is associated with heightened systemic inflammation and of dysregulated nutrient transport in the placenta and provide insight into the basis of fetal reprogramming.

15.
Gut Microbes ; 9(4): 338-356, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29517944

RESUMEN

Alcohol use disorder (AUD) results in increased intestinal permeability, nutrient malabsorption, and increased risk of colorectal cancer (CRC). Our understanding of the mechanisms underlying these morbidities remains limited because studies to date have relied almost exclusively on short-term heavy/binge drinking rodent models and colonic biopsies/fecal samples collected from AUD subjects with alcoholic liver disease (ALD). Consequently, the dose- and site-dependent impact of chronic alcohol consumption in the absence of overt liver disease remains poorly understood. In this study, we addressed this knowledge gap using a nonhuman primate model of voluntary ethanol self-administration where rhesus macaques consume varying amounts of 4% ethanol in water for 12 months. Specifically, we performed RNA-Seq and 16S rRNA gene sequencing on duodenum, jejunum, ileum, and colon biopsies collected from 4 controls and 8 ethanol-consuming male macaques. Our analysis revealed that chronic ethanol consumption leads to changes in the expression of genes involved in protein trafficking, metabolism, inflammation, and CRC development. Additionally, we observed differences in the relative abundance of putatively beneficial bacteria as well as those associated with inflammation and CRC. Given that the animals studied in this manuscript did not exhibit signs of ALD or CRC, our data suggest that alterations in gene expression and bacterial communities precede clinical disease and could serve as biomarkers as well as facilitate future studies aimed at developing interventions to restore gut homeostasis.


Asunto(s)
Alcoholismo/genética , Alcoholismo/microbiología , Bacterias/genética , Microbioma Gastrointestinal , Alcoholismo/metabolismo , Alcoholismo/patología , Animales , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Modelos Animales de Enfermedad , Etanol/efectos adversos , Etanol/metabolismo , Femenino , Humanos , Mucosa Intestinal , Intestinos/microbiología , Intestinos/patología , Macaca mulatta , Masculino , Transcriptoma
16.
Viral Immunol ; 29(4): 212-27, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27008411

RESUMEN

microRNAs (miRNAs) are small noncoding RNAs that are key regulators of biological processes, including the immune response to viral infections. Differential expression levels of cellular miRNAs and their predicted targets have been described in the lungs of H1N1-infected BALB/c mice, the lungs of H5N1 influenza-infected cynomolgus macaques, and in peripheral blood mononuclear cells (PBMCs) of critically ill patients infected with 2009 pandemic H1N1. However, a longitudinal analysis of changes in the expression of miRNAs and their targets during influenza infection and how they relate to viral replication and host response has yet to be carried out. In the present study, we conducted a comprehensive analysis of innate and adaptive immune responses as well as the expression of several miRNAs and their validated targets in both peripheral blood and bronchoalveolar lavage (BAL) collected from rhesus macaques over the course of infection with the 2009 H1N1 virus A/Mexico/4108/2009 (MEX4108). We describe a distinct set of differentially expressed miRNAs in BAL and PBMCs, which regulate the expression of genes involved in inflammation, immune response, and regulation of cell cycle and apoptosis.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Macaca mulatta/inmunología , MicroARNs/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Apoptosis/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Ciclo Celular/inmunología , Femenino , Perfilación de la Expresión Génica , Inflamación/inmunología , Inflamación/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Macaca mulatta/sangre , Macaca mulatta/virología , MicroARNs/metabolismo , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/virología , Carga Viral/inmunología , Replicación Viral/inmunología
17.
J Immunol ; 196(1): 182-95, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26621857

RESUMEN

Several lines of evidence indicate that chronic alcohol use disorder leads to increased susceptibility to several viral and bacterial infections, whereas moderate alcohol consumption decreases the incidence of colds and improves immune responses to some pathogens. In line with these observations, we recently showed that heavy ethanol intake (average blood ethanol concentrations > 80 mg/dl) suppressed, whereas moderate alcohol consumption (blood ethanol concentrations < 50 mg/dl) enhanced, T and B cell responses to modified vaccinia Ankara vaccination in a nonhuman primate model of voluntary ethanol consumption. To uncover the molecular basis for impaired immunity with heavy alcohol consumption and enhanced immune response with moderate alcohol consumption, we performed a transcriptome analysis using PBMCs isolated on day 7 post-modified vaccinia Ankara vaccination, the earliest time point at which we detected differences in T cell and Ab responses. Overall, chronic heavy alcohol consumption reduced the expression of immune genes involved in response to infection and wound healing and increased the expression of genes associated with the development of lung inflammatory disease and cancer. In contrast, chronic moderate alcohol consumption upregulated the expression of genes involved in immune response and reduced the expression of genes involved in cancer. To uncover mechanisms underlying the alterations in PBMC transcriptomes, we profiled the expression of microRNAs within the same samples. Chronic heavy ethanol consumption altered the levels of several microRNAs involved in cancer and immunity and known to regulate the expression of mRNAs differentially expressed in our data set.


