Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Blood Transfus ; 21(2): 146-156, 2023 03.
Article in English | MEDLINE | ID: mdl-35175191

ABSTRACT

BACKGROUND: Platelets are anucleated blood cells and contain various RNA species. We investigated the changes in the whole transcriptome expression profile of platelet concentrates (PC) during storage to explore biological functions and biomarkers in platelet storage damage. MATERIALS AND METHODS: Platelets were collected by apheresis from eight healthy blood donors and stored from day 0 to day 4. Platelet phenotyping and function analysis were used to detect platelet activity during storage. RNA-sequencing was used to detect changes in expression of mRNA, lncRNA and circRNA in the PC during storage. Gene ontology and KEGG analyses were applied to predict the functional distribution of differential expression of mRNA. Gene set enrichment analysis was used to analyze the differential levels of gene pathways. Finally, polymerase chain reaction (PCR) analysis was performed to verify the expression of three mRNA (POLE2, DCUN1D4, DAD1). RESULTS: In total, 10,767 mRNA, 2,923 lncRNA and 68,550 circRNA were detected in the PC by RNA-sequencing. The expression levels of 222 mRNA changed significantly from day 0 to day 4 of storage: 58 increased continuously and 145 decreased continuously. Differentially expressed mRNA may be involved in physiological processes such as platelet activation, platelet aggregation, endocytosis, and apoptosis. Expression levels of 1,413 lncRNA were obvious. The levels of 42 species increased and the levels of 28 species decreased. The expression levels of 198 species of circRNA changed significantly, with those of 13 species changing continuously. The differential levels of expression of DAD1, DCUN1D4 and POLE1 mRNA, shown by RNA sequencing, were validated by PCR assay. DISCUSSION: Changes in mRNA, lncRNA and circRNA during platelet storage may be closely related to platelet apoptosis and physiological functions in the platelet storage lesion. The expression levels of DAD1, DCUN1D4 and POLE1 could be biomarkers to monitor platelet status in PC bags.


Subject(s)
RNA, Circular , RNA, Long Noncoding , Humans , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Blood Preservation , Blood Platelets/metabolism , Biomarkers/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression Profiling
2.
FASEB J ; 36(4): e22244, 2022 04.
Article in English | MEDLINE | ID: mdl-35262964

ABSTRACT

Cancer immunotherapy has achieved impressive therapeutic effects in many cancers, while only a small subset of patients benefit from it and some patients even have experienced severe toxicity. It is urgent to develop a feasible large-cohort humanized mouse model to evaluate the pre-clinical efficacy and safety of cancer immunotherapy. Furthermore, developing potentially effective combination therapy between cancer immunotherapy and other therapies also needs humanized mouse model to adequately mimic clinical actual setting. Herein, we established a humanized mouse model engrafted with less human CD34+ HSCs than ever before and then evaluated reconstitution efficiency and the profiles of human immune cells in this humanized mouse model. Also, this humanized mouse model was used to evaluate the preclinical efficacy and safety of cancer immunotherapy. For each batch of CD34+ HSCs humanized mouse model, a relatively-large cohort with over 25% human CD45+ cells in peripheral blood was established. This humanized mouse model could efficiently reconstitute human innate and adaptive immune cells. This humanized mouse model supported patient-derived xenograft tumor growth and tumor infiltration of PD-1+ human T cells. Furthermore, therapeutic efficacy, re-activation of tumor-infiltrated T cells, and side effects of checkpoint blockade therapy could be monitored in this humanized mouse model. Human T cells from this humanized mouse model were successfully engineered with CD19-CAR. CD19 CAR-T cells could effectively deplete B cells and suppress tumor growth of acute lymphoblastic leukemia in vivo in this humanized mouse model. This humanized mouse model also could be used to demonstrate the efficacy of bispecific antibodies, such as anti-CD19/CD3. Overall, our work provides a feasible large-cohort humanized mouse model for evaluating a variety of cancer immunotherapy approaches including checkpoint inhibitors, adoptive cell therapy, and bispecific antibody therapy, and demonstrates that human T cells from this humanized mouse model possess anti-tumor activities in vitro and in vivo.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/pharmacology , Antigens, CD34 , Disease Models, Animal , Hematopoietic Stem Cells , Humans , Immunotherapy , Mice , Xenograft Model Antitumor Assays
3.
Platelets ; 33(3): 462-470, 2022 Apr 03.
Article in English | MEDLINE | ID: mdl-34223794

