Barwicz, T., Bo Peng, Leidy, R., Janta-Polczynski, A., Houghton, T., Khater, M., Kamlapurkar, S., Engelmann, S., Fortier, P., Boyer, N., and Green, W.M.J.
IEEE Journal of Selected Topics in Quantum Electronics IEEE J. Select. Topics Quantum Electron. Selected Topics in Quantum Electronics, IEEE Journal of. 25(3):1-13 Jun, 2019
Bo Peng, Hang Gong, Han Tian, Quan Zhuang, Junhui Li, Ke Cheng, and Yingzi Ming
Journal of Translational Medicine, Vol 18, Iss 1, Pp 1-11 (2020)
Subjects
Immune monitoring, Kidney transplant, Machine learning, Pneumonia, Immunosuppression, and Medicine
Abstract
Abstract Background Kidney transplantation is the optimal treatment to cure the patients with end-stage renal disease (ESRD). However, the infectious complication, especially pneumonia, is the main cause of mortality in the early stage. Immune monitoring by relevant biomarkers provides direct evidence of immune status. We aimed to study the association between immune monitoring and pneumonia in kidney transplant patients through machine learning models. Methods A total of 146 patients receiving the immune monitoring panel in our center, including 46 pneumonia recipients and 100 stable recipients, were retrospectively reviewed to develop the models. All the models were validated by external data containing 10 pneumonia recipients and 32 stable recipients. The immune monitoring panel consisted of the percentages and absolute cell counts of CD3+CD4+ T cells, CD3+CD8+ T cells, CD19+ B cells and natural killer (NK) cells, and median fluorescence intensity (MFI) of human leukocyte antigen (HLA)-DR on monocytes and CD64 on neutrophils. The machine learning models including support vector machine (SVM), logistic regression (LR), multi-layer perceptron (MLP) and random forest (RF) were applied for analysis. Results The pneumonia and stable groups showed significant difference in cell counts of each subpopulation and MFI of monocyte HLA-DR and neutrophil CD64. The SVM model by monocyte HLA-DR (MFI), neutrophil CD64 (MFI), CD8+ T cells (cells/μl), NK cells (cell/μl) and TBNK (T cells, B cells and NK cells, cells/μl) had the best performance with the average area under the curve (AUC) of 0.940. The RF model best predicted the patients who would progress into severe pneumonia, with the average AUC of 0.760. All the models had good performance validated by external data. Conclusions The immune monitoring panel was tightly associated with pneumonia in kidney transplant recipients. The models developed by machine learning techniques identified patients at risk and predicted the prognosis. Based on the results of immune monitoring, better individualized therapy might be achieved.
Tao Yao, Yan Gao, Qin Cui, Bo Peng, Yan Chen, Jiansheng Li, Chao Huang, Chunping He, Jie Pu, Jiajun Wei, Yanqiang Zhan, Jie Yan, Jinghua Tian, Zhaohui Zhang, and Zhichao Liu
BMC Infectious Diseases, Vol 20, Iss 1, Pp 1-9 (2020)
Subjects
Characteristics, COVID-19, Pneumonia, Death, Infectious and parasitic diseases, and RC109-216
Abstract
Abstract Background With the widespread outbreak of novel coronavirus diseases 2019(COVID-19), more and more death cases were reported, however, limited data are available for the patients who died. We aimed to explore the clinical characteristics of deaths with COVID-19 pneumonia. Methods We abstracted and analyzed epidemiological, demographic, clinical, and laboratory data from 83 death cases with COVID-19 pneumonia in East Hospital of Wuhan University Renmin Hospital, between January 26, 2020, and February 28, 2020. Results Of the 83 deaths, none was the medical staff. The mean age was 71.8 years (SD 13.2; range, 34–97 years) and 53(63.9%) were male. The median from onset to admission was 10 days (IQR 7–14: range, 2–43 days), to death was 17 days (IQR 14–21: range, 6–54 days). Most deaths (66[80%]) had underlying comorbid diseases, the most of which was hypertension [47(57%)]. The main initial symptoms of these 83 deaths were shortness of breath(98.8%), fever(94%), and myalgia or fatigue(90.4%). Laboratory analyses showed the lymphocytopenia in 69(83%) deaths, hypoalbuminemia in 77(93%) deaths, the elevation of lactate dehydrogenase in 79(95%) deaths, procalcitonin in 69(83%) deaths and C-reactive protein in 79(95%) deaths. All 83 patients received antiviral treatment, 81(97.6%) deaths received antibiotic therapy, 54(65.1%) deaths received glucocorticoid therapy, and 20(24.1%) patients received invasive mechanical ventilation. Conclusion Most of the deaths with COVID-19 pneumonia were elderly patients with underlying comorbid diseases, especially those over 70 years of age. The time of death after the onset of the disease was mostly 15–21 days. More care should be given to the elderly in further prevention and control strategies of COVID-19.
Yiyuan Wan, Hesheng Luo, Ming Yang, Xia Tian, Bo Peng, Ting Zhan, Xiaoli Chen, Yu Ding, Jinrong He, Xueting Cheng, Xiaodong Huang, and Yadong Zhang
Molecular Therapy: Oncolytics, Vol 18, Iss , Pp 432-442 (2020)
Subjects
miR-324-5p, KLF3, proliferation, apoptosis, pancreatic cancer, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, and RC254-282
Abstract
Pancreatic cancer cells are characterized by high cell proliferation and low cell apoptosis, but the factors involved in these processes remain to be further studied. In this study, we report that miR-324-5p regulates the proliferation and apoptosis of pancreatic cancer cells through regulating the expression of Krüppel-like factor 3 (KLF3). In both pancreatic cancer tissues and cell lines, the levels of miR-324-5p are significantly increased. Inhibition of miR-324-5p represses cell proliferation but promotes cell apoptosis, whereas overexpression of miR-324-5p exerts the opposite effect. Furthermore, we identified KLF3, a factor regulating pancreatic cancer cell proliferation and apoptosis, as a new direct downstream target of miR-324-5p. Our results suggest that miR-324-5p plays an important role in pancreatic cancer cell proliferation and apoptosis via downregulating the expression of KLF3.
Cheng Qin, Xin-Sheng Zeng, Bo Peng, Yu-Ni Tang, You-Di Zhou, and Bo Song
Guoji Yanke Zazhi, Vol 20, Iss 9, Pp 1493-1497 (2020)
Subjects
posterior cataract, d-δ-tocopherol, human lens epithelial sra cells, proliferation, Ophthalmology, and RE1-994
Abstract
AIM: To investigate the effect of d-δ-tocopherol on the growth of human lens epithelial SRA cells and its related molecular mechanism, and to provide experimental basis for the treatment and prevention of posterior cataract with d-δ-tocopherol. METHODS: The experiment was divided into 6 groups, blank control group and experimental group, that is, five different concentrations of d-δ-tocopherol(40, 60, 80, 100, 120)μmoL/L. The proliferation inhibition rate of each group was detected by thiazolam(MTT)assay. The morphology of human lens epithelial SRA cells was observed under inverted microscope. Cell cycle was detected by flow cytometry and the expression of bcl-2, bax, Cyclin D1, P21 protein was detected by Western Blot(WB). RESULTS: With the increase of d-δ-tocopherol concentration, the SRA cells decreased significantly compared with the control group; the MTT results showed that with the increase of d-δ-tocopherol concentration, the inhibition rate of cell proliferation increased gradually, the difference was statistically significant(PPPCONCLUSION: d-δ-tocopherol can significantly inhibit the proliferation of human lens epithelial SRA cells and block the cell cycle in S phase. d-δ-tocopherol can inhibit the proliferation of human lens epithelial cells. The proliferation of human lens epithelial SRA cells may be achieved by inhibiting the expression of bcl-2, P21, Cyclin D1 and inducing the expression of bax.
Haiping Jiang, Yulong Zheng, Jiong Qian, Chenyu Mao, Xin Xu, Ning Li, Cheng Xiao, Huan Wang, Lisong Teng, Hui Zhou, Shuyan Wang, Donglei Zhu, Bo Peng, Lin Shen, and Nong Xu
BMC Cancer, Vol 20, Iss 1, Pp 1-8 (2020)
Subjects
Sintilimab, Capecitabine, Oxaliplatin, Gastric/gastroesophageal junction adenocarcinoma, Tumor mutation burden, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, and RC254-282
Abstract
Abstract Background Sintilimab blocks the interaction between programmed death-1 (PD-1) and its ligands. The safety and efficacy of sintilimab combined with oxaliplatin/capecitabine (CapeOx) as first-line treatment were evaluated in patients with gastric (G)/gastroesophageal junction (GEJ) adenocarcinoma in a phase Ib clinical trial. Methods Patients with locally advanced or metastatic G/GEJ adenocarcinoma without previous systemic treatment were enrolled as one cohort of a multi-cohort study. Sintilimab was administered at a dose of 200 mg intravenously (IV) in combination with CapeOx (1000 mg/m2 capecitabine orally, bid, D1–14 and 130 mg/m2 oxaliplatin IV, D1) every 21 days for up to 6 cycles. After combination treatment, patients continued to receive sintilimab (200 mg) at 3 weekly intervals as maintenance therapy until progressive disease (PD), unacceptable toxicity, withdrawal of informed consent, or for up to 24 months. Adverse events (AEs) were monitored to assess safety in terms of their frequency, intensity and causality. The efficacy endpoints included the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS) and overall survival (OS). Tumor mutation burden (TMB) was evaluated for its association with clinical response. Results A total of 20 patients were enrolled and received sintilimab plus CapeOx. All patients reported treatment-related AEs (TRAEs). Grade 3–4 TRAEs were found in 11 (55.0%) patients. Seventeen patients obtained partial response and the ORR was 85.0% (95% CI: 62.1–96.8%). Three (15.0%) had stable disease and DCR was 100.0% (95% CI: 83.2–100.0%). As data cutoff of May 1, 2019, the median follow-up was 7.8 months. The median PFS was 7.5 months (95% CI: 6.2–9.4) and median OS had not been reached. The OS rates at 6 months and 12 months were 100.0 and 68.0%. No association was observed between TMB and efficacy. Conclusions Sintilimab combined with CapeOx as first-line treatment demonstrated acceptable safety and promising efficacy. Trial registration ClinicalTrials.gov, NCT02937116 . Registered 8 October 2016.
Zhiyan Wang, Xiaodong Cai, Rirang Qiu, Chen Yao, Ye Tian, Chen Gong, Yingli Zhang, Bin Xu, Doudou Zhang, Yu Zang, Jiali Liu, Bo Peng, and Luming Li
Frontiers in Psychiatry, Vol 11 (2021)
Subjects
lateral habenula, patient, local field potentials, deep brain stimualtion, treatment resistant depression, Psychiatry, and RC435-571
Abstract
Treatment-resistant depression (TRD) is a chronic and severe psychiatric illness associated with limited therapeutic options. Deep brain stimulation (DBS) is a promising therapy for TRD patients. However, its safety and efficacy are still unclear. Here we reported the safety and efficacy of lateral habenula (LHb) DBS for a TRD patient who had failed medical, psychological, electroconvulsive, and ketamine therapy. The DBS system is compatible with 3T magnetic resonance imaging along with local field potential (LFP) streaming. Two DBS electrodes were implanted at the bilateral LHb without any complication. The patient showed acute stimulation effects and achieved long-term improvements in his depression, anxiety, and sleep with left LHb 160 Hz frequency stimulation, accompanying the change of LFPs. These results provided clinical evidence toward the safety and efficacy and electrophysiological basis of LHb DBS for TRD.
The intricate interplay between malignant cells and host cellular and non-cellular components play crucial role in different stages of tumor development, progression, and metastases. Tumor and stromal cells communicate to each other through receptors such as integrins and secretion of signaling molecules like growth factors, cytokines, chemokines and inflammatory mediators. Chemokines mediated signaling pathways have emerged as major mechanisms underlying multifaceted roles played by host cells during tumor progression. In response to tumor stimuli, host cells-derived chemokines further activates signaling cascades that support the ability of tumor cells to invade surrounding basement membrane and extra-cellular matrix. The host-derived chemokines act on endothelial cells to increase their permeability and facilitate tumor cells intravasation and extravasation. The tumor cells-host neutrophils interaction within the vasculature initiates chemokines driven recruitment of inflammatory cells that protects circulatory tumor cells from immune attack. Chemokines secreted by tumor cells and stromal immune and non-immune cells within the tumor microenvironment enter the circulation and are responsible for formation of a “pre-metastatic niche” like a “soil” in distant organs whereby circulating tumor cells “seed’ and colonize, leading to formation of metastatic foci. Given the importance of host derived chemokines in cancer progression and metastases several drugs like Mogamulizumab, Plerixafor, Repertaxin among others are part of ongoing clinical trial which target chemokines and their receptors against cancer pathogenesis. In this review, we focus on recent advances in understanding the complexity of chemokines network in tumor microenvironment, with an emphasis on chemokines secreted from host cells. We especially summarize the role of host-derived chemokines in different stages of metastases, including invasion, dissemination, migration into the vasculature, and seeding into the pre-metastatic niche. We finally provide a brief description of prospective drugs that target chemokines in different clinical trials against cancer.