Primary liver cancer (PLC) is one type of cancer with high incidence rate and high mortality rate in the worldwide. Systemic therapy is the major treatment for PLC, including surgical resection, immunotherapy and targeted therapy. However, mainly due to the heterogeneity of tumors, responses to the above drug therapy differ from person to person, indicating the urgent needs for personalized treatment for PLC. Organoids are 3D models derived from adult liver tissues or pluripotent stem cells. Based on the ability to recapitulate the genetic and functional features of in vivo tissues, organoids have assisted biomedical research to make tremendous progress in understanding disease origin, progression and treatment strategies since their invention and application. In liver cancer research, liver organoids contribute greatly to reflecting the heterogeneity of liver cancer and restoring tumor microenvironment (TME) by co-organizing tumor vasculature and stromal components in vitro . Therefore, they provide a promising platform for further investigation into the biology of liver cancer, drug screening and precision medicine for PLC. In this review, we discuss the recent advances of liver organoids in liver cancer, in terms of generation methods, application in precision medicine and TME modeling.
Exposure to particulate matters with diameters below 2.5 µm (PM2.5) is considered a major risk factor for cardiovascular diseases (CVDs). The closest associations between PM2.5 and CVDs have been observed in hyperbetalipoproteinemia cases, although the detailed underpinning mechanism remains undefined. In this work, hyperlipidemic mice and H9C2 cells were used to examine the effects of PM2.5 on myocardial injury and their underlying mechanisms. The results revealed that PM2.5 exposure caused severe myocardial damage in the high-fat mouse model. Oxidative stress and pyroptosis were also observed along with myocardial injury. After inhibiting pyroptosis with disulfiram (DSF), the level of pyroptosis was effectively reduced as well as myocardial injury, suggesting that PM2.5 induced the pyroptosis pathway and further caused myocardial injury and cell death. Afterwards, by suppressing PM2.5-induced oxidative stress with N-acetyl-L-cysteine (NAC), myocardial injury was markedly ameliorated, and the upregulation of pyroptosis markers was reversed, which indicated that PM2.5-pyroptosis was also improved. Taken together, this study revealed that PM2.5 induce myocardial injury through the ROS-pyroptosis signaling pathway in hyperlipidemia mice models, providing a potential approach for clinical interventions.
Mouse islets are widely used in diabetes research. Thus an adequate quality, quantity, and purity of islets are needed for high-quality investigations. We performed a combination of filtration and density gradient separation and optimized many steps in the islet isolation procedure, including perfusion, digestion, and purification. Our results show that an increased quality, quantity, and purity of isolated islets can be achieved using these modifications. Moreover, this method can guarantee maximal recovery and purity of the isolated islets and is easy to perform with practice.
Objective
To observe the effect of immune protection of heart allograft in mice induced by immune escape mechanism of trichinella spiralis,and explore new and potential ways against acute rejection in clinic.
Methods
The heterotopic cardiac transplantation models were divided into three groups: rejection group, treatment group,and isograft group.Mean survival time was measured.Twenty- eight days before transplantation, recipients(C57BL/6)of rejection group were orally infected with 300 muscle larvae,and donors were untreated.Hearts in each group were harvested on the day 7 post- transplantation for pathological observation.Proportion of CD4+,CD8+ T cells and regulatory T cells(Tregs)was detected by flow cytometry.
Results
As compared with rejection group(11 days),survival of allografts was significantly prolonged in treatment group(24 days)(P<0.01),alleviated pathologic signd were observed,and proportion of CD8+ T cells(36.6%)was reduced(48.8%, P<0.01), but there was no significant difference in CD4+ T cells between treatment group and rejection group.Meanwhile, proportion of Tregs in treatment group(16.9%)was significantly higher than in rejection group(10.8%,P<0.05).
Conclusion
Trichinella spiralis infection lowered the proportion of CD8+ T cells,augmented Tregs frequency,and alleviated acute rejection of heterotopic cardiac transplantation in mice, which immunologically protected mice heart allograft.
Key words:
Trichinella spiralis; Immune escape; Cardiac transplantation
Background and Aims: Lymph node metastasis is a significant risk factor for patients with cholangiocarcinoma, but the mechanisms underlying cholangiocarcinoma colonization in the lymph node microenvironment remain unclear. We aimed to determine whether metabolic reprogramming fueled the adaptation and remodeling of cholangiocarcinoma cells to the lymph node microenvironment. Approach and Results: Here, we applied single-cell RNA sequencing of primary tumor lesions and paired lymph node metastases from patients with cholangiocarcinoma and revealed significantly reduced intertumor heterogeneity and syntropic lipid metabolic reprogramming of cholangiocarcinoma after metastasis to lymph nodes, which was verified by pan-cancer single-cell RNA sequencing analysis, highlighting the essential role of lipid metabolism in tumor colonization in lymph nodes. Metabolomics and in vivo CRISPR/Cas9 screening identified PPARγ as a crucial regulator in fueling cholangiocarcinoma colonization in lymph nodes through the oleic acid-PPARγ-fatty acid–binding protein 4 positive feedback loop by upregulating fatty acid uptake and oxidation. Patient-derived organoids and animal models have demonstrated that blocking this loop impairs cholangiocarcinoma proliferation and colonization in the lymph node microenvironment and is superior to systemic inhibition of fatty acid oxidation. PPARγ-regulated fatty acid metabolic reprogramming in cholangiocarcinoma also contributes to the immune-suppressive niche in lymph node metastases by producing kynurenine and was found to be associated with tumor relapse, immune-suppressive lymph node microenvironment, and poor immune checkpoint blockade response. Conclusions: Our results reveal the role of the oleic acid-PPARγ-fatty acid–binding protein 4 loop in fueling cholangiocarcinoma colonization in lymph nodes and demonstrate that PPARγ-regulated lipid metabolic reprogramming is a promising therapeutic target for relieving cholangiocarcinoma lymph node metastasis burden and reducing further progression.
In 2006, the first Chinese Tahyna virus isolate (XJ0625) was obtained in Xinjiang province and human infection were found in the same region. In this study, cell culture, animal experiments, electron microscopy, immunofluorescence assay and cross neutralization tests were performed to see the cell susceptibility, animal pathogenicity, morphology and antigenic and other biological characteristics of XJ0625. In addition, molecular biology software was used to analyze the characteristics of molecular evolution. The results showed that BHK-21 cell line was susceptible to XJ0625 and the virus was lethal to suckling mice when injected by intracranial ways. Similar to the other Bunyavirus, Tahyna virus is spherical enveloped virus under electron microscopy. XJ0625 infected cells showed strong fluorescent signal and could be neutralized by immune asities fluid with immnity to protype Tahyna virus Bardos 92. The sequence of the S and M segments showed 91.8% and 81.9% homology with Bardos 92.
Abstract In 2006, Tahyna virus was isolated from Culex spp. mosquitoes collected in Xinjiang, People's Republic of China. In 2007, to determine whether this virus was infecting humans, we tested serum from febrile patients. We found immunoglobulin (Ig) M and IgG against the virus, which suggests human infection in this region.
Objective
To study the regulation of high expression of suppressor of cytokine signaling 1 (SOCS1) molecule in dendritic cells (DC), and to verify the effect of DC-SOCS1 on T cell function and allografts immune protective effects in vitro and in vivo.
Methods
DC were modified with high affinity replication-defective adenoviral vector Ad5F35 over-expressing SOCS1. The phenotype of transduced mature DC and the function were analyzed, and the mechanism of inducing donor-specific CD4+ T cells hyporesponsiveness was explored. The mouse allogenic islet transplantation model was established, and 24 h prior to transplantation each recipient mouse was given DC cells, DC-SOCS1 cells or equal amount of phosphate buffer (PBS), named DC group, DC-SOCS1 group and control group respectively. The survival of islet grafts of transplanted recipients was observed, and on the day 7 and the rejection day post transplantation, the islet graft glucose tolerance, pathological changes, the percentage of T cell subsets in the spleen and draining lymph nodes as well as related cytokines were observed. The possible associated mechanisms of SOCS1 gene-modified mature DC mitigating immune rejection and inducing transplant tolerance in vivo were explored.
Results
Genetic modified Ad5F35 dramatically increased the transduction efficiency of DC compared to regular Ad. The experimental data demonstrated there was a tendency that DC overexpressing SOCS1 can induce DC low down the express of MHC and costimulatory molecules, potentiate the ability of DC-SOCS1 to induce donor specific T cell anergy in mixed lymphocytes reaction (MLR) assay. DC-SOCS1 significantly prolonged the survival time of the islet allograft with a median survival time of (37.00±2.19) days, which is significantly longer than control [(20.00±0.55) d, P=0.008] and DC group [(10.00±0.45) d, P=0.001]. Flow cytometric analysis revealed that the DC-SOCS1 dramatically reduced the population of Th1 and Tc1 [(9.92±1.57)%, P=0.027], CD4 [(17.77±2.80)%, P=0.007] and CD8 [(14.92±2.18)%, P=0.004] in spleen lymphocytes compared to control group and DC group on day 7, and a reduction of CD4 [(26.00±2.02)%, P=0.034] was observed in draining lymph nodes compared to DC group.
Conclusion
SOCS1 genetic modification efficiently confers mDC with low express of MHC and costimulatory molecules, and allows them to induce T cell hyporesponsiveness in vitro. The DC-SOCS1 pretreated recipients exhibit immune tolerance and prolong islet allograft survival in vivo.
Key words:
Dendritic cells; Gene transduction; Suppressor of cytokine signaling 1; Islet transplantation; Immune tolerance
Members of TLR/IL-1R (TIR) superfamily mediate maturation of dendritic cells (DCs) and initiate immune response in transplanted organs. In this study, we tested the hypothesis that TIR8, also known as single Ig IL-1 receptor-related molecule (SIGIRR), refrain DCs from maturation and induce immune tolerance of transplant organ. DCs were transduced with the recombinant adenovirus Ad5F35 to highly express SIGIRR (DC-SIGIRR), then injected to murine recipient before islet transplantation. It revealed that DCs transduced with SIGIRR had low expression of major histocompatibility and costimulatory molecules along with strong phagocytic ability in vitro assay. The data demonstrated that recipients treated with DC-SIGIRR had satisfying islet allograft function and long survival times, with an increase of Treg and reduction of Th17 in both spleen and draining lymph nodes in vivo. Therefore, genetic modification of SIGIRR inhibits DC activation and maturation, affects differentiation of T cell subsets, protects allograft biological function, and prolongs graft survival.