ABSTRACT

In the clinic, the supply of platelets is frequently insufficient to meet transfusion needs. To address this issue, many scientists have established the derivation of functional platelets from CD34+ cells or human pluripotent stem cells (PSCs). However, the yield of platelets is still far below what is required. Here we found that the plant hormone abscisic acid (ABA) could increase the generation of megakaryocytes (MKs) and platelets from human induced PSCs (hiPSCs). During platelet derivation, ABA treatment promoted the generation of CD34+/CD45+ HPCs and CD41+ MKs on day 14 and then increased CD41+/CD42b+ MKs and platelets on day 19. Moreover, we found ABA-mediated activation of Akt and ERK1/2 signal pathway through receptors LANCL2 and GRP78 in a PKA-dependent manner on CD34+/CD45+ cells. In conclusion, our data suggest that ABA treatment can promote CD34+/CD45+ HPC proliferation and CD41+ MK differentiation.


Subject(s)
Abscisic Acid/therapeutic use , Induced Pluripotent Stem Cells/metabolism , Megakaryocytes/drug effects , Plant Growth Regulators/metabolism , Abscisic Acid/pharmacology , Cell Differentiation , Humans
4.
Blood Sci ; 2(3): 71-75, 2020 Jul.
Article in English | MEDLINE | ID: mdl-35402827

ABSTRACT

The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in China led to a public health emergency of international concern, putting all health organizations on high alert in the beginning of 2020. Corona virus disease 2019 (COVID-19) is highly infectious and has resulted in thousands of deaths which exceeded that of the SARS coronavirus (SARS-CoV) outbreak back in 2002 and 2003 in China. Besides, the number of diagnosed patients, patients who are suspected to have contracted the disease, and deaths are increasing worldwide. Unfortunately, effective drugs and vaccines to combat SARS-CoV-2 are still lacking. Convalescent plasma, a seemingly successful treatment for COVID-19 patients, proved to be of huge value in terms of saving severely ill patients. This review introduces the reported effects, potential mechanisms, and future uncertainties of convalescent plasma therapy in the treatment of COVID-19 patients, in the hopes that it will provide useful information for relevant physicians and researchers.

5.
Acta Haematol ; 143(1): 9-18, 2020.
Article in English | MEDLINE | ID: mdl-31167182

ABSTRACT

BACKGROUND: Aggressive growth of primitive and immature cells in the bone marrow results in reductions in megakaryocyte and platelet (PLT) counts, leading to thrombocytopenia in acute myeloid leukemia (AML). However, not all AML patients show thrombocytopenia at the time of diagnosis, and the association of PLT count with patient survival is largely unknown. METHODS: A retrospective study was performed to determine PLT counts at diagnosis in the peripheral blood in 291 newly diagnosed AML patients and assess the association of PLT counts with the overall survival (OS) and disease-free survival (DFS) of these patients. RESULTS: Low PLT counts (≤40 × 109/L) were associated with better outcomes for the whole cohort (5-year OS, 55.1 ± 3.8 vs. 35.3 ± 3.5%, p < 0.001; 5-year DFS, 49.1 ± 3.8 vs. 25.7 ± 4.0%, p < 0.001) and intermediate-risk patients (5-year OS, 64.5 ± 5.4 vs. 41.0 ± 4.8%, p < 0.001; 5-year DFS, 60.8 ± 5.6 vs. 28.6 ± 5.6%, p < 0.001). Moreover, low PLT counts were related to deeper molecular remission. Low PLT counts correlated with better survival of intermediate-risk AML patients treated with chemotherapy only. Allogeneic hematopoietic stem cell transplantation attenuated the negative impact of high PLT counts on the survival of intermediate-risk patients. Furthermore, univariate and multivariate analyses demonstrated that PLT count at diagnosis was an independent prognostic factor for intermediate-risk AML. CONCLUSION: PLT count at diagnosis predicts survival for patients with intermediate-risk AML.


Subject(s)
Blood Platelets/cytology , Leukemia, Myeloid, Acute/diagnosis , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Platelet Count , Prognosis , Proportional Hazards Models , Remission Induction , Retrospective Studies , Risk Factors , Young Adult
6.
Cytometry B Clin Cytom ; 98(2): 193-202, 2020 03.
Article in English | MEDLINE | ID: mdl-31571381

ABSTRACT

BACKGROUND: Several studies have used CD157 in white blood cells with or without proaerolysin (fluorescein-labeled proaerolysin [FLAER])-based flow cytometry assays in the diagnosis of paroxysmal nocturnal hemoglobinuria (PNH). METHODS: We designed a seven-color CD marker panel comprising FLAER, CD15, CD64, CD24, CD14, CD157, and CD45 to verify CD157's clinical applicability and diagnostic performance in a clinical setting. RESULTS: A total of 356 samples were tested. These included 43 PNH-positive samples and 313 PNH-negative samples. PNH clones confirmed by the CD157/FLAER combination were almost identical in size to the clones detected by the CD24/CD14/FLAER combination, and the accuracy of the CD157/FLAER combination was 100% in granulocytes and 99.7% in monocytes. Substitution of FLAER with CD157 resulted in 1.9% and 3.5% false-positives in granulocytes and monocytes, respectively. The accuracy was 98.3% and 96.9% in granulocytes and monocytes, respectively. Moreover, the loss of CD157 expression in granulocytes and monocytes was commonly observed in non-PNH patients. Some monocytes in non-PNH patients had weak expression of CD14 but normal expression of FLAER. In this study, PNH clones in granulocytes were always lower than those in matched monocytes. CONCLUSIONS: We performed the first prospective exploration of the clinical usefulness of FLAER and CD157 in simultaneously recognizing PNH clones in granulocytes and monocytes and verified the applicability of CD157 in substitute for both CD14 and CD24. In the conditions where FLAER is not available, substitution of FLAER with CD157 is acceptable for the identification of PNH clones under the premise of giving full attention to the potential for false-positives.


Subject(s)
ADP-ribosyl Cyclase/blood , Antigens, CD/blood , Flow Cytometry/methods , Hemoglobinuria, Paroxysmal/diagnosis , ADP-ribosyl Cyclase/analysis , Antigens, CD/analysis , Biomarkers/analysis , Biomarkers/blood , Blood Cells/metabolism , Blood Cells/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Diagnosis, Differential , Feasibility Studies , Flow Cytometry/instrumentation , GPI-Linked Proteins/analysis , GPI-Linked Proteins/blood , Hemoglobinuria, Paroxysmal/blood , Humans , Immunophenotyping/instrumentation , Immunophenotyping/methods , Monocytes/metabolism , Monocytes/pathology , Neutrophils/metabolism , Neutrophils/pathology , Predictive Value of Tests , Prospective Studies
7.
Cell Death Dis ; 10(4): 271, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894510

ABSTRACT

Mechanisms underlying the generation of induced pluripotent stem cells (iPSC) and keeping iPSC stability remain to be further defined. Accumulated evidences showed that iPSC reprogramming may be controlled by the cell-division-rate-dependent model. Here we reported effects of absence of mouse p27 or p18 on iPSC generation efficiency and genomic stability. Expression levels of cyclin-dependent kinases inhibitors (CDKIs), p21, p27, and p18 decreased during iPSC reprogramming. Like p21 loss, p27 or p18 deficiency significantly promoted efficiency of iPSC generation, whereas ectopic expression of p27, p18, or treatment with CDK2 or CDK4 inhibitors repressed the reprogramming rate, suggesting that CDKIs-regulated iPSC reprogramming is directly related with their functions as CDK inhibitors. However, unlike p21 deletion, absence of p27 or p18 did not increase DNA damage or chromosomal aberrations during iPSC reprogramming and at iPSC stage. Our data not only support that cell cycle regulation is critical for iPSC reprogramming, but also reveal the distinction of CDKIs in somatic cell reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Genomic Instability/genetics , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Division/genetics , Chromosome Aberrations , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Transduction, Genetic
8.
Stem Cell Res Ther ; 9(1): 163, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29907164

