A role for thrombopoietin in hemangioblast development.
Vascular endothelial growth factor (VEGF) and stem cell factor (SCF) act as growth factors for the hemangioblast,an embryonic progenitor of the hematopoietic and endothelial lineages. Because thrombopoietin (TPO) and its receptor,c-Mpl,regulate primitive hematopoietic populations,including bone marrow hematopoietic stem cells,we investigated whether TPO acts on the hemangioblasts that derive from differentiation of embryonic stem cells in vitro. Reverse transcriptase polymerase chain reaction analysis detected expression of c-Mpl beginning on day 3 of embryoid body differentiation when the hemangioblast first arises. In assays of the hemangioblast colony-forming cell (BL-CFC),TPO alone supported BL-CFC formation and nearly doubled the number of BL-CFC when added together with VEGF and SCF. When replated under the appropriate conditions,TPO-stimulated BL-CFC gave rise to secondary hematopoietic colonies,as well as endothelial cells,confirming their nature as hemangioblasts. Addition of a neutralizing anti-VEGF antibody did not block TPO enhancement of BL-CFC formation,suggesting that TPO acts independently of VEGF. These results establish that Mpl signaling plays a role in the earliest stages of hematopoietic development and that TPO represents a third growth factor influencing hemangioblast formation.
View Publication
产品类型:
产品号#:
03434
03444
产品名:
MethoCult™GF M3434
MethoCult™GF M3434
Ciceri P et al. ( 2014)
Nature chemical biology 10 4 305--312
Dual kinase-bromodomain inhibitors for rationally designed polypharmacology.
Concomitant inhibition of multiple cancer-driving kinases is an established strategy to improve the durability of clinical responses to targeted therapies. The difficulty of discovering kinase inhibitors with an appropriate multitarget profile has,however,necessitated the application of combination therapies,which can pose major clinical development challenges. Epigenetic reader domains of the bromodomain family have recently emerged as new targets for cancer therapy. Here we report that several clinical kinase inhibitors also inhibit bromodomains with therapeutically relevant potencies and are best classified as dual kinase-bromodomain inhibitors. Nanomolar activity on BRD4 by BI-2536 and TG-101348,which are clinical PLK1 and JAK2-FLT3 kinase inhibitors,respectively,is particularly noteworthy as these combinations of activities on independent oncogenic pathways exemplify a new strategy for rational single-agent polypharmacological targeting. Furthermore,structure-activity relationships and co-crystal structures identify design features that enable a general platform for the rational design of dual kinase-bromodomain inhibitors.
View Publication
产品类型:
产品号#:
73472
73474
产品名:
TG101348
TG101348
T. Hara et al. (Apr 2024)
Cancer Science 115 7
High N6‐methyladenosine‐activated TCEAL8 mRNA is a novel pancreatic cancer marker
N6‐methyladenosine (m6A) is an RNA modification involved in RNA processing and widely found in transcripts. In cancer cells,m6A is upregulated,contributing to their malignant transformation. In this study,we analyzed gene expression and m6A modification in cancer tissues,ducts,and acinar cells derived from pancreatic cancer patients using MeRIP‐seq. We found that dozens of RNAs highly modified by m6A were detected in cancer tissues compared with ducts and acinar cells. Among them,the m6A‐activated mRNA TCEAL8 was observed,for the first time,as a potential marker gene in pancreatic cancer. Spatially resolved transcriptomic analysis showed that TCEAL8 was highly expressed in specific cells,and activation of cancer‐related signaling pathways was observed relative to TCEAL8‐negative cells. Furthermore,among TCEAL8‐positive cells,the cells expressing the m6A‐modifying enzyme gene METTL3 showed co‐activation of Notch and mTOR signaling,also known to be involved in cancer metastasis. Overall,these results suggest that m6A‐activated TCEAL8 is a novel marker gene involved in the malignant transformation of pancreatic cancer.
View Publication
产品类型:
产品号#:
100-0781
100-0820
产品名:
PancreaCult™ 类器官生长培养基 (人)
PancreaCult™ 类器官启动培养基 (人)
Z. Chen et al. (nov 2019)
Gastroenterology 156 4 1127--1139.e8
Integrated Analysis of Mouse and Human Gastric Neoplasms Identifies Conserved microRNA Networks in Gastric Carcinogenesis.
BACKGROUND AIMS microRNAs (miRNAs) are small non-coding RNAs that bind to 3'UTR regions of mRNAs to promote their degradation or block their translation. Mice with disruption of the trefoil factor 1 gene (Tff1) develop gastric neoplasms. We studied these mice to identify conserved miRNA networks involved in gastric carcinogenesis. METHODS We performed next-generation miRNA sequencing analysis of normal gastric tissues (based on histology) from subjects without evidence of gastric neoplasm from patients (n=64) and TFF1-knockout mice (n=22). We validated our findings using 270 normal gastric tissues (including 61 samples from patients without evidence of neoplastic lesions) and 234 gastric tumor tissues from 3 separate cohorts of patients and from mice. We performed molecular and functional assays using cell lines (MKN28,MKN45,STKM2,and AGS cells),gastric organoids,and mice with xenograft tumors. RESULTS We identified 117 miRNAs that were significantly deregulated in mouse and human gastric tumor tissues,compared with non-tumor tissues. We validated changes in levels of 6 miRNAs by quantitative real-time PCR analyses of neoplastic gastric tissues from mice (n=39) and 3 independent cohorts patients (332 patients total). We found levels of MIR135B-5p,MIR196B-5p,and MIR92A-5p to be increased in tumor tissues whereas levels of MIR143-3p,MIR204-5p,and MIR133-3p were decreased in tumor tissues. Levels of MIR143-3p were reduced not only in gastric cancer tissues,but also in normal tissues adjacent to tumors in humans and low-grade dysplasia in mice. Transgenic expression of MIR143-3p in gastric cancer cell lines reduced their proliferation and restored their sensitivity to cisplatin. AGS cells with stable transgenic expression of MIR143-3p grew more slowly as xenograft tumors in mice than control AGS cells; tumor growth from AGS cells that expressed MIR143-3p,but not control cells,was sensitive to cisplatin. We identified and validated bromodomain containing 2 (BRD2) as a direct target of MIR143-3p; increased levels of BRD2 in gastric tumors associated with shorter survival times of patients. CONCLUSIONS In an analysis of miRNA profiles of gastric tumors from mice and human patients,we identified a conserved signature associated with early stages of gastric tumorigenesis. Strategies to restore MIR143-3p or inhibit BRD2 might be developed for treatment of gastric cancer.
View Publication
产品类型:
产品号#:
06005
产品名:
IntestiCult™ 肠道类器官生长培养基 (小鼠)
R. Wang et al. (Jul 2025)
Journal of Experimental & Clinical Cancer Research : CR 44 3
MiR-181a-driven downregulation of cholesterol biosynthesis through SREBP2 inhibition suppresses uveal melanoma metastasis
uveal melanoma (UM) is the most common primary intraocular tumor in adults,with metastasis being the leading cause of death. However,effective treatments for metastatic UM remain limited. Emerging evidence suggests that cholesterol metabolism plays a role in cancer progression,but its impact on UM metastasis is not well understood. we investigated the effects of miR-181a on UM metastasis using multiple UM cell lines and a suprachoroidal injection mouse model. Functional assays,including migration,invasion,and cancer stem-like cell (CSC) formation,were performed. The target of miR-181a was identified through bioinformatics,luciferase assays,and western blotting. Cholesterol levels were measured,and in vitro and in vivo studies assessed the therapeutic potential of combining miR-181a with crizotinib. miR-181a significantly decreases UM cell migration,invasion,and metastasis. Mechanistically,miR-181a was found to target sterol regulatory element-binding protein 2 (SREBP2),thereby inhibiting cholesterol biosynthesis. This decrease in cholesterol levels hindered reduced epithelial-to-mesenchymal transition (EMT) and led to a decline in cancer stem-like cell (CSC) populations in UM. Furthermore,elevated cholesterol or overexpression of SREBP2 abrogated the anti-metastatic effects of miR-181a. Additionally,a combination of miR-181a and crizotinib significantly inhibited metastasis,both in vitro and in vivo. miR-181a inhibits UM metastasis by targeting SREBP2 and reducing cholesterol biosynthesis. Its combination with crizotinib may provide a promising therapeutic strategy for metastatic UM. The online version contains supplementary material available at 10.1186/s13046-025-03459-8.
View Publication
产品类型:
产品号#:
01700
01702
01705
产品名:
ALDEFLUOR™ 试剂盒
ALDEFLUOR™测定缓冲液
ALDEFLUOR™ DEAB试剂
Murphy SV et al. (JAN 2013)
Journal of biomedical materials research. Part A 101 1 272--84
Evaluation of hydrogels for bio-printing applications.
In the United States alone,there are approximately 500,000 burn injuries that require medical treatment every year. Limitations of current treatments necessitate the development of new methods that can be applied quicker,result in faster wound regeneration,and yield skin that is cosmetically similar to undamaged skin. The development of new hydrogel biomaterials and bioprinting deposition technologies has provided a platform to address this need. Herein we evaluated characteristics of twelve hydrogels to determine their suitability for bioprinting applications. We chose hydrogels that are either commercially available,or are commonly used for research purposes. We evaluated specific hydrogel properties relevant to bioprinting applications,specifically; gelation time,swelling or contraction,stability,biocompatibility and printability. Further,we described regulatory,commercial and financial aspects of each of the hydrogels. While many of the hydrogels screened may exhibit characteristics suitable for other applications,UV-crosslinked Extracel,a hyaluronic acid-based hydrogel,had many of the desired properties for our bioprinting application. Taken together with commercial availability,shelf life,potential for regulatory approval and ease of use,these materials hold the potential to be further developed into fast and effective wound healing treatments.
View Publication
产品类型:
产品号#:
70025
70025.1
70025.2
70025.3
70047
70047.1
70047.2
70048
70048.1
70048.2
产品名:
冻存的人外周血单个核细胞
冻存的人外周血单个核细胞
冻存的人外周血单个核细胞
冻存的人外周血单个核细胞
X. Wang et al. (jun 2022)
Journal of experimental & clinical cancer research : CR 41 1 210
Expanding anti-CD38 immunotherapy for lymphoid malignancies.
BACKGROUND Lymphoid neoplasms,including multiple myeloma (MM),non-Hodgkin lymphoma (NHL),and NK/T cell neoplasms,are a major cause of blood cancer morbidity and mortality. CD38 (cyclic ADP ribose hydrolase) is a transmembrane glycoprotein expressed on the surface of plasma cells and MM cells. The high expression of CD38 across MM and other lymphoid malignancies and its restricted expression in normal tissues make CD38 an attractive target for immunotherapy. CD38-targeting antibodies,like daratumumab,have been approved for the treatment of MM and tested against lymphoma and leukemia in multiple clinical trials. METHODS We generated chimeric antigen receptor (CAR) T cells targeting CD38 and tested its cytotoxicity against multiple CD38high and CD38low lymphoid cancer cells. We evaluated the synergistic effects of all-trans retinoic acid (ATRA) and CAR T cells or daratumumab against cancer cells and xenograft tumors. RESULTS CD38-CAR T cells dramatically inhibited the growth of CD38high MM,mantle cell lymphoma (MCL),Waldenstrom's macroglobulinemia (WM),T-cell acute lymphoblastic leukemia (T-ALL),and NK/T-cell lymphoma (NKTCL) in vitro and in mouse xenografts. ATRA elevated CD38 expression in multiple CD38low cancer cells and enhanced the anti-tumor activity of daratumumab and CD38-CAR T cells in xenograft tumors. CONCLUSIONS These findings may expand anti-CD38 immunotherapy to a broad spectrum of lymphoid malignancies and call for the incorporation of ATRA into daratumumab or other anti-CD38 immunological agents for cancer therapy.
View Publication
RNAi screen identifies MAPK14 as a druggable suppressor of human hematopoietic stem cell expansion.
We report on a forward RNAi screen in primary human hematopoietic stem and progenitor cells,using pooled lentiviral shRNA libraries deconvoluted by next generation sequencing. We identify MAPK14/p38α as a modulator of ex vivo stem cell proliferation and show that pharmacologic inhibition of p38 dramatically enhances the stem cell activity of cultured umbilical cord blood derived hematopoietic cells. p38 inhibitors should thus be considered in strategies aiming at expanding stem cells for clinical benefit.
View Publication
产品类型:
产品号#:
72682
72684
产品名:
BIRB - 796
BIRB - 796
(Feb 2025)
Communications Biology 8
Aberrant choroid plexus formation drives the development of treatment-related brain toxicity
Brain tumors are commonly treated with radiotherapy,but the efficacy of the treatment is limited by its toxicity to the normal tissue including post-irradiation contrast enhanced lesions often linked to necrosis. The poorly understood mechanisms behind such brain lesions were studied using cerebral organoids. Here we show that irradiation of such organoids leads to dose-dependent growth retardation and formation of liquid-filled cavities but is not correlated with necrosis. Instead,the radiation-induced changes comprise of an enhancement of cortical hem markers,altered neuroepithelial stem cell differentiation,and an increase of ZO1+/AQP1+/CLDN3+-choroid plexus (CP)-like structures accompanied by an upregulation of IGF2 mRNA,known to be expressed in CP and cerebrospinal fluid. The altered differentiation is attributed to changes in the WNT/BMP signaling pathways. We conclude that aberrant CP formation can be involved in radiation-induced brain lesions providing additional strategies for possible countermeasures. Human cerebral organoids provide insights into mechanisms behind the formation of choroid plexus (CP)-like structures that may contribute to radiation-induced brain image changes.
View Publication
SummaryInterleukin-33 (IL-33) is an immunoregulatory cytokine that moderately suppresses experimental autoimmune encephalomyelitis (EAE),a murine model of multiple sclerosis (MS). However,poor pharmacokinetics and toxicity hinder its clinical translation. To address these limitations,we develop an activity-attenuated IL-33 by recombinant fusion to serum albumin (SA). SA-IL-33 exhibits reduced toxicity and prolonged residence in the secondary lymphoid organs (SLOs),sites of T cell priming in autoimmunity,compared to wild-type (WT) IL-33. Prophylactic SA-IL-33 administration prevents EAE with superior efficacy to WT IL-33 and comparable efficacy to fingolimod (FTY720),a Food and Drug Administration (FDA)-approved MS drug. Therapeutic SA-IL-33 treatment also reduces disease severity in both chronic and relapsing-remitting EAE. SA-IL-33 modulates immunity in EAE by suppressing CD45+ cell infiltration (including myelin-reactive T helper 17 [TH17] cells) in the spinal cord,while expanding type 2 immune cells (including type 2 innate lymphoid cells [ILC2s],ST2+ regulatory T cells [Tregs],T helper 2 [TH2] cells,and M2-polarized macrophages) in the SLOs. These findings suggest that SA-IL-33 is a promising therapeutic for neuroinflammatory diseases. Graphical abstract Highlights•Fusion of serum albumin (SA) to interleukin-33 (IL-33) