Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 11: 620688, 2021.
Article in English | MEDLINE | ID: mdl-33833986

ABSTRACT

Background: Colorectal cancer, the fourth leading cause of cancer mortality, is prone to metastasis, especially to the liver. The pre-metastatic microenvironment comprising various resident stromal cells and immune cells is essential for metastasis. However, how the dynamic evolution of immune components facilitates pre-metastatic niche formation remains unclear. Methods: Utilizing RNA-seq data from our orthotopic colorectal cancer mouse model, we applied single sample gene set enrichment analysis and Cell type Identification By Estimating Relative Subsets Of RNA Transcripts to investigate the tumor microenvironment landscape of pre-metastatic liver, and define the exact role of myeloid-derived suppressor cells (MDSCs) acting in the regulation of infiltrating immune cells and gene pathways activation. Flow cytometry analysis was conducted to quantify the MDSCs levels in human and mice samples. Results: In the current work, based on the high-throughput transcriptome data, we depicted the immune cell infiltration pattern of pre-metastatic liver and highlighted MDSCs as the dominant altered cell type. Notably, flow cytometry analysis showed that high frequencies of MDSCs, was detected in the pre-metastatic liver of orthotopic colorectal cancer tumor-bearing mice, and in the peripheral blood of patients with stage I-III colorectal cancer. MDSCs accumulation in the liver drove immunosuppressive factors secretion and immune checkpoint score upregulation, consequently shaping the pre-metastatic niche with sustained immune suppression. Metabolic reprogramming such as upregulated glycolysis/gluconeogenesis and HIF-1 signaling pathways in the primary tumor was also demonstrated to correlate with MDSCs infiltration in the pre-metastatic liver. Some chemokines were identified as a potential mechanism for MDSCs recruitment. Conclusion: Collectively, our study elucidates the alterations of MDSCs during pre-metastatic niche transformation, and illuminates the latent biological mechanism by which primary tumors impact MDSC aggregation in the targeted liver.

2.
Theranostics ; 10(15): 7002-7014, 2020.
Article in English | MEDLINE | ID: mdl-32550918

ABSTRACT

Immune-checkpoint blockades (ICBs) have been routinely implemented to treat metastatic urothelial cancer (mUC), whereas robust biomarkers are urgently warranted. Herein, we explored latent promising biomarkers based on 348 pretreatment mUC samples from IMvigor210. Methods: The genome, transcriptome, immunome, and metabolome were systemically analyzed using the external TCGA dataset for validation. Kaplan-Meier and ROC curve analyses were performed to estimate the predictive capacity of M1-macrophage infiltration. Chi-square/Spearman/Mann Whitney U test are used to determine its correlation to genetic, biochemical, and clinicopathological parameters. Results: M1 frequency is a robust biomarker for predicting the prognosis and response to ICBs, which is non-inferior to tumor mutation burden (TMB) or tumor neoantigen burden (TNB), and exceeds CD8 T cells, T cell inflamed gene expression profile (GEP), and PD-L1 expression. Moreover, M1 infiltration is associated with immune phenotypes (AUC = 0.785) and is negatively correlated with immune exclusion. Additionally, transcriptomic analysis showed immune activation in the high-M1 subgroup, whereas it showed steroid and drug metabolism reprograming in the M1-deficient subset, which characterized the limited sensitivity to ICB therapy. Notably, investigation of the corresponding intrinsic genomic profiles highlighted the significance of TP53 and FGFR alterations. Conclusions: M1 infiltration is a robust biomarker for immunotherapeutic response and immunophenotype determination in an mUC setting. Innate immunity activation involving macrophage polarization remodeling and anti-FGFR mutations may be promising strategies for synergy with anti-PD-L1 treatments and may help prolong the clinical survival of patients with mUC.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/immunology , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Macrophages/immunology , Transcriptome/drug effects , Urinary Bladder Neoplasms/pathology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Databases, Genetic/statistics & numerical data , Humans , Immunotherapy/methods , Macrophages/drug effects , Macrophages/metabolism , Mutation , Prognosis , Survival Rate , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
3.
ACS Nano ; 14(7): 7940-7958, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32413258

ABSTRACT

Gold nanoparticles (AuNPs) are a promising nanomaterial due to their drug-delivery properties and inherent anti-neoplastic activity. Here, we focused on the anti-neoplastic effects of an improved targeting polymer and folic acid-modified gold nanoparticles (AuNPP-FA) without therapeutic drugs. AuNPP-FA inhibited tumor proliferation both in vitro and in vivo, and tumor metastasis was controlled in vivo. We also found that, in addition to inhibiting tumor angiogenesis, AuNPP-FA normalized tumor vasculature by increasing pericyte coverage and strengthening tight junctions by upregulating VE-cadherin (VE-cad) levels on endothelial cells. This decreased vascular permeability, improved vascular perfusion, and alleviated tissue hypoxia. The immunotherapeutic response was enhanced due to the increased infiltration of CD3+CD8+ T lymphocytes. AuNPP-FA increased the expression and secretion of semaphorin 3A (SEMA3A) in cancer cells to further inhibit Smad2/3 signaling in human umbilical vein endothelial cells (HUVECs). This normalized tumor vasculature and inhibited metastasis. In conclusion, AuNPP-FA normalized tumor vasculature; therefore, AuNPP-FA has great potential for future clinical applications.


Subject(s)
Metal Nanoparticles , Neoplasms , Smad2 Protein , Smad3 Protein , Cell Line, Tumor , Gold , Humans , Neovascularization, Pathologic/drug therapy , Signal Transduction
4.
Histol Histopathol ; 33(7): 681-690, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29319126

ABSTRACT

Proteinuria (albuminuria) is an important cause of aggravating tubulointerstitial injury. Previous studies have shown that autophagy activation can alleviate renal tubular epithelial cell injury caused by urinary protein, but the mechanism is not clear. Here, we investigated the role of clearance of damaged mitochondria in this protective effect. We found that albumin overload induces a significant increase in turnover of LC3-II and decrease in p62 protein level in renal proximal tubular (HK-2) cells in vitro. Albumin overload also induces an increase in mitochondrial damage. ALC, a mitochondrial torpent, alleviates mitochondrial damage induced by albumin overload and also decreases autophagy, while mitochondrial damage revulsant CCCP further increases autophagy. Furthermore, pretreatment of HK-2 cells with rapamycin reduced the amount of damaged mitochondria and the level of apoptosis induced by albumin overload. In contrast, blocking autophagy with chloroquine exerted an opposite effect. Taken together, our results indicated autophagy activation promotes removal of damaged mitochondria and protects against renal tubular injury caused by albumin overload. This further confirms previous research that autophagy activation is an adaptive response in renal tubular epithelial cells after urinary protein overload.


Subject(s)
Acute Kidney Injury/physiopathology , Albuminuria/physiopathology , Autophagy/physiology , Kidney Tubules, Proximal/physiopathology , Mitochondria/pathology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Albumins/toxicity , Albuminuria/pathology , Cell Line , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology
5.
Histol Histopathol ; 33(3): 237-246, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28696497

ABSTRACT

Hypoxia is a kind of common pathological condition existing in various diseases such as sleep apnea syndrome, myocardial infarction and stroke, which can precipitate the onset of diseases through inducing cell apoptosis. Ca²âº is the ubiquitous message in cell. Given the crucial role of Ca2²âº in physiology, intracellular Ca²âº overload is a significant regulator of apoptosis. Numerous experiments show that hypoxia may cause changes of multiple cellular Ca²âº channels, for instance, Na⁺/ Ca²âº Exchanger (NCX), L-type voltage dependent Ca²âº channel (L-VDCC), inositol triphosphate receptors (IP3R) and so on, which contribute to intracellular Ca²âº overload, thus eventually triggering cell apoptosis. However, the mechanisms underlying different Ca²âº channels involved in hypoxic apoptosis are complex. For example, chronic hypoxia or acute hypoxia may select different Ca²âº channels to influence cell apoptosis. In addition, intracellular Ca²âº overload may initiate different apoptotic pathways due to hypoxic duration. Furthermore, different locations in the cell of specific Ca²âº channels activated by hypoxia will determine different apoptosis signaling pathways. Moreover, activation of different Ca²âº channel isoforms will result in different outcomes of the cell under hypoxia. Hence, we aim to highlight the potential mechanisms of the main Ca²âº channels in regulation of apoptosis under hypoxic stress.


Subject(s)
Apoptosis/physiology , Calcium Channels/metabolism , Calcium/metabolism , Cell Hypoxia/physiology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...