ABSTRACT

BACKGROUND: Generation of induced pluripotent stem cells (iPSCs) from human peripheral blood provides a convenient and low-invasive way to obtain patient-specific iPSCs. The episomal vector is one of the best approaches for reprogramming somatic cells to pluripotent status because of its simplicity and affordability. However, the efficiency of episomal vector reprogramming of adult peripheral blood cells is relatively low compared with cord blood and bone marrow cells. METHODS: In the present study, integration-free human iPSCs derived from peripheral blood were established via episomal technology. We optimized mononuclear cell isolation and cultivation, episomal vector promoters, and a combination of transcriptional factors to improve reprogramming efficiency. RESULTS: Here, we improved the generation efficiency of integration-free iPSCs from human peripheral blood mononuclear cells by optimizing the method of isolating mononuclear cells from peripheral blood, by modifying the integration of culture medium, and by adjusting the duration of culture time and the combination of different episomal vectors. CONCLUSIONS: With this optimized protocol, a valuable asset for banking patient-specific iPSCs has been established.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming/physiology , Induced Pluripotent Stem Cells/metabolism , Peripheral Blood Stem Cells/metabolism , Cell Differentiation , Humans
9.
Int Immunopharmacol ; 61: 132-139, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29870918

ABSTRACT

CD4+CXCR5+Foxp3+ follicular regulatory T (Tfr) cells possess critical roles in suppressing the germinal center reaction, B cell activation, and follicular helper T cell (Tfh) cytokine secretion. Since diffuse large B cell lymphoma (DLBCL) can arise from B cells undergoing germinal center reaction and/or differentiation, we hypothesized that Tfr cells might be involved in DLBCL. In the present study, we recruited thirty-five DLBCL patients and twenty-five healthy controls. Data showed that DLBCL patients presented an enrichment of circulating CD4+CXCR5+Foxp3+ Tfr cells compared to controls. In the primary tumor isolated from enlarged lymph nodes, Tfr cells made up of roughly 3% to 16% of infiltrating T cells. Higher levels of tumor-infiltrating Tfr cells were observed in patients with less advanced DLBCL stages, and in patients that stayed in remission 24 months after the initial R-CHOP treatment. High BCL6 and high FOXP3 expression was observed in Tfr cells ex vivo. After anti-CD3/CD28 and IL-2 stimulation, the Tfr cells more closely resembled Treg cells and presented high IL10 and TGFB1 expression. CD4+CD25+CXCR5+ Tfr cells and CD4+CD25+CXCR5- non-Tfr Treg cells could suppress CD4+CD25- Tconv cell and CD8+ T cell proliferation with similar capacity. However, Tfr cells were less capable of suppressing IFNG expression than Treg cells, and although both cell types supported CD19+ tumor cell proliferation, Tfr cells were less supportive than the non-Tfr Treg cells. Overall, this study suggested that Tfr cells were involved in intratumoral immunity, were likely beneficial to DLBCL patients, and were functionally distinctive from non-Tfr Treg cells. The distribution pattern and the prognostic value of Tfr cells in DLBCL should be examined in further studies.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, Large B-Cell, Diffuse/epidemiology , T-Lymphocytes, Regulatory/immunology , Adult , Cell Proliferation , Cells, Cultured , China/epidemiology , Female , Forkhead Transcription Factors/metabolism , Germinal Center/immunology , Humans , Immune Tolerance , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , Prevalence , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/metabolism
10.
Stem Cell Res Ther ; 9(1): 92, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29625575

ABSTRACT

BACKGROUND: Replacement therapy for hemophilia remains a lifelong treatment. Only gene therapy can cure hemophilia at a fundamental level. The clustered regularly interspaced short palindromic repeats-CRISPR associated nuclease 9 (CRISPR-Cas9) system is a versatile and convenient genome editing tool which can be applied to gene therapy for hemophilia. METHODS: A patient's induced pluripotent stem cells (iPSCs) were generated from their peripheral blood mononuclear cells (PBMNCs) using episomal vectors. The AAVS1-Cas9-sgRNA plasmid which targets the AAVS1 locus and the AAVS1-EF1α-F9 cDNA-puromycin donor plasmid were constructed, and they were electroporated into the iPSCs. When insertion of F9 cDNA into the AAVS1 locus was confirmed, whole genome sequencing (WGS) was carried out to detect the off-target issue. The iPSCs were then differentiated into hepatocytes, and human factor IX (hFIX) antigen and activity were measured in the culture supernatant. Finally, the hepatocytes were transplanted into non-obese diabetic/severe combined immunodeficiency disease (NOD/SCID) mice through splenic injection. RESULTS: The patient's iPSCs were generated from PBMNCs. Human full-length F9 cDNA was inserted into the AAVS1 locus of iPSCs of a hemophilia B patient using the CRISPR-Cas9 system. No off-target mutations were detected by WGS. The hepatocytes differentiated from the inserted iPSCs could secrete hFIX stably and had the ability to be transplanted into the NOD/SCID mice in the short term. CONCLUSIONS: PBMNCs are good somatic cell choices for generating iPSCs from hemophilia patients. The iPSC technique is a good tool for genetic therapy for human hereditary diseases. CRISPR-Cas9 is versatile, convenient, and safe to be used in iPSCs with low off-target effects. Our research offers new approaches for clinical gene therapy for hemophilia.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Genetic Therapy/methods , Hemophilia B/therapy , Hepatocytes/transplantation , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Cell Transplantation/methods , Cells, Cultured , Factor IX/genetics , Factor IX/metabolism , HEK293 Cells , Hemophilia B/genetics , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Inbred NOD , Mice, SCID
11.
Cell Reprogram ; 19(6): 344-353, 2017 12.
Article in English | MEDLINE | ID: mdl-29135280

ABSTRACT

Successful development of interspecies somatic cell nuclear transfer (iSCNT) embryos depends on compatibilities between ooplasmic and nuclear components. However, the mechanisms by which the compatibilities are regulated are still unknown. In this study, using mouse Oct4-green fluorescent protein (GFP) cells as donors and rabbit oocytes as recipients, we show that Oct4 and other pluripotency related genes were reactivated in some of mouse-rabbit iSCNT embryos, which could also activate Oct4 promoter-driven GFP reporter gene expression. Series nuclear transfer improved the efficiency of Oct4 reactivation. DNA demethylation of Oct4 promoter was detected in GFP positive iSCNT blastocysts, whereas GFP negative iSCNT embryos showed a low efficiency. Our results demonstrate that Oct4-GFP can well label the embryos with epigenetic remodeling and reactivation of pluripotent gene expression. Abundant rabbit mitochondria specific DNAs were identified in reconstructed mouse-rabbit embryos throughout preimplantation stages. Our data demonstrate that epigenetic remodeling and the complete mitochondrial match are not necessary for successful iSCNT embryo development before implantation.


Subject(s)
Cellular Reprogramming , Embryo, Mammalian/physiology , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Nuclear Transfer Techniques/veterinary , Oocytes/physiology , Animals , Cell Nucleus/physiology , Cloning, Organism , Cytoplasm , DNA Methylation , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Embryo, Mammalian/cytology , Embryonic Development , Embryonic Stem Cells/cytology , Female , Green Fluorescent Proteins/metabolism , Hybrid Cells , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oocytes/cytology , Rabbits
12.
Mol Neurobiol ; 54(4): 2395-2405, 2017 05.
Article in English | MEDLINE | ID: mdl-26960328

ABSTRACT

Schwann cells (SCs) proliferation is crucial for nerve regeneration following nerve injury. This study aims to investigate effects of interleukin-22 (IL-22) on SCs proliferation in vitro, as well as the corresponding mechanism. Rat SCs were treated with 100 ng/ml rat IL-22 for 48 h, and cell proliferation and apoptosis were detected using fluorescent staining and flow cytometry. After transcriptome sequencing, raw reads were filtered and mapped to reference genome rn5. Then, differentially expressed genes (DEGs) and long non-coding RNAs (DElncRNAs) between IL-22 and control groups were identified (tool: Cuffdiff). Functional and pathway enrichment analyses were performed (tool: GOFunction), and protein-protein interaction (PPI) network was constructed (tool: STRING and Cytoscape). Furthermore, Pearson's correlations between DEGs and DElncRNAs were analyzed, and regulatory network of DEGs, DElncRNAs, and transcription factors (TFs) was constructed. IL-22 significantly inhibited proliferation (p value < 0.05) and promoted apoptosis of Schwann cells. Totally, 932 DEGs and 118 DElncRNAs were identified, among which Ccl2 and Ccna2 were hub genes in PPI network. Up-regulated DEGs were enriched in apoptosis related terms, whereas down-regulated DEGs were enriched in proliferation related terms. DElncRNAs like NONRATT023505, NONRATG020400, and NONRATT022748 were correlated with multiple DEGs enriched in cell cycle and division. Moreover, up-regulated TFs Egr1, Cebpd, and Atf4 play crucial roles in regulatory network, and NONRATG020400-Cebpd-Ccl2, NONRATT023505/NONRATT022748-Atf4-Ccna2, and NONRATT022748-Egr1-Id1/Aldoc/Eno2/F3/Serpine1 regulatory pathways were identified in SCs after IL-22 treatment. IL-22 might influence SCs proliferation and apoptosis via regulating lncRNA-TF-gene pathways in SCs. However, more studies are required to confirm these results.


Subject(s)
Computational Biology/methods , Interleukins/pharmacology , Schwann Cells/cytology , Schwann Cells/metabolism , Sequence Analysis, RNA , Transcriptome/genetics , Animals , Cell Proliferation/drug effects , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Protein Interaction Maps/drug effects , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats , Schwann Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/metabolism , Transcriptome/drug effects , Interleukin-22
13.
Mol Cell Biol ; 37(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28031328

ABSTRACT

RAD6, an E2 ubiquitin-conjugating enzyme, is a key node for determining different DNA damage repair pathways, controlling both the error-prone and the error-free DNA damage repair pathways through differential regulation of the ubiquitination of the proliferating cell nuclear antigen (PCNA) protein. However, whether other pathways are involved in the RAD6-mediated regulation of DNA damage repair is still unclear. To deeply understand the molecular mechanisms of RAD6 in DNA damage repair, we performed a proteomic analysis and identified the changes of the protein-protein interaction (PPI) networks of RAD6 before and after X-ray irradiation. Furthermore, our study indicated that a proteasome-related event is likely involved in the DNA damage repair process. Moreover, we found that RAD6 promotes proteasome activity and nuclear translocation by enhancing the degradation of PSMF1 and the lamin B receptor (LBR). Therefore, we provide a novel pathway that is employed by RAD6 in response to DNA damage.


Subject(s)
DNA Damage , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Maps , Proteomics/methods , Ubiquitin-Conjugating Enzymes/metabolism , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Repair/radiation effects , Down-Regulation/genetics , Down-Regulation/radiation effects , HEK293 Cells , Humans , Models, Biological , Proteins/metabolism , Proteolysis/radiation effects , Receptors, Cytoplasmic and Nuclear/metabolism , Ubiquitin/metabolism , X-Rays , Lamin B Receptor
14.
Exp Cell Res ; 350(1): 154-160, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27888017

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is a common and aggressive cancer caused by the malignant transformation of B cells. Although it has been established that the follicular helper T (Tfh) cells play a central role in B cell development, little information is available on their involvement in DLBCL pathogenesis. We studied the role of the peripheral Tfh equivalent, the CXCR5+ CD4+ T cells, in DLBCL. Data showed that compared to CXCR5- CD4+ T cells, CXCR5+ CD4+ T cells were significantly more effective at promoting the proliferation as well as inhibiting the apoptosis of primary autologous DLBCL tumor cells. Surprisingly, we found that at equal cell numbers, CXCR5+ CD4+ T cells in DLBCL patients secreted significantly less interleukin (IL)-21 than CXCR5- CD4+ T cells, while the level of IL-10 secretion was significant elevated in the CXCR5+ compartment compared to the CXCR5- compartment. Neutralization of IL-10 in the primary DLBCL-CXCR5+ CD4+ T cell coculture compromised the CXCR5+ CD4+ T cell-mediated pro-tumor effects, in a manner that was dependent on the concentration of anti-IL-10 antibodies. The CXCR5+ compartment also contained significantly lower frequencies of cytotoxic CD4+ T cells than the CXCR5- compartment. In conclusion, our investigations discovered a previously unknown pro-tumor role of CXCR5-expressing circulating CD4+ T cells, which assisted the survival and proliferation of primary DLBCL cells through IL-10.


Subject(s)
Lymphocyte Activation/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Receptors, CXCR5/immunology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry/methods , Humans , Interleukin-10/metabolism , Interleukins/metabolism , Signal Transduction
17.
Tumour Biol ; 37(1): 829-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26254098

ABSTRACT

Rituximab is a chimeric monoclonal antibody directed against the CD20 antigen. Treatment using rituximab in combination with chemotherapy has dramatically improved overall survival rate of diffuse large B cell lymphoma (DLBCL). Since rituximab can deplete both lymphoma B cells and normal B cells, how rituximab-treatment affects normal B cell function in DLBCL patients under remission is unclear. Here, we examined peripheral blood B cell composition and antigen-specific B cell responses in DLBCL patients in remission and observed reductions in the frequencies of total B cell as well as several major B cell subsets, including CD19(+)IgD(+) naive B cells, CD19(+)IgD(-)CD27(+) memory B cells, and CD19(lo)CD27(hi) plasmablasts. Moreover, tetanus toxin (TT)-specific B cell proliferation was reduced in DLBCL patients in remission. On the other hand, HA-specific IgG-secreting B cell responses could be stimulated by influenza vaccination in DLBCL patients in remission, demonstrating that the machinery for generating de novo adaptive B cell responses was functional in DLBCL patients in remission. Our results provided insights in normal B cell function in DLBCL patients in remission.


Subject(s)
Antineoplastic Agents/therapeutic use , B-Lymphocytes/immunology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Rituximab/therapeutic use , Adult , Antigens, CD19/metabolism , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoglobulin G/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Phenotype , Remission Induction , Young Adult
18.
Zhonghua Xue Ye Xue Za Zhi ; 36(4): 331-6, 2015 Apr.
Article in Chinese | MEDLINE | ID: mdl-25916298

ABSTRACT

OBJECTIVE: To improve the MigR1-CD19-CAR (chimeric antigen receptor) that contains a single chain variable region (scFv) which targeted to CD19 through a retroviral vector transduction efficiency of T-lymphocytes. METHODS: Insert the CD19-CAR fragment into the retroviral vector (MigR1) through recombinant DNA technology, after transfecting plat-A packaging cell lines, viral supernatant was collected to transduce K562 cell line and activated human T-lymphocytes. We used flow cytometry to determine the transduction efficiency and RT-PCR to confirm the transcription of CD19-CAR gene. The ability of the transduced T cells to produce IFN-γ and TNF-α in a CD19-specific manner was measured in an enzyme-linked immunosorbent (ELISA) assay. RESULTS: (1)Using MigR1-CD19-CAR retroviral vector to produce the high titer retrovirus. (2)MigR1-CD19-CAR transduction efficiency of K562 cell line was significantly higher than human T-lymphocytes (P<0.01). (3)120 min centrifugation could significantly improve transduction efficiency of T-lymphocytes to (54.5±14.6)%. (4)Transduction efficiency could be improved by deciding transduce time according to T-lymphocytes proliferation fold in vitro individually, and the highest transduction efficiency in the study was 69.3%. The CD19-CAR gene sequence was transcripted specificly with high efficiency. (5) IFN-γ and TNF-α released by CD19-CAR transduced T-lymphocytes significantly increased to (13 230±1 543) pg/ml and (4 217±211) pg/ml when coculture with CD19-K562 cells. CONCLUSION: We have successfully constructed a second generation CAR which targeted to CD19 through a retroviral vector called MigR1 (MigR1-CD19-CAR). Deciding transduce time according to T-lymphocytes proliferation fold in vitro individually and 120 min centrifugation could improve the CAR transduction efficiency of T-lymphocytes. RT-PCR confirmed that the CD19-CAR gene was specificly transcripted with high efficiency. IFN-γ and TNF-α released by CD19-CAR transduced T-lymphocytes significantly increased when activated by target cells.


Subject(s)
T-Lymphocytes , Antigens, CD19 , Cell Proliferation , Flow Cytometry , Genetic Vectors , Humans , K562 Cells , Recoverin , Retroviridae , Transfection
19.
Stem Cells Transl Med ; 4(4): 320-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25742692

ABSTRACT

Reprogramming human adult blood mononuclear cells (MNCs) cells by transient plasmid expression is becoming increasingly popular as an attractive method for generating induced pluripotent stem (iPS) cells without the genomic alteration caused by genome-inserting vectors. However, its efficiency is relatively low with adult MNCs compared with cord blood MNCs and other fetal cells and is highly variable among different adult individuals. We report highly efficient iPS cell derivation under clinically compliant conditions via three major improvements. First, we revised a combination of three EBNA1/OriP episomal vectors expressing five transgenes, which increased reprogramming efficiency by ≥10-50-fold from our previous vectors. Second, human recombinant vitronectin proteins were used as cell culture substrates, alleviating the need for feeder cells or animal-sourced proteins. Finally, we eliminated the previously critical step of manually picking individual iPS cell clones by pooling newly emerged iPS cell colonies. Pooled cultures were then purified based on the presence of the TRA-1-60 pluripotency surface antigen, resulting in the ability to rapidly expand iPS cells for subsequent applications. These new improvements permit a consistent and reliable method to generate human iPS cells with minimal clonal variations from blood MNCs, including previously difficult samples such as those from patients with paroxysmal nocturnal hemoglobinuria. In addition, this method of efficiently generating iPS cells under feeder-free and xeno-free conditions allows for the establishment of clinically compliant iPS cell lines for future therapeutic applications.


Subject(s)
Cell Culture Techniques , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Adult , Blood Cells/cytology , Culture Media, Serum-Free , Feeder Cells/cytology , Flow Cytometry , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/transplantation , Leukocytes, Mononuclear/cytology , Vitronectin/administration & dosage
20.
J Anesth ; 29(4): 570-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25716536

ABSTRACT

OBJECTIVE: The aim of this meta-analysis is to compare the potential effects of inhalation anesthetics with total intravenous anesthetics on alveolar cytokine expression and lung-related clinical outcomes in patients undergoing one-lung ventilation (OLV) for thoracic surgery. METHODS: We retrieved the PubMed, EMBASE, and the Cochrane Library respectively to identify randomized controlled trials comparing different anesthetics (volatile anesthetics vs. intravenous anesthetics) on the pulmonary inflammatory response to OLV. The primary outcomes were the levels of alveolar concentrations of inflammatory cytokines. RESULTS: Eight randomized controlled trials that included 365 patients were screened. Overall, there were significant differences in the concentration of alveolar inflammatory mediators between volatile group and intravenous group, in which volatile group had lower levels of TNF-α (SMD -1.51; 95 % CI -2.15 to -0.87; p < 0.001), IL-6 (SMD -0.70; 95 % CI -0.99 to -0.41; p < 0.001) and IL-8 (SMD -1.32; 95 % CI -2.20 to -0.45; p = 0.003). The overall number of pulmonary complications in the volatile group was smaller (RR 0.42; 95 % CI 0.23-0.77; p = 0.005) and patients in that group had significantly abridged hospitalization stay (WMD -3.59 days; 95 % CI -5.70 to -1.48 days; p = 0.001). CONCLUSIONS: Inhalation anesthetics might be preferable in patients undergoing OLV for thoracic surgery and their protective effects might work via attenuating inflammatory responses.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Anesthetics, Intravenous/administration & dosage , One-Lung Ventilation/methods , Propofol/administration & dosage , Cytokines/metabolism , Humans , Length of Stay , Lung/metabolism , Randomized Controlled Trials as Topic , Thoracic Surgical Procedures/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...