Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Front Immunol ; 13: 1008764, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36159851

RESUMO

Colorectal cancer (CRC) is one of the most common cancers worldwide and demands more effective treatments. We sought to identify tumor selective CRC antigens and their therapeutic potential for cytotoxic T-cell targeting by transcriptomic and immunohistochemical analysis. LY6G6D was identified as a tumor selectively expressed CRC antigen, mainly in the microsatellite stable (MSS) subtype. A specific anti LY6G6D/CD3 T cell engager (TcE) was generated and demonstrated potent tumor cell killing and T cell activation in vitro. Ex vivo treatment of primary patient-derived CRC tumor slice cultures with the LY6G6D/CD3 TcE led to IFNγ secretion in LY6G6D positive tumor samples. In vivo, LY6G6D/CD3 TcE monotherapy demonstrated tumor regressions in pre-clinical mouse models of engrafted human CRC tumor cells and PBMCs. Lastly, 2D and 3D cocultures of LY6G6D positive and negative cells were used to explore the bystander killing of LY6G6D negative cells after specific activation of T cells by LY6G6D positive cells. LY6G6D/CD3 TcE treatment was shown to lyse target negative cells in the vicinity of target positive cells through a combined effect of IFNγ, TNFα and Fas/FasL. In summary, LY6G6D was identified as a selectively expressed CRC antigen that can be utilized to potently re-direct and activate cytotoxic T-cells to lyse LY6G6D expressing CRC using a TcE. This effect can be spread to target negative neighboring tumor cells, potentially leading to improved therapeutic efficacy.


Assuntos
Neoplasias Colorretais , Fator de Necrose Tumoral alfa , Animais , Antígenos de Neoplasias , Humanos , Imunoglobulinas , Ativação Linfocitária , Camundongos , Linfócitos T Citotóxicos
2.
J Med Internet Res ; 24(7): e32487, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35797104

RESUMO

BACKGROUND: Insufficient physical activity and unhealthy diets are contributing to the rise in noncommunicable diseases. Preventative mobile health (mHealth) interventions may help reverse this trend, but present bias might reduce their effectiveness. Future-self avatar interventions have resulted in behavior change in related fields, yet evidence of whether such interventions can change health behavior is lacking. OBJECTIVE: We aimed to investigate the impact of a future-self avatar mHealth intervention on physical activity and food purchasing behavior and examine the feasibility of a novel automated nutrition tracking system. We also aimed to understand how this intervention impacts related attitudinal and motivational constructs. METHODS: We conducted a 12-week parallel randomized controlled trial (RCT), followed by semistructured interviews. German-speaking smartphone users aged ≥18 years living in Switzerland and using at least one of the two leading Swiss grocery loyalty cards, were recruited for the trial. Data were collected from November 2020 to April 2021. The intervention group received the FutureMe intervention, a physical activity and food purchase tracking mobile phone app that uses a future-self avatar as the primary interface and provides participants with personalized food basket analysis and shopping tips. The control group received a conventional text- and graphic-based primary interface intervention. We pioneered a novel system to track nutrition by leveraging digital receipts from loyalty card data and analyzing food purchases in a fully automated way. Data were consolidated in 4-week intervals, and nonparametric tests were conducted to test for within- and between-group differences. RESULTS: We recruited 167 participants, and 95 eligible participants were randomized into either the intervention (n=42) or control group (n=53). The median age was 44 years (IQR 19), and the gender ratio was balanced (female 52/95, 55%). Attrition was unexpectedly high with only 30 participants completing the intervention, negatively impacting the statistical power. The FutureMe intervention led to small statistically insignificant increases in physical activity (median +242 steps/day) and small insignificant improvements in the nutritional quality of food purchases (median -1.28 British Food Standards Agency Nutrient Profiling System Dietary Index points) at the end of the intervention. Intrinsic motivation significantly increased (P=.03) in the FutureMe group, but decreased in the control group. Outcome expectancy directionally increased in the FutureMe group, but decreased in the control group. Leveraging loyalty card data to track the nutritional quality of food purchases was found to be a feasible and accepted fully automated nutrition tracking system. CONCLUSIONS: Preventative future-self avatar mHealth interventions promise to encourage improvements in physical activity and food purchasing behavior in healthy population groups. A full-powered RCT is needed to confirm this preliminary evidence and to investigate how future-self avatars might be modified to reduce attrition, overcome present bias, and promote sustainable behavior change. TRIAL REGISTRATION: ClinicalTrials.gov NCT04505124; https://clinicaltrials.gov/ct2/show/NCT04505124.


Assuntos
Telefone Celular , Aplicativos Móveis , Telemedicina , Adolescente , Adulto , Exercício Físico , Feminino , Comportamentos Relacionados com a Saúde , Humanos , Telemedicina/métodos
3.
Front Nutr ; 9: 855223, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35464035

RESUMO

Postbariatric hypoglycemia (PBH) is an increasingly recognized late metabolic complication of bariatric surgery, characterized by low blood glucose levels 1-3 h after a meal, particularly if the meal contains rapid-acting carbohydrates. PBH can often be effectively managed through appropriate nutritional measures, which remain the cornerstone treatment today. However, their implementation in daily life continues to challenge both patients and health care providers. Emerging digital technologies may allow for more informed and improved decision-making through better access to relevant data to manage glucose levels in PBH. Examples include applications for automated food analysis from meal images, digital receipts of purchased food items or integrated platforms allowing the connection of continuously measured glucose with food and other health-related data. The resulting multi-dimensional data can be processed with artificial intelligence systems to develop prediction algorithms and decision support systems with the aim of improving glucose control, safety, and quality of life of PBH patients. Digital innovations, however, face trade-offs between user burden vs. amount and quality of data. Further challenges to their development are regulatory non-compliance regarding data ownership of the platforms acquiring the required data, as well as user privacy concerns and compliance with regulatory requirements. Through navigating these trade-offs, digital solutions could significantly contribute to improving the management of PBH.

4.
Glycobiology ; 31(1): 44-54, 2021 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-32501471

RESUMO

Siglec-15 is a conserved sialic acid-binding Ig-like lectin expressed on osteoclast progenitors, which plays an important role in osteoclast development and function. It is also expressed by tumor-associated macrophages and by some tumors, where it is thought to contribute to the immunosuppressive microenvironment. It was shown previously that engagement of macrophage-expressed Siglec-15 with tumor cells expressing its ligand, sialyl Tn (sTn), triggered production of TGF-ß. In the present study, we have further investigated the interaction between Siglec-15 and sTn on tumor cells and its functional consequences. Based on binding assays with lung and breast cancer cell lines and glycan-modified cells, we failed to see evidence for recognition of sTn by Siglec-15. However, using a microarray of diverse, structurally defined glycans, we show that Siglec-15 binds with higher avidity to sialylated glycans other than sTn or related antigen sequences. In addition, we were unable to demonstrate enhanced TGF-ß secretion following co-culture of Siglec-15-expressing monocytic cell lines with tumor cells expressing sTn or following Siglec-15 cross-linking with monoclonal antibodies. However, we did observe activation of the SYK/MAPK signaling pathway following antibody cross-linking of Siglec-15 that may modulate the functional activity of macrophages.


Assuntos
Antígenos Glicosídicos Associados a Tumores/metabolismo , Imunoglobulinas/metabolismo , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Ácidos Siálicos/metabolismo , Antígenos Glicosídicos Associados a Tumores/química , Antígenos Glicosídicos Associados a Tumores/genética , Técnicas de Cocultura , Humanos , Imunoglobulinas/química , Imunoglobulinas/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Polissacarídeos/química , Ácidos Siálicos/química , Células Tumorais Cultivadas
5.
Pulm Pharmacol Ther ; 44: 96-105, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28315490

RESUMO

Smoking is an important risk factor for the development of chronic obstructive pulmonary disease (COPD) and viral infections are believed to be major triggers of exacerbations, which periodically lead to a worsening of symptoms. The pro-inflammatory IL-1 family members IL-1α and IL-1ß are increased in COPD patients and might contribute to disease pathology. We investigated whether individual or combined inhibition of these cytokines reduced lung inflammation in cigarette smoke (CS)-exposed and H1N1-infected BALB/c mice. Animals were treated with individual or combined antibodies (Abs) directed against IL-1α, IL-1ß or IL-1R1. Cells in BAL fluid and cytokines/chemokines in lung homogenate were determined. The viral load was investigated. Blocking IL-1α had significant suppressive effects on total cells, neutrophils, and macrophages. Furthermore, it reduced KC levels significantly. Blocking of IL-1ß did not provide significant activity. In primary human bronchial epithelial air-liquid-interface cell cultures infected with H1N1, IL-1α Abs but not IL-1ß Abs reduced levels of TNF-α and IL-6. Concomitant usage of Abs against IL-1α/IL-1ß revealed strong effects in vivo and reduced total cells, neutrophils and macrophages. Additionally, levels of KC, IL-6, TNF-α, MCP-1, MIP-1α and MIP-1ß were significantly reduced and ICAM-1 and MUC5 A/C mRNA expression was attenuated. The viral load decreased significantly upon combined IL-1α/IL-1ß Ab treatment. Blocking the IL-1R1 provided significant effects on total cells, neutrophils and macrophages but was inferior compared to inhibiting both its soluble ligands IL-1α/IL-1ß. Our results suggest that combined inhibition of IL-1α/IL-1ß might be beneficial to reduce CS/H1N1-induced airway inflammation. Moreover, combined targeting of both IL-1α/IL-1ß might be more efficient compared to individual neutralization IL-1α or IL-1ß or inhibition of the IL-1R1.


Assuntos
Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pneumonia/prevenção & controle , Fumar/efeitos adversos , Animais , Anticorpos , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/etiologia , Inflamação/patologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Influenza Humana/complicações , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo , Infecções por Orthomyxoviridae/complicações , Pneumonia/etiologia , Fatores de Risco , Fumaça/efeitos adversos , Nicotiana , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA