Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Commun Biol ; 6(1): 1169, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37973823

RESUMEN

Durotaxis and negative durotaxis are processes in which cell migration is directed by extracellular stiffness. Durotaxis is the tendency of cells to migrate toward stiffer areas, while negative durotaxis occurs when cells migrate toward regions with lower stiffness. The mechanisms of both processes are not yet fully understood. Additionally, the connection between durotaxis and negative durotaxis remains unclear. In this review, we compare the mechanisms underlying durotaxis and negative durotaxis, summarize the basic principles of both, discuss the possible reasons why some cell types exhibit durotaxis while others exhibit negative durotaxis, propose mechanisms of switching between these processes, and emphasize the challenges in the investigation of durotaxis and negative durotaxis.


Asunto(s)
Fenómenos Biomecánicos , Movimiento Celular
3.
Adv Sci (Weinh) ; 10(24): e2300881, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37341301

RESUMEN

The application of immunotherapy in gastrointestinal (GI) cancers remains challenging because of the limited response rate and emerging therapeutic resistance. Combining clinical cohorts, multi-omics study, and functional/molecular experiments, it is found that ANO1 amplification or high-expression predicts poor outcomes and resistance to immunotherapy for GI cancer patients. Knocking-down or inhibiting ANO1 suppresses the growth/metastasis/invasion of multiple GI cancer cell lines, cell-derived xenograft, and patient-derived xenograft models. ANO1 contributes to an immune-suppressive tumor microenvironment and induces acquired resistance to anti-PD-1 immunotherapy, while ANO1 knockdown or inhibition enhances immunotherapeutic effectiveness and overcomes resistance to immunotherapy. Mechanistically, through inhibiting cancer ferroptosis in a PI3K-Akt signaling-dependent manner, ANO1 enhances tumor progression and facilitates cancer-associated fibroblast recruitment by promoting TGF-ß release, thus crippling CD8+ T cell-mediated anti-tumor immunity and generating resistance to immunotherapy. This work highlights ANO1's role in mediating tumor immune microenvironment remodeling and immunotherapeutic resistance, and introduces ANO1 as a promising target for GI cancers' precision treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Ferroptosis , Neoplasias Gastrointestinales , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Fosfatidilinositol 3-Quinasas , Proliferación Celular , Proteínas de Neoplasias/metabolismo , Inmunoterapia , Microambiente Tumoral , Anoctamina-1/metabolismo
4.
Insects ; 14(6)2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37367370

RESUMEN

There is growing concern that massive loss of honey bees can cause serious negative effects on biodiversity and ecosystems. Surveys of colony losses have been performed worldwide to monitor the dynamic changes and health status of honey bee colonies. Here, we present the results of surveys regarding winter colony losses from 21 provinces in China from 2009 to 2021, with a total of 1,744,324 colonies managed by 13,704 beekeepers. The total colony losses were low (9.84%; 95% Confidence Interval (CI): 9.60-10.08%) but varied among years, provinces, and scales of apiaries. As little is known about the overwintering mortality of Apis cerana, in this study, we surveyed and compared the loss rates between Apis mellifera and A. cerana in China. We found colonies of A. mellifera suffered significantly lower losses than A. cerana in China. Larger apiaries resulted in higher losses in A. mellifera, whereas the opposite was observed in A. cerana. Furthermore, we used generalized linear mixed-effects models (GLMMs) to evaluate the effects of potential risk factors on winter colony losses and found that the operation size, species, migration, migration×species interaction, and queen problems were significantly related to the loss rates. New queens can increase their colony overwintering survival. Migratory beekeepers and large operations reported lower loss rates.

5.
Eur J Nucl Med Mol Imaging ; 50(9): 2802-2817, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37099132

RESUMEN

PURPOSE: Claudin 18.2 (CLDN18.2) is a reliable target for lesion detection and could have clinical implications for epithelial tumors, especially digestive system neoplasms. However, there is no predictive technology for accurate whole-body mapping of CLDN18.2 expression in patients. This study assessed the safety of the 124I-18B10(10L) tracer and the feasibility of mapping whole-body CLDN18.2 expression using PET functional imaging. METHODS: The 124I-18B10(10L) probe was synthesized manually, and preclinical experiments including binding affinity and specific targeting ability were conducted after testing in vitro model cells. Patients with pathologically confirmed digestive system neoplasms were enrolled in an ongoing, open-label, single-arm, first-in-human (FiH) phase 0 trial (NCT04883970). 124I-18B10(10L) PET/CT or PET/MR and 18F-FDG PET were performed within one week. RESULTS: 124I-18B10(10L) was successfully constructed with an over 95% radiochemical yield. The results of preclinical experiments showed that it had high stability in saline and high affinity in CLDN18.2 overexpressing cells (Kd = 4.11 nM). Seventeen patients, including 12 with gastric cancers, 4 with pancreatic cancers, and 1 with cholangiocarcinoma were enrolled. 124I-18B10(10L) displayed high uptake in the spleen and liver, and slight uptake in the bone marrow, lung, stomach and pancreas. The tracer uptake SUVmax in tumor lesions ranged from 0.4 to 19.5. Compared with that in lesions that had been treated with CLDN18.2-targeted therapy, 124I-18B10(10L) uptake was significantly higher in lesions that had not. Regional 124I-18B10(10L) PET/MR in two patients showed high tracer uptake in metastatic lymph nodes. CONCLUSIONS: 124I-18B10(10L) was successfully prepared and exhibited a high binding affinity and CLDN18.2 specificity in preclinical studies. As an FiH CLDN18.2 PET tracer, 124I-18B10(10L) was shown to be safe with acceptable dosimetry and to clearly reveal most lesions overexpressing CLDN18.2. TRIAL REGISTRATION: NCT04883970; URL: https://register. CLINICALTRIALS: gov/ . Registered 07 May 2021.


Asunto(s)
Tomografía Computarizada por Tomografía de Emisión de Positrones , Neoplasias Gástricas , Humanos , Radioisótopos de Yodo , Tomografía de Emisión de Positrones/métodos , Fluorodesoxiglucosa F18 , Claudinas
6.
Cancer Res Treat ; 55(2): 626-635, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36550683

RESUMEN

PURPOSE: The human epidermal growth factor receptor 2 (HER2) is an established therapeutic target for various kinds of solid tumors. HER2 amplification occurs in approximately 1% to 6% of colorectal cancer. In this study, we aimed to assess the efficacy and safety of trastuzumab in combination with chemotherapy in HER2-positive metastatic colorectal cancer (mCRC). Materials and Methods: An open-label, phase II trial (Clinicaltrials.gov: NCT03185988) was designed to evaluate the antitumor activity of trastuzumab and chemotherapy in HER2-positive digestive cancers excluding gastric cancer in 2017. Patients from this trial with HER2-positive, KRAS/BRAF wild-type, unresectable mCRC were analyzed in this manuscript. Eligible patients were treated with trastuzumab (8 mg/kg loading dose and then 6 mg/kg every 3 weeks) and irinotecan (120 mg/m2 days 1 and 8 every 3 weeks). The primary endpoint was the objective response rate. RESULTS: Twenty-one HER2-positive mCRC patients were enrolled in this study. Seven patients (33.3%) achieved an objective res-ponse, and 11 patients (52.4%) had stable disease as their best response. The median progression-free survival (PFS) was 4.3 months (95% confidence interval, 2.7 to 5.9). Four of the 21 patients (19.0%) had grade 3 adverse events, including leukopenia, neutropenia, urinary tract infection, and diarrhea. No treatment-related death was reported. Exploratory analyses revealed that high tumor tissue HER2 copy number was associated with better therapeutic response and PFS. Alterations in the mitogen-activated protein kinase pathway, HER2 gene, phosphoinositide 3-kinase/AKT pathway, and cell cycle control genes were potential drivers of trastuzumab resistance in mCRC. CONCLUSION: Trastuzumab combined with chemotherapy is a potentially effective and well-tolerated therapeutic regimen in mCRC with a high HER2 copy number.


Asunto(s)
Neoplasias de la Mama , Neoplasias del Colon , Neoplasias del Recto , Humanos , Femenino , Trastuzumab/efectos adversos , Irinotecán/efectos adversos , Fosfatidilinositol 3-Quinasas , Anticuerpos Monoclonales Humanizados , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Recto/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico
7.
Ther Adv Med Oncol ; 14: 17588359221105022, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35747165

RESUMEN

Background: Combinatorial inhibition of epidermal growth factor receptor (EGFR) and BRAF shows remarkable clinical benefits in patients with BRAF V600E-mutant metastatic colorectal cancer (mCRC). However, the tumor may inevitably develop resistance to the targeted therapy, thereby limiting the response rate and durability. This study aimed to determine the genetic alterations associated with intrinsic and acquired resistance to EGFR/BRAF inhibitors in BRAF V600E-mutant mCRC. Methods: Targeted sequencing of 520 cancer-related genes was performed in tumor tissues and in plasma samples collected from patients with BRAF V600E-mutant mCRC, who were treated with EGFR/BRAF ±â€„MEK inhibitors, before and after the targeted treatment. Clinical benefit was defined as an objective response or a stable disease lasting longer than the median progression-free survival (PFS). Results: In all, 25 patients with BRAF V600E-mutant mCRC were included in this study. Those with RNF43 mutations (n = 8) were more likely to achieve clinical benefit from EGFR/BRAF inhibitors than those with wild-type RNF43 (87.5% versus 37.5%, p = 0.034). Genetic alterations in receptor tyrosine kinase genes (n = 6) were associated with worse PFS (p = 0.005). Among the 23 patients whose disease progressed after the EGFR/BRAF-targeted therapy, at least one acquired resistance-related mutation was detected in 12 patients. Acquired mutations were most frequently observed in the mitogen-activated protein kinase pathway-related genes (n = 9), including KRAS (G12D and Q61H/R), NRAS (Q61L/R/K and amplification), BRAF (amplification), and MEK1 (K57T). MET amplification and PIK3R1 Q579fs mutation emerged in three patients and one patient, respectively, after disease progression. Conclusion: Multiple genetic alterations are associated with clinical benefits and resistance to EGFR/BRAF inhibitors in BRAF V600E-mutant mCRC. Our findings provide novel insights into strategies for overcoming resistance to EGFR/BRAF inhibitors in patients with BRAF V600E-mutant mCRC.

8.
Front Oncol ; 10: 568911, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194656

RESUMEN

BACKGROUND: Early onset colorectal cancer (EO CRC) is a heterogeneous colorectal cancer subtype with obvious hereditary tendencies and increasing incidence. We sought to determine the susceptibility genes and molecular characteristics of EO CRC. METHODS: 330 EO metastatic CRC (mCRC) (≤55 years) and 110 average-onset (AO) mCRC patients (>55 years) were enrolled. Capture-based targeted sequencing was performed on tumor tissue and paired white blood cells using a sequencing panel of 520 genes. The association between molecular alterations and overall survival (OS) was analyzed. RESULTS: Of the 330 EO mCRC patients, 31 carried pathogenic or likely pathogenic germline mutations, with 16 of them diagnosed with lynch syndrome. Fifteen patients had germline mutations in non-mismatch repair genes, including four in MUTHY, three in RAD50, one in TP53, and eight in other genes. Twenty-nine genes were recurrently mutated in EO mCRC, including TP53, APC, KRAS, SMAD4, and BRCA2. The majority of genomic alterations were comparable between EO and AO mCRC. EO mCRC patients were more likely to have a high tumor mutation burden (p < 0.05). RNF43, RBM10, TSC, and BRAF V600E mutations were more commonly observed in EO mCRC, while APC, ASXL1, DNMT3B, and MET genes were more commonly altered in AO patients. At the pathway level, the WNT pathway was the only differentially mutated pathway between EO and AO mCRC (p < 0.0001). The wild-type WNT pathway (p = 0.0017) and mutated TGF-ß pathway (p = 0.023) were associated with unfavorable OS in EO mCRC. CONCLUSIONS: Approximately one in 10 EO mCRC was associated with hereditary tumors. The spectrum of somatic alterations was largely comparable between EO and AO mCRC with several notable differences.

9.
J Cancer Res Clin Oncol ; 146(2): 329-342, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31912229

RESUMEN

PURPOSE: Members of the aaRS (aminoacyl-tRNA synthetase) family are proteins controlling the aminoacylation process, in which YARS (tyrosyl-tRNA synthetase) catalyzes the binding of tyrosine to its cognate tRNA and plays an important role in basic biosynthesis. Several studies have demonstrated the association between YARS mutation and certain developmental abnormalities/diseases, yet YARS's linkage with cancer remains uncategorized. In this study, by combining in silico, in vitro, and in vivo studies, we explored the expressions and functions of YARS in gastric cancer (GC). METHODS: We evaluated YARS's distribution in tumor and paired normal tissues/specimens of GC by referring to large cohort online datasets and patient-derived tissue specimens. YARS-related changes were assessed by phenotypical/molecular experiments and RNA-sequencing analysis in GC cell lines harboring YARS knockdown or overexpression. RESULTS: Both the transcript and protein levels of YARS were evidently higher in gastric cancer tissues than in paired normal tissues. YARS knockdown induced repressed proliferation and invasiveness, as well as enhanced apoptosis in GC cell lines, while abnormally upregulating YARS expression promoted gastric cancer growth in vivo. We inferred based on RNA-sequencing that YARS modulates multiple cancerous signaling pathways and proved through cellular experiments that YARS promoted GC progression, as well as homologous recombination by activating PI3K-Akt signaling. CONCLUSIONS: By revealing the existence of a YARS-PI3K-Akt signaling axis in gastric cancer, we discovered that tRNA synthetase YARS is a novel tumorigenic factor, characterized by its upregulation in tumor-derived specimens, as well as its functions in promoting gastric cancer progression.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Gástricas/enzimología , Tirosina-ARNt Ligasa/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Tirosina-ARNt Ligasa/biosíntesis , Tirosina-ARNt Ligasa/genética , Regulación hacia Arriba
10.
Theranostics ; 9(12): 3485-3500, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281492

RESUMEN

Background: Liver is the most common metastatic site in advanced colorectal cancer. Most patients with colorectal cancer liver metastasis (CRLM) do not benefit from current treatment. Patient-derived xenografts (PDXs) with defined molecular signatures are attractive models for preclinical studies. Methods: Successfully established PDXs were evaluated to elucidate their fidelity of patients' biologic characteristics (pathologic, genetic and protein properties, together with chemosensitivity). The genomic variations of PDXs were analyzed by next-generation sequencing to explore the underlying molecular mechanism of metastasis and potential therapeutic targets. Results: CRLM (N=73) showed a significantly higher successful PDX establishment rate than primary specimens (N=26; 76.7% vs. 57.7%). CRLM PDXs recapitulated the pathologic, genetic and protein properties of parental tumors, as well as chemosensitivity. Frequent altered genes in PDXs showed high consistency compared to patients' genomic alterations and were enriched in MAPK, ErbB, cell cycle, focal adhesion pathways for CRLM PDXs, whereas primary tumor-derived PDXs only exhibited genomic variations involving ErbB and cell cycle. The genetic alterations showed high concordance between paired PDXs from primary and metastatic tissues, except for recurrent gene mutations (ARID1A, CDK8, ETV1, STAT5B and WNK3) and common copy number gains in chromosomes 20q (e.g., SRC/AURKA). Several potential drug targets such as KRAS, HER2, and FGFR2 were validated using corresponding inhibitors. Additionally, PDX models could also be used in screening efficient regimens for patients with no druggable alterations. Conclusion: This study has successfully established and validated a large panel of molecularly annotated platforms from patients with CRLM for preclinical studies.


Asunto(s)
Neoplasias Colorrectales , Xenoinjertos , Neoplasias Hepáticas , Adulto , Anciano , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Perfilación de la Expresión Génica , Xenoinjertos/efectos de los fármacos , Xenoinjertos/metabolismo , Xenoinjertos/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Masculino , Ratones , Persona de Mediana Edad , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Dev Comp Immunol ; 78: 160-168, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987482

RESUMEN

Rhodaneses are known to catalyze in vitro the transfer of a sulfane sulfur atom from thiosulfate to cyanide with concomitant formation of thiocyanate, however, their biological functions remain speculative despite the main role is considered as detoxifying cyanide especially in animal livers. In this study, we characterized a single-domain rhodanese homologue, MnRDH1, from Macrobrachium nipponense. We found MnRDH1 with the highest expression in hemocytes. Upon Aeromonas hydrophila challenge, expression of MnRDH1 was up-regulated in various tissues, including hepatopancreas, gill, intestine and hemocytes. RNAi knockdown of MnRDH1 led to rapid increases of malondialdehyde content, which reveals that MnRDH1 deficiency causes oxidative stress. The expression of MnRDH1 in hepatopancreas was significantly increased in response to the doxorubicin-induced oxidative stress, indicating the gene is oxidative stress inducible. We transformed E. coli with MnRDH1 and the mutant MnRDH1C75A, and found significant rhodanese activity of the recombinant protein of MnRDH1 in vitro, but detected no enzyme activity of the mutant MnRDH1C75A. When under the oxidative insult by H2O2, the MnRDH1 transformed E. coli had significantly enhanced survival rates compared to those bacteria transformed with MnRDH1C75A. In conclusion, our study demonstrates that rhodanese in M. nipponense confers oxidative stress tolerance, and thus renders an evidence for the notion that rhodanese family genes act a critical role in antioxidant defenses.


Asunto(s)
Aeromonas hydrophila/inmunología , Proteínas de Artrópodos/genética , Infecciones por Bacterias Gramnegativas/inmunología , Hemocitos/fisiología , Palaemonidae/fisiología , Dominios Proteicos/genética , Tiosulfato Azufretransferasa/genética , Animales , Proteínas de Artrópodos/metabolismo , Células Cultivadas , Clonación Molecular , Doxorrubicina/metabolismo , Hepatopáncreas/fisiología , Homeostasis , Mutación/genética , Oxidación-Reducción , Estrés Oxidativo/genética , ARN Interferente Pequeño/genética , Homología de Secuencia de Aminoácido , Tiosulfato Azufretransferasa/metabolismo
12.
Fish Shellfish Immunol ; 70: 327-334, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28882803

RESUMEN

In Macrobrachium nipponense, the rhodanese homologue 2 (MnRDH2) gene codes for a single rhodanese domain protein. Considering the lack of information on the biological role of the ubiquitous rhodaneses in invertebrate, we examined the functions of MnRDH2 using both in silico and in vitro approaches. Quantitative PCR analysis of different tissues indicated that expression of MnRDH2 was enriched in hepatopancreas, in which bacterial challenge by Aeromonas hydrophila induced MnRDH2 expression. Knocking down MnRDH2 by RNA interference caused significant accumulations of reactive oxygen species and malondialdehyde (MDA). Using Escherichia coli (DE3), we expressed MnRDH2 and the mutant MnRDH2C78A, in which the predicted catalytic cysteine was mutated to alanine, and found significant rodanese activity of the recombinant MnRDH2 in vitro, but not for the mutant rMnRDH2C78A. We observed that rMnRDH2 was able to significantly increase tolerance of the host bacteria to oxidative stressor phenazine methosulfate. These results suggest that MnRDH2 might have the potential to buffer general levels of oxidants via regulation of redox reactions. In conclusion, our study begins to hint a possible biological functionality of MnRDH2 as a redox switch to activate defensive activities against oxidative damage, which helps host in maintaining the cellular redox balance. These characteristics will facilitate future investigations into the physiological functions for invertebrate rhodanese family genes.


Asunto(s)
Antioxidantes/metabolismo , Inmunidad Innata , Palaemonidae/genética , Palaemonidae/inmunología , Tiosulfato Azufretransferasa/genética , Tiosulfato Azufretransferasa/inmunología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Secuencia de Bases , Escherichia coli/fisiología , Especificidad de Órganos , Alineación de Secuencia
13.
J Photochem Photobiol B ; 133: 65-72, 2014 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-24705372

RESUMEN

The redox properties of cytochrome c (Cyt c), hemoglobin (Hb) and myoglobin (Mb) were studied based on electrostatic interactions between Thioglycolic acid (TGA) capped CdSe/ZnS quantum dots (QDs) and proteins. Results indicated that only Cyt c quenched the fluorescence of the QDs at pH>8.0. Under the optimized conditions, a significant fluorescence recovery of the QDs' system was observed when the reduced form of Cyt c incubated with TGA capped QDs, however, the reduced state of Hb and Mb resulted in a more fluorescence quenching on the same size of QDs. Interestingly, the fluorescence changes of QDs-proteins could be switched by modulating the redox potentials of proteins-attached QDs. Moreover, only the oxidized Cyt c form was reduced by the generated O2(-) that significantly enhanced the fluorescence of the QDs' system, which was also demonstrated by fluorescence imaging in HeLa cells.


Asunto(s)
Citocromos c/química , Hemoglobinas/química , Mioglobina/química , Puntos Cuánticos/química , Compuestos de Cadmio/química , Citocromos c/metabolismo , Células HeLa , Hemoglobinas/metabolismo , Humanos , Microscopía Confocal , Mioglobina/metabolismo , Oxidación-Reducción , Compuestos de Selenio/química , Espectrometría de Fluorescencia , Sulfuros/química , Tioglicolatos/química , Compuestos de Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA