24 research outputs found

    Development of Space-Time-Controlled Multi-Stage Pulsed Magnetic Field Forming and Manufacturing Technology at the WHMFC*

    Get PDF
    In November 2011, the Project of Basic Research of Forming by Space-Time-Controlled Multi-Stage Pulsed Magnetic Field (Stic-Must-PMF) was supported by the National Basic Research Program of China (973 Project, 2011.11-2016.08). It is aimed at achieving breakthroughs in manufacturing technology to solve current problems in forming largescale and complex sheet and tube parts and components, imposed by the limitations of existing equipment and materials forming properties. The objective of our research group focuses on the design principles and structural layout optimization of Stic-Must-PMF facility. And this paper will report the development of Stic-Must-PMF forming and manufacturing technology at the Wuhan National High Magnetic Field Center (WHMFC) including numerical modeling, experimental setup and experimental studies

    A Unique Human Immunoglobulin Heavy Chain Variable Domain-Only CD33 CAR for the Treatment of Acute Myeloid Leukemia

    Get PDF
    Acute myeloid leukemia (AML) remains a challenging pediatric and adult disease. Given the elevated expression of the CD33 antigen on leukemic blasts, therapeutic approaches to AML now feature the approved antibody drug conjugate (Mylotarg, GO) and investigational CART cell approaches incorporating CD33-binding domains derived from humanized scFvs. We designed a functional chimeric antigen receptor utilizing a human targeting sequence, derived from a heavy chain variable domain, termed CAR33VH. Lentiviral-based expression vectors which encoded CAR constructs incorporating the novel binding domain (CAR33VH), or the My96 scFv control binder (My96CAR) in frame with a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain and a CD3 zeta activation domain, were transduced into primary human CD4+ and CD8+ T cells, and CAR expression was confirmed by flow cytometry. CAR33VH, similar to My96CAR, demonstrated robust and specific cytotoxicity in short-term and long-term co-incubation killing assays against CD33+ AML lines. In overnight cytokine release assays in which CAR T cells were challenged with the CD33+ tumor cells HL-60, MOLM-14 and KG-1a, CAR33VH elicited IFN-gamma, TNF-alpha and IL-2. This was seen with CD33+ cell lines, but not when CAR T were cultured alone. Studies with a CD33āˆ’ cell line engineered to stably express the full length CD33 variant 1, or the naturally occurring CD33 splice variant 2, revealed that both CAR33VH and My96CAR, target the V domain of CD33, suggesting a similar therapeutic profile. Colony-formation assays utilizing peripheral blood CD34+ hematopoietic stem cells treated with CAR33VH, My96CAR, or with an untransduced T cell control, yielded similar numbers of BFU-E erythroid and CFU-GM myeloid colonies, suggesting a lack of CAR-related overt toxicity. In an in vivo AML model, NSG mice engrafted with MOLM-14 cells stably expressing firefly luciferase, both CAR33VH and CARMy96 efficiently eliminated tumors. In conclusion, we demonstrate for the first time the feasibility and efficacy of employing human variable domain-only binder derived from a phage display library in an anti-AML CAR design. CAR33VH, comprised of a human heavy-chain variable fragment-only antigen binding domain, was efficient in tumor killing in vitro and in vivo, and showed comparable functionality to the scFv-based My96CAR

    Graphene Quantum Dots Doped PVDF(TBT)/PVP(TBT) Fiber Film with Enhanced Photocatalytic Performance

    Get PDF
    We report the fabrication of polyvinylidene fluoride (tetrabutyl titanate)/polyvinyl pyrrolidone ((tetrabutyl titanate))-graphene quantum dots [PVDF(TBT)/PVP(TBT)-GQDs] film photocatalyst with enhanced photocatalytic performance. The polyvinylidene fluoride (tetrabutyl titanate)/polyvinyl pyrrolidone ((tetrabutyl titanate)) [PVDF(TBT)/PVP(TBT)] film was first prepared with a dual-electrospinning method and then followed by attaching graphene quantum dots (GQDs) to the surface of the composite film through a hydrothermal method. Later, part of the PVP in the composite film was dissolved by a hydrothermal method. As a result, a PVDF(TBT)/PVP(TBT)-GQDs film photocatalyst with a larger specific surface area was achieved. The photocatalytic degradation behavior of the PVDF(TBT)/PVP(TBT)-GQDs film photocatalyst was examined by using Rhodamine B as the target contaminant. The PVDF(TBT)/PVP(TBT)-GQDs photocatalyst showed a higher photocatalytic efficiency than PVDF(TBT)-H2O, PVDF(TBT)/PVP(TBT)-H2O, and PVDF(TBT)-GQDs, respectively. The enhanced photocatalytic efficiency can be attributed to the broader optical response range of the PVDF(TBT)/PVP(TBT)-GQDs photocatalyst, which makes it useful as an effective photocatalyst under white light irradiation

    Metabolic Brain Network Analysis of FDG-PET in Alzheimerā€™s Disease Using Kernel-Based Persistent Features

    No full text
    Recent research of persistent homology in algebraic topology has shown that the altered network organization of human brain provides a promising indicator of many neuropsychiatric disorders and neurodegenerative diseases. However, the current slope-based approach may not accurately characterize changes of persistent features over graph filtration because such curves are not strictly linear. Moreover, our previous integrated persistent feature (IPF) works well on an rs-fMRI cohort while it has not yet been studied on metabolic brain networks. To address these issues, we propose a novel univariate network measurement, kernel-based IPF (KBI), based on the prior IPF, to quantify the difference between IPF curves. In our experiments, we apply the KBI index to study fluorodeoxyglucose positron emission tomography (FDG-PET) imaging data from 140 subjects with Alzheimer’s disease (AD), 280 subjects with mild cognitive impairment (MCI), and 280 healthy normal controls (NC). The results show the disruption of network integration in the progress of AD. Compared to previous persistent homology-based measures, as well as other standard graph-based measures that characterize small-world organization and modular structure, our proposed network index KBI possesses more significant group difference and better classification performance, suggesting that it may be used as an effective preclinical AD imaging biomarker

    ANP32E induces tumorigenesis of tripleā€negative breast cancer cells by upregulating E2F1

    No full text
    Tripleā€negative breast cancer (TNBC) lacks expression of estrogen receptor (ER), progesterone receptor, and the HER2 receptor; it is highly proliferative and becomes the deadliest forms of breast cancer. Effective prognostic methods and therapeutic targets for TNBC are required to improve patient outcomes. Here, we report that acidic nuclear phosphoprotein 32 family member E (ANP32E), which promotes cell proliferation in mammalian development, is highly expressed in TNBC cells compared to other types of breast cancer. High expression of ANP32E correlates significantly with worse overall survival (OS; PĀ <Ā 0.001) and higher risks of disease recurrence (PĀ <Ā 0.001) in patients with TNBC. Univariate and multivariate Coxā€regression models show that ANP32E is an independent prognostic factor in TNBC. Furthermore, we discovered that ANP32E promotes tumor proliferation inĀ vitro by inducing G1/S transition, and ANP32E inhibition suppresses tumor formation inĀ vivo. By examining the expression of E2F1, cyclin E1, and cyclin E2, we discovered that ANP32E promotes the G1/S transition by transcriptionally inducing E2F1. Taken together, our study shows that ANP32E is an efficient prognostic marker, and it promotes the G1/S transition and induces tumorigenesis of TNBC cells by transcriptionally inducing E2F1
    corecore