Asunto(s)
Consumo de Bebidas Alcohólicas/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Humoral/genética , Linfocitos T/inmunología , Virus Vaccinia/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Enfermedades Cardiovasculares/inmunología , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Etanol/sangre , Perfilación de la Expresión Génica , Enfermedades Pulmonares Obstructivas/inmunología , Macaca mulatta , Masculino , MicroARNs/biosíntesis , MicroARNs/genética , Neoplasias/genética , Neoplasias/inmunología , ARN Mensajero/biosíntesis , Vacuna contra Viruela/inmunología , Vacunación , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología
18.
Artículo en Inglés | MEDLINE | ID: mdl-26375241

RESUMEN

Several studies have described a dose-dependent effect of alcohol on human health with light to moderate drinkers having a lower risk of all-cause mortality than abstainers, while heavy drinkers are at the highest risk. In the case of the immune system, moderate alcohol consumption is associated with reduced inflammation and improved responses to vaccination, while chronic heavy drinking is associated with a decreased frequency of lymphocytes and increased risk of both bacterial and viral infections. However, the mechanisms by which alcohol exerts a dose-dependent effect on the immune system remain poorly understood due to a lack of systematic studies that examine the effect of multiple doses and different time courses. This review will summarize our current understanding of the impact of moderate versus excessive alcohol consumption on the innate and adaptive branches of the immune system derived from both in vitro as well as in vivo studies carried out in humans and animal model studies.


Asunto(s)
Trastornos Relacionados con Alcohol/inmunología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Sistema Inmunológico/efectos de los fármacos , Animales , Depresores del Sistema Nervioso Central/metabolismo , Depresores del Sistema Nervioso Central/toxicidad , Etanol/metabolismo , Etanol/toxicidad , Humanos
19.
Alcohol Res ; 37(2): 185-97, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26695744

RESUMEN

Alcohol exposure, and particularly chronic heavy drinking, affects all components of the adaptive immune system. Studies both in humans and in animal models determined that chronic alcohol abuse reduces the number of peripheral T cells, disrupts the balance between different T-cell types, influences T-cell activation, impairs T-cell functioning, and promotes T-cell apoptosis. Chronic alcohol exposure also seems to cause loss of peripheral B cells, while simultaneously inducing increased production of immunoglobulins. In particular, the levels of antibodies against liver-specific autoantigens are increased in patients with alcoholic liver disease and may promote alcohol-related liver damage. Finally, chronic alcohol exposure in utero interferes with normal T-cell and B-cell development, which may increase the risk of infections during both childhood and adulthood. Alcohol's impact on T cells and B cells increases the risk of infections (e.g., pneumonia, HIV infection, hepatitis C virus infection, and tuberculosis), impairs responses to vaccinations against such infections, exacerbates cancer risk, and interferes with delayed-type hypersensitivity. In contrast to these deleterious effects of heavy alcohol exposure, moderate alcohol consumption may have beneficial effects on the adaptive immune system, including improved responses to vaccination and infection. The molecular mechanisms underlying ethanol's impact on the adaptive immune system remain poorly understood.


Asunto(s)
Inmunidad Adaptativa/inmunología , Alcoholismo/inmunología , Linfocitos B/inmunología , Linfocitos T/inmunología , Tuberculosis/inmunología , Virosis/inmunología , Infecciones por VIH/inmunología , Hepatitis C/inmunología , Humanos , Inmunoglobulinas/inmunología , Gripe Humana/inmunología , Hepatopatías Alcohólicas/inmunología , Activación de Linfocitos/inmunología , Vacunación
20.
J Biol Chem ; 289(38): 26167-26177, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25086045

RESUMEN

The serine/threonine protein kinase YpkA is an essential virulence factor produced by pathogenic Yersinia species. YpkA is delivered into host mammalian cells via a type III secretion system and localizes to the inner side of the plasma membrane. We have previously shown that YpkA binds to and phosphorylates the α subunit of the heterotrimeric G protein complex, Gαq, resulting in inhibition of Gαq signaling. To identify residues in YpkA involved in substrate binding activity we generated GFP-YpkA N-terminal deletion mutants and performed coimmunoprecipitation experiments. We located a substrate-binding domain on amino acids 40-49 of YpkA, which lies within the previously identified membrane localization domain on YpkA. Deletion of amino acids 40-49 on YpkA interfered with substrate binding, substrate phosphorylation and substrate inhibition. Autophosphorylation regulates the kinase activity of YpkA. To dissect the mechanism by which YpkA transmits signals, we performed nano liquid chromatography coupled to tandem mass spectrometry to map in vivo phosphorylation sites. Multiple serine phosphorylation sites were identified in the secretion/translocation region, kinase domain, and C-terminal region of YpkA. Using site-directed mutagenesis we generated multiple YpkA constructs harboring specific serine to alanine point mutations. Our results demonstrate that multiple autophosphorylation sites within the N terminus regulate YpkA kinase activation, whereas mutation of serine to alanine within the C terminus of YpkA had no effect on kinase activity. YpkA autophosphorylation on multiple sites may be a strategy used by pathogenic Yersinia to prevent inactivation of this important virulence protein by host proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Yersinia enterocolitica/enzimología , Proteínas Bacterianas/química , Dominio Catalítico , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA