Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Immun Ageing ; 20(1): 49, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37752597

RESUMEN

BACKGROUND: Preclinical models are often used for cancer studies and evaluation of novel therapeutics. The relevance of these models has vastly improved with mice bearing a human immune system, especially in the context of immunotherapy. Nonetheless, cancer is an age-related disease, and studies often overlook the effects of aging. Here we have established a humanized mouse model of human immune aging to investigate the role of this phenomenon on liver tumor dynamics. METHODS: Multiple organs and tissues (blood, thymus, lung, liver, spleen and bone marrow) were harvested from NOD-scid IL2rγ-/- (NIKO) mice reconstituted with human immune cells, over a period of 60 weeks post-birth, for immune profiling. Young and aging immune cells were compared for transcriptomic changes and functional differences. Effect of immune aging was investigated in a liver cancer humanized mouse model. RESULTS: Focusing on the T cell population, which is central to cancer immunosurveillance and immunotherapy, we showed that the proportion of naïve T cells declined while memory subsets and senescent-like cells increased with age. RNA-sequencing revealed that downregulated genes were related to immune responses and processes, and this was corroborated by reduced cytokine production in aging T cells. Finally, we showed faster liver tumor growth in aging than younger humanized mice, which could be attributed to specific pathways of aging T cell exhaustion. CONCLUSION: Our work improves on existing humanized (immune) mouse model and highlights the importance of considering immune aging in liver cancer modeling.

2.
Sci Rep ; 13(1): 12484, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528224

RESUMEN

The COVID-19 pandemic has sickened millions, cost lives and has devastated the global economy. Various animal models for experimental infection with SARS-CoV-2 have played a key role in many aspects of COVID-19 research. Here, we describe a humanized hACE2 (adenovirus expressing hACE2) NOD-SCID IL2Rγ-/- (NIKO) mouse model and compare infection with ancestral and mutant (SARS-CoV-2-∆382) strains of SARS-CoV-2. Immune cell infiltration, inflammation, lung damage and pro-inflammatory cytokines and chemokines was observed in humanized hACE2 NIKO mice. Humanized hACE2 NIKO mice infected with the ancestral and mutant SARS-CoV-2 strain had lung inflammation and production of pro-inflammatory cytokines and chemokines. This model can aid in examining the pathological basis of SARS-CoV-2 infection in a human immune environment and evaluation of therapeutic interventions.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Animales , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pandemias , Modelos Animales de Enfermedad , Citocinas , Ratones Transgénicos , Pulmón
3.
Adv Sci (Weinh) ; 10(28): e2302175, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37544893

RESUMEN

In keeping with the rule of "form follows function", morphological aspects of a cell can reflect its role. Here, it is shown that the cellular granularity of a lymphocyte, represented by its intrinsic side scatter (SSC), is a potent indicator of its cell state and function. The granularity of a lymphocyte increases from naïve to terminal effector state. High-throughput cell-sorting yields a SSChigh population that can mediate immediate effector functions, and a highly prolific SSClow population that can give rise to the replenishment of the memory pool. CAR-T cells derived from the younger SSClow population possess desirable attributes for immunotherapy, manifested by increased naïve-like cells and stem cell memory (TSCM )-like cells together with a balanced CD4/CD8 ratio, as well as enhanced target-killing in vitro and in vivo. Altogether, lymphocyte segregation based on biophysical properties is an effective approach for label-free selection of cells that share collective functions and can have important applications for cell-based immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia Adoptiva , Inmunoterapia , Linfocitos T CD4-Positivos , Células Madre
4.
Sci Adv ; 8(47): eadd1187, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36417514

RESUMEN

In recent decades, chimeric antigen receptor (CAR)-engineered immune effector cells have demonstrated promising antileukemic activity. Nevertheless, their efficacy remains unsatisfactory on solid cancers, plausibly due to the influence of tumor microenvironments (TME). In a novel mouse cancer model with a humanized immune system, tumor-infiltrating immunosuppressive leukocytes and exhausted programmed death protein-1 (PD-1)high T cells were found, which better mimic patient TME, allowing the screening and assessment of immune therapeutics. Particularly, membrane-bound programmed death ligand 1 (PD-L1) level was elevated on a tumor cell surface, which serves as an attractive target for natural killer (NK) cell-mediated therapy. Hematopoietic stem cell-derived CAR-NK (CAR pNK) cells targeting the PD-L1 showed enhanced in vitro and in vivo anti-solid tumor function. The CAR pNK cells and nivolumab resulted in a synergistic anti-solid tumor response. Together, our study highlights a robust platform to develop and evaluate the antitumor efficacy and safety of previously unexplored therapeutic regimens.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Ratones , Animales , Receptores Quiméricos de Antígenos/metabolismo , Nivolumab/farmacología , Receptor de Muerte Celular Programada 1 , Antígeno B7-H1/metabolismo , Neoplasias/metabolismo , Células Asesinas Naturales , Modelos Animales de Enfermedad , Ligandos , Microambiente Tumoral
5.
Hepatology ; 74(3): 1395-1410, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33738839

RESUMEN

BACKGROUND AND AIMS: Recent development of multiple treatments for human hepatocellular carcinoma (HCC) has allowed for the selection of combination therapy to enhance the effectiveness of monotherapy. Optimal selection of therapies is based on both HCC and its microenvironment. Therefore, it is critical to develop and validate preclinical animal models for testing clinical therapeutic solutions. APPROACH AND RESULTS: We established cell line-based or patient-derived xenograft-based humanized-immune-system mouse models with subcutaneous and orthotopic HCC. Mice were injected with human-specific antibodies (Abs) to deplete human immune cells. We analyzed the transcription profiles of HCC cells and human immune cells by using real-time PCR and RNA sequencing. The protein level of HCC tumor cells/tissues or human immune cells was determined by using flow cytometry, western blotting, and immunohistochemistry. The HCC tumor size was measured after single, dual-combination, and triple-combination treatment using N-(1',2-Dihydroxy-1,2'-binaphthalen-4'-yl)-4-methoxybenzenesulfonamide (C188-9), bevacizumab, and pembrolizumab. In this study, human immune cells in the tumor microenvironment were strongly selected and modulated by HCC, which promoted the activation of the IL-6/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in tumor cells and led to augmented HCC proliferation and angiogenesis by releasing angiogenic cytokines in humanized-immune-system mice with HCC. In particular, intratumor human cluster of differentiation-positive (hCD14+ ) cells could produce IL-33 through damage-associated molecular pattern/Toll-like receptor 4/activator protein 1, which up-regulated IL-6 in other intratumor immune cells and activated the JAK2/STAT3 pathway in HCC. Specific knockdown of the CD14 gene in human monocytes could impair IL-33 production induced by cell lysates. Subsequently, we evaluated the in vivo anti-HCC effect of C188-9, bevacizumab, and pembrolizumab. The results showed that the anti-HCC effect of triple-combination therapy was superior to that of single or dual treatments. CONCLUSIONS: Humanized-immune-system HCC mouse models are suitable for identifying targets from cancer and immune components and for testing combinational therapies.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neovascularización Patológica/inmunología , Microambiente Tumoral/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Bevacizumab/farmacología , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Humanos , Interleucina-6/inmunología , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Receptores de Lipopolisacáridos/metabolismo , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/genética , Ratones , Naftoles/farmacología , Trasplante de Neoplasias , Neovascularización Patológica/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Sulfonamidas/farmacología , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Front Immunol ; 11: 553362, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193321

RESUMEN

Advancements in science enable researchers to constantly innovate and create novel biologics. However, the use of non-human animal models during the development of biologics impedes identification of precise in vivo interactions between the human immune system and treatments. Due to lack of this understanding, adverse effects are frequently observed in healthy volunteers and patients exposed to potential biologics during clinical trials. In this study, we evaluated and compared the effects of known immunotoxic biologics, Proleukin®/IL-2 and OKT3 in humanized mice (reconstituted with human fetal cells) to published clinical outcomes. We demonstrated that humanized mice were able to recapitulate in vivo pathological changes and human-specific immune responses, such as elevated cytokine levels and modulated lymphocytes and myeloid subsets. Given the high similarities of immunological side effects observed between humanized mice and clinical studies, this model could be used to assess immunotoxicity of biologics at a pre-clinical stage, without placing research participants and/or patients at risk.


Asunto(s)
Interleucina-2/análogos & derivados , Modelos Inmunológicos , Animales , Evaluación de Medicamentos , Feto , Humanos , Interleucina-2/efectos adversos , Interleucina-2/inmunología , Interleucina-2/farmacología , Ratones , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología
7.
Front Immunol ; 11: 580968, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013934

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) has been on a global rise. While animal models have rendered valuable insights to the pathogenesis of NAFLD, discrepancy with patient data still exists. Since non-alcoholic steatohepatitis (NASH) involves chronic inflammation, and CD4+ T cell infiltration of the liver is characteristic of NASH patients, we established and characterized a humanized mouse model to identify human-specific immune response(s) associated with NAFLD progression. Immunodeficient mice engrafted with human immune cells (HIL mice) were fed with high fat and high calorie (HFHC) or chow diet for 20 weeks. Liver histology and immune profile of HIL mice were analyzed and compared with patient data. HIL mice on HFHC diet developed steatosis, inflammation and fibrosis of the liver. Human CD4+ central and effector memory T cells increased within the liver and in the peripheral blood of our HIL mice, accompanied by marked up-regulation of pro-inflammatory cytokines (IL-17A and IFNγ). In vivo depletion of human CD4+ T cells in HIL mice reduced liver inflammation and fibrosis, but not steatosis. Our results highlight CD4+ memory T cell subsets as important drivers of NAFLD progression from steatosis to fibrosis and provides a humanized mouse model for pre-clinical evaluation of potential therapeutics.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Cirrosis Hepática Experimental/etiología , Cirrosis Hepática Experimental/inmunología , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Citocinas/sangre , Dieta Alta en Grasa/efectos adversos , Femenino , Células Madre Fetales/trasplante , Hepatocitos/trasplante , Xenoinjertos , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Cirrosis Hepática Experimental/patología , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Enfermedad del Hígado Graso no Alcohólico/patología
8.
Cancers (Basel) ; 12(4)2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32331230

RESUMEN

Immune checkpoint blockade (ICB) monotherapy shows early promise for the treatment of nasopharyngeal carcinoma (NPC) in patients. Nevertheless, limited representative NPC models hamper preclinical studies to evaluate the efficacy of novel ICB and combination regimens. In the present study, we engrafted NPC biopsies in non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain-null (NSG) mice and established humanized mouse NPC-patient-derived xenograft (NPC-PDX) model successfully. Epstein-Barr virus was detected in the NPC in both NSG and humanized mice as revealed by Epstein-Barr virus-encoded small RNA (EBER) in situ hybridization (ISH) and immunohistochemical (IHC) staining. In the NPC-bearing humanized mice, the percentage of tumor-infiltrating CD8+ cytotoxic T cells was lowered, and the T cells expressed higher levels of various inhibitory receptors, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) than those in blood. The mice were then treated with nivolumab and ipilimumab, and the anti-tumor efficacy of combination immunotherapy was examined. In line with paired clinical data, the NPC-PDX did not respond to the treatment in terms of tumor burden, whilst an immunomodulatory response was elicited in the humanized mice. From our results, human proinflammatory cytokines, such as interferon-gamma (IFN-γ) and interleukin-6 (IL-6) were significantly upregulated in plasma. After treatment, there was a decrease in CD4/CD8 ratio in the NPC-PDX, which also simulated the modulation of intratumoral CD4/CD8 profile from the corresponding donor. In addition, tumor-infiltrating T cells were re-activated and secreted more IFN-γ towards ex vivo stimulation, suggesting that other factors, including soluble mediators and metabolic milieu in tumor microenvironment may counteract the effect of ICB treatment and contribute to the tumor progression in the mice. Taken together, we have established and characterized a novel humanized mouse NPC-PDX model, which plausibly serves as a robust platform to test for the efficacy of immunotherapy and may predict clinical outcomes in NPC patients.

9.
J Leukoc Biol ; 107(5): 797-807, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31922289

RESUMEN

Recently a G-protein-coupled receptor, MAS Related GPR Family Member X2 (MRGPRX2), was identified as a specific receptor on human mast cells responsible for IgE independent adverse drug reactions (ADR). Although a murine homologue, Mrgprb2, has been identified for this receptor, its affinity for many ADR-causing drugs is poor making it difficult to undertake in vivo studies to examine mechanisms of ADR and to develop therapeutic strategies. Here, we have created humanized mice capable of generating MRGPRX2-expressing human MCs allowing for the study of MRGPRX2 MCs-mediated ADR in vitro as well as in vivo. Humanized mice were generated by hydrodynamic-injection of plasmids expressing human GM-CSF and IL-3 into NOD-scid IL2R-γ-/- strain of mice that had been transplanted with human hematopoietic stem cells. These GM/IL-3 humice expressed high numbers of tissue human MCs but the MRGPRX2 receptor expressed in MCs were limited to few body sites including the skin. Importantly, large numbers of MRGPRX2-expressing human MCs could be cultured from the bone marrow of GM/IL-3 humice revealing these mice to be an important source of human MCs for in vitro studies of MRGPRX2-related MCs activities. When GM/IL-3 humice were exposed to known ADR causing contrast agents (meglumine and gadobutrol), the humice were found to experience anaphylaxis analogous to the clinical situation. Thus, GM/IL-3 humice represent a valuable model for investigating in vivo interactions of ADR-causing drugs and human MCs and their sequelae, and these mice are also a source of human MRGPRX2-expressing MCs for in vitro studies.


Asunto(s)
Modelos Animales de Enfermedad , Erupciones por Medicamentos/inmunología , Mastocitos/inmunología , Proteínas del Tejido Nervioso/inmunología , Receptores Acoplados a Proteínas G/inmunología , Receptores de Neuropéptido/inmunología , Animales , Medios de Contraste/toxicidad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Interleucina-3/genética , Mastocitos/efectos de los fármacos , Meglumina/toxicidad , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Compuestos Organometálicos/toxicidad
10.
J Virol ; 93(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30429352

RESUMEN

Since the discovery of enterovirus A71 (EV-A71) half a century ago, it has been recognized as the cause of large-scale outbreaks of hand-foot-and-mouth disease worldwide, particularly in the Asia-Pacific region, causing great concern for public health and economic burdens. Detailed mechanisms on the modulation of immune responses after EV-A71 infection have not been fully known, and the lack of appropriate models hinders the development of promising vaccines and drugs. In the present study, NOD-scid IL2Rγ-/- (NSG) mice with a human immune system (humanized mice) at the age of 4 weeks were found to be susceptible to a human isolate of EV-A71 infection. After infection, humanized mice displayed limb weakness, which is similar to the clinical features found in some of the EV-A71-infected patients. Histopathological examination indicated the presence of vacuolation, gliosis, or meningomyelitis in brain stem and spinal cord, which were accompanied by high viral loads detected in these organs. The numbers of activated human CD4+ and CD8+ T cells were upregulated after EV-A71 infection, and EV-A71-specific human T cell responses were found. Furthermore, the secretion of several proinflammatory cytokines, such as human gamma interferon (IFN-γ), interleukin-8 (IL-8), and IL-17A, was elevated in the EV-A71-infected humanized mice. Taken together, our results suggested that the humanized mouse model permits insights into the human immune responses and the pathogenesis of EV-A71 infection, which may provide a platform for the evaluation of anti-EV-A71 drug candidates in the future.IMPORTANCE Despite causing self-limited hand-food-and-mouth disease in younger children, EV-A71 is consistently associated with severe forms of neurological complications and pulmonary edema. Nevertheless, only limited vaccines and drugs have been developed over the years, which is possibly due to a lack of models that can more accurately recapitulate human specificity, since human is the only natural host for wild-type EV-A71 infection. Our humanized mouse model not only mimics histological symptoms in patients but also allows us to investigate the function of the human immune system during infection. It was found that human T cell responses were activated, accompanied by an increase in the production of proinflammatory cytokines in EV-A71-infected humanized mice, which might contribute to the exacerbation of disease pathogenesis. Collectively, this model allows us to delineate the modulation of human immune responses during EV-A71 infection and may provide a platform to evaluate anti-EV-A71 drug candidates in the future.


Asunto(s)
Linfocitos T CD8-positivos/patología , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/patología , Feto/patología , Carga Viral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Enterovirus Humano A/aislamiento & purificación , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/virología , Feto/inmunología , Feto/virología , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID
11.
Gut ; 67(10): 1845-1854, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29602780

RESUMEN

OBJECTIVE: As the current therapeutic strategies for human hepatocellular carcinoma (HCC) have been proven to have limited effectiveness, immunotherapy becomes a compelling way to tackle the disease. We aim to provide humanised mouse (humice) models for the understanding of the interaction between human cancer and immune system, particularly for human-specific drug testing. DESIGN: Patient-derived xenograft tumours are established with type I human leucocyte antigen matched human immune system in NOD-scid Il2rg-/- (NSG) mice. The longitudinal changes of the tumour and immune responses as well as the efficacy of immune checkpoint inhibitors are investigated. RESULTS: Similar to the clinical outcomes, the human immune system in our model is educated by the tumour and exhibits exhaustion phenotypes such as a significant declination of leucocyte numbers, upregulation of exhaustion markers and decreased the production of human proinflammatory cytokines. Notably, cytotoxic immune cells decreased more rapidly compared with other cell types. Tumour infiltrated T cells have much higher expression of exhaustion markers and lower cytokine production compared with peripheral T cells. In addition, tumour-associated macrophages and myeloid-derived suppressor cells are found to be highly enriched in the tumour microenvironment. Interestingly, the tumour also changes gene expression profiles in response to immune responses by upregulating immune checkpoint ligands. Most importantly, in contrast to the NSG model, our model demonstrates both therapeutic and side effects of immune checkpoint inhibitors pembrolizumab and ipilimumab. CONCLUSIONS: Our work provides a model for immune-oncology study and a useful parallel-to-human platform for anti-HCC drug testing, especially immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Carcinoma Hepatocelular , Inmunoterapia/métodos , Ipilimumab/farmacología , Neoplasias Hepáticas , Microambiente Tumoral/inmunología , Animales , Antineoplásicos Inmunológicos/farmacología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Citocinas/metabolismo , Modelos Animales de Enfermedad , Xenoinjertos/inmunología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Ratones , Ratones Endogámicos NOD
12.
Sci Rep ; 8(1): 4726, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549333

RESUMEN

Bats are an important animal model with long lifespans, low incidences of tumorigenesis and an ability to asymptomatically harbour pathogens. Currently, in vivo studies of bats are hampered due to their low reproduction rates. To overcome this, we transplanted bat cells from bone marrow (BM) and spleen into an immunodeficient mouse strain NOD-scid IL-2R-/- (NSG), and have successfully established stable, long-term reconstitution of bat immune cells in mice (bat-mice). Immune functionality of our bat-mouse model was demonstrated through generation of antigen-specific antibody response by bat cells following immunization. Post-engraftment of total bat BM cells and splenocytes, bat immune cells survived, expanded and repopulated the mouse without any observable clinical abnormalities. Utilizing bat's remarkable immunological functions, this novel model has a potential to be transformed into a powerful platform for basic and translational research.


Asunto(s)
Trasplante de Médula Ósea/métodos , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Linfocitos/inmunología , Inmunodeficiencia Combinada Grave/terapia , Quimera por Trasplante/inmunología , Animales , Quirópteros , Rechazo de Injerto/prevención & control , Ratones , Ratones Endogámicos NOD , Ratones SCID , Inmunodeficiencia Combinada Grave/inmunología
13.
Asia Pac J Clin Nutr ; 27(2): 421-432, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29384332

RESUMEN

BACKGROUND AND OBJECTIVES: This study aimed to determine the relationship between breakfast consumption and body weight status among primary and secondary school children in Malaysia. METHODS AND STUDY DESIGN: This nationwide cross-sectional study involved 5,332 primary school children aged 6 to 12 years and 3,000 secondary school children aged 13 to 17 years. Height and weight were measured and BMI-for-age was determined. Socio-demographic backgrounds, breakfast habits and physical activity levels were assessed using questionnaires. Breakfast frequency was defined as follows: breakfast skippers (ate breakfast 0-2 days/week), irregular breakfast eaters (ate breakfast 3-4 days/week) and regular breakfast eaters (ate breakfast ≥5 days/week). RESULTS: The overall prevalence of breakfast skippers and irregular breakfast eaters was 11.7% and 12.7% respectively. Breakfast skipping was related to age, sex, ethnicity, income and physical activity level. Among primary school boys and secondary school girls, the proportion of overweight/obesity was higher among breakfast skippers (boys: 43.9%, girls: 30.5%) than regular breakfast eaters (boys: 31.2%, girls: 22.7%). Among primary school children, only boys who skipped breakfast had a higher mean BMI-for-age z-score than regular breakfast eaters. Among secondary school boys and girls, BMI-for-age z-score was higher among breakfast skippers than regular breakfast eaters. Compared to regular breakfast eaters, primary school boys who skipped breakfast were 1.71 times (95% CI=1.26-2.32, p=0.001) more likely to be overweight/obese, while the risk was lower in primary school girls (OR=1.36, 95% CI=1.02-1.81, p=0.039) and secondary school girls (OR=1.38, 95% CI=1.01-1.90, p=0.044). CONCLUSION: Regular breakfast consumption was associated with a healthier body weight status and is a dietary behaviour which should be encouraged.


Asunto(s)
Peso Corporal , Desayuno , Adolescente , Índice de Masa Corporal , Niño , Estudios Transversales , Femenino , Humanos , Malasia , Masculino , Encuestas Nutricionales
14.
Sci Rep ; 7(1): 16642, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29192160

RESUMEN

Mouse models have contributed to the bulk of knowledge on Systemic Lupus Erythematosus (SLE). Nevertheless, substantial differences exist between human and mouse immune system. We aimed to establish and characterise a SLE model mediated by human immune system. Injection of pristane into immunodeficient mice reconstituted with human immune system (humanised mice) recapitulated key SLE features, including: production of human anti-nuclear autoantibodies, lupus nephritis, and pulmonary serositis. There was a reduction in the number of human lymphocytes in peripheral blood, resembling lymphopenia in SLE patients. Concurrently, B cells and T cells were systemically hyperactivated, with a relative expansion of CD27+ and CD27-IgD- memory B cells, increased number of plasmablasts/plasma cells, and accumulation of effector memory T cells. There was also an increased production of human pro-inflammatory cytokines, including: IFN-γ, IL-8, IL-18, MCP-1, and IL-6, suggesting their role in SLE pathogenesis. Increased expression of type I IFN signature genes was also found in human hepatocytes. Altogether, we showed an SLE model that was mediated by human immune system, and which recapitulated key clinical and immunological SLE features. The advancements of humanised mice SLE model would provide an in vivo platform to facilitate translational studies and pre-clinical evaluations of human-specific mechanisms and immunotherapies.


Asunto(s)
Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/etiología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Biomarcadores , Biopsia , Citocinas/metabolismo , Perfilación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hepatocitos/metabolismo , Humanos , Isotipos de Inmunoglobulinas/sangre , Isotipos de Inmunoglobulinas/inmunología , Inmunohistoquímica , Inmunofenotipificación , Mediadores de Inflamación , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/etiología , Nefritis Lúpica/patología , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Transgénicos
15.
J Hematol Oncol ; 10(1): 162, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28985760

RESUMEN

BACKGROUND: Xenotransplantation of patient-derived AML (acute myeloid leukemia) cells in NOD-scid Il2rγ null (NSG) mice is the method of choice for evaluating this human hematologic malignancy. However, existing models constructed using intravenous injection in adult or newborn NSG mice have inferior engraftment efficiency, poor peripheral blood engraftment, or are difficult to construct. METHODS: Here, we describe an improved AML xenograft model where primary human AML cells were injected into NSG newborn pups intrahepatically. RESULTS: Introduction of primary cells from AML patients resulted in high levels of engraftment in peripheral blood, spleen, and bone marrow (BM) of recipient mice. The phenotype of engrafted AML cells remained unaltered during serial transplantation. The mice developed features that are consistent with human AML including spleen enlargement and infiltration of AML cells into multiple organs. Importantly, we demonstrated that although leukemic stem cell activity is enriched and mediated by CD34+CD117+ subpopulation, CD34+CD117- subpopulation can acquire CD34+CD117+ phenotype through de-differentiation. Lastly, we evaluated the therapeutic potential of Sorafenib and Regorafenib in this AML model and found that periphery and spleen AML cells are sensitive to these treatments, whereas BM provides a protective environment to AML. CONCLUSIONS: Collectively, our improved model is robust, easy-to-construct, and reliable for pre-clinical AML studies.


Asunto(s)
Leucemia Mieloide Aguda/genética , Trasplante Heterólogo/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID
16.
PLoS One ; 12(9): e0184127, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28886065

RESUMEN

Hepatitis C is a liver disease caused by infection of the Hepatitis C virus (HCV). Many individuals infected by the virus are unable to resolve the viral infection and develop chronic hepatitis, which can lead to formation of liver cirrhosis and cancer. To understand better how initial HCV infections progress to chronic liver diseases, we characterised the long term pathogenic effects of HCV infections with the use of a humanised mouse model (HIL mice) we have previously established. Although HCV RNA could be detected in infected mice up to 9 weeks post infection, HCV infected mice developed increased incidences of liver fibrosis, granulomatous inflammation and tumour formation in the form of hepatocellular adenomas or hepatocellular carcinomas by 28 weeks post infection compared to uninfected mice. We also demonstrated that chronic liver inflammation in HCV infected mice was mediated by the human immune system, particularly by monocytes/macrophages and T cells which exhibited exhaustion phenotypes. In conclusion, HIL mice can recapitulate some of the clinical symptoms such as chronic inflammation, immune cell exhaustion and tumorigenesis seen in HCV patients. Our findings also suggest that persistence of HCV-associated liver disease appear to require initial infections of HCV and immune responses but not long term HCV viraemia.


Asunto(s)
Carcinoma Hepatocelular/etiología , Transformación Celular Neoplásica , Hepacivirus , Hepatitis C Crónica/complicaciones , Hepatitis C Crónica/inmunología , Neoplasias Hepáticas/etiología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica/inmunología , Citocinas/sangre , Modelos Animales de Enfermedad , Hepacivirus/inmunología , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/virología , Pruebas de Función Hepática , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Albúmina Sérica/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Viremia/inmunología , Viremia/virología
17.
Blood ; 130(18): 1995-2005, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28851698

RESUMEN

Many immune parameters show circadian rhythms during the 24-hour day in mammals. The most striking circadian oscillation is the number of circulating immune cells that display an opposite rhythm between humans and mice. The physiological roles and mechanisms of circadian variations in mouse leukocytes are well studied, whereas for humans they remain unclear because of the lack of a proper model. In this study, we found that consistent with their natural host species, mouse and human circulating leukocytes exhibited opposite circadian oscillations in humanized mice. This cyclic pattern of trafficking correlated well with the diurnal expression levels of C-X-C chemokine receptor 4, which were controlled by the intracellular hypoxia-inducible factor 1α/aryl hydrocarbon receptor nuclear translocator-like heterodimer. Furthermore, we also discovered that p38 mitogen-activated protein kinases/mitogen-activated 2 had opposite effects between mice and humans in generating intracellular reactive oxygen species, which subsequently regulated HIF-1α expression. In conclusion, we propose humanized mice as a robust model for human circadian studies and reveal insights on a novel molecular clock network in the human circadian rhythm.


Asunto(s)
Ritmo Circadiano/fisiología , Leucocitos/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Movimiento Celular , Quimiocina CXCL12/metabolismo , Humanos , Síndrome Jet Lag/fisiopatología , Ratones , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Food Nutr Res ; 61(1): 1304692, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28469545

RESUMEN

Background: The association between different types of breakfast meals and nutrient intakes has been studied to a lesser extent. Objective: This study compared nutrient intakes at breakfast and throughout the day between Malaysian children who consumed ready-to-eat cereals (RTEC) and those who did not. Methods: Anthropometric and dietary data for 1955 children aged 6-12 years from the MyBreakfast study were used in the analysis. Results: Overall, 18% of the children consumed RTEC at breakfast on at least one of the recall days. RTEC consumption was associated with younger age, urban areas, higher income and education level of parents. Among consumers, RTEC contributed 10% and 15% to daily intakes of calcium and iron respectively and ≥20% to daily intakes of vitamin C, thiamin, riboflavin and niacin. RTEC consumers had significantly higher mean intakes of vitamin C, thiamin, riboflavin, niacin, calcium, iron and sugar but lower intakes of fat and sodium than non-RTEC consumers at breakfast and for the total day. Conclusion: Consumption of fortified RTEC at breakfast was associated with lower fat and sodium intakes and higher intakes of several micronutrients both at breakfast and for the total day. However, total sugar intakes appeared to be higher.

19.
Nutrients ; 8(12)2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27916932

RESUMEN

BACKGROUND: The 2010 World Health Organisation (WHO) Infant and Young Child Feeding (IYCF) indicators are useful for monitoring feeding practices. METHODS: A total sample of 300 subjects aged 6 to 23 months was recruited from urban suburbs of Kuala Lumpur and Putrajaya. Compliance with each IYCF indicator was computed according to WHO recommendations. Dietary intake based on two-day weighed food records was obtained from a sub-group (N = 119) of the total sample. The mean adequacy ratio (MAR) value was computed as an overall measure of dietary intake adequacy. Contributions of core IYCF indicators to MAR were determined by multinomial logistic regression. RESULTS: Generally, the subjects showed high compliance for (i) timely introduction of complementary foods at 6 to 8 months (97.9%); (ii) minimum meal frequency among non-breastfed children aged 6 to 23 months (95.2%); (iii) consumption of iron-rich foods at 6 to 23 months (92.3%); and minimum dietary diversity (78.0%). While relatively high proportions achieved the recommended intake levels for protein (87.4%) and iron (71.4%), lower proportions attained the recommendations for calcium (56.3%) and energy (56.3%). The intake of micronutrients was generally poor. The minimum dietary diversity had the greatest contribution to MAR (95% CI: 3.09, 39.87) (p = 0.000) among the core IYCF indicators. CONCLUSION: Malaysian urban infants and toddlers showed moderate to high compliance with WHO IYCF indicators. The robustness of the analytical approach in this study in quantifying contributions of IYCF indicators to MAR should be further investigated.


Asunto(s)
Dieta Saludable , Dieta/efectos adversos , Métodos de Alimentación/efectos adversos , Fenómenos Fisiológicos Nutricionales del Lactante , Política Nutricional , Cooperación del Paciente , Salud Suburbana , Lactancia Materna/etnología , Cuidadores , Guarderías Infantiles , Desarrollo Infantil , Estudios Transversales , Dieta/etnología , Registros de Dieta , Dieta Saludable/etnología , Femenino , Indicadores de Salud , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante/etnología , Malasia , Masculino , Encuestas Nutricionales , Padres , Cooperación del Paciente/etnología , Salud Suburbana/etnología , Organización Mundial de la Salud
20.
Gut ; 65(10): 1744-53, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26149491

RESUMEN

OBJECTIVE: HCV infection affects millions of people worldwide, and many patients develop chronic infection leading to liver cancers. For decades, the lack of a small animal model that can recapitulate HCV infection, its immunopathogenesis and disease progression has impeded the development of an effective vaccine and therapeutics. We aim to provide a humanised mouse model for the understanding of HCV-specific human immune responses and HCV-associated disease pathologies. DESIGN: Recently, we have established human liver cells with a matched human immune system in NOD-scid Il2rg(-/-) (NSG) mice (HIL mice). These mice are infected with HCV by intravenous injection, and the pathologies are investigated. RESULTS: In this study, we demonstrate that HIL mouse is capable of supporting HCV infection and can present some of the clinical symptoms found in HCV-infected patients including hepatitis, robust virus-specific human immune cell and cytokine responses as well as liver fibrosis and cirrhosis. Similar to results obtained from the analysis of patient samples, the human immune cells, particularly T cells and macrophages, play critical roles during the HCV-associated liver disease development in the HIL mice. Furthermore, our model is demonstrated to be able to reproduce the therapeutic effects of human interferon alpha 2a antiviral treatment. CONCLUSIONS: The HIL mouse provides a model for the understanding of HCV-specific human immune responses and HCV-associated disease pathologies. It could also serve as a platform for antifibrosis and immune-modulatory drug testing.


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis C Crónica , Interferón-alfa/uso terapéutico , Ratones Endogámicos NOD , Animales , Antivirales/uso terapéutico , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/fisiopatología , Humanos , Inmunidad Celular/inmunología , Interferón alfa-2 , Ratones , Proteínas Recombinantes/uso terapéutico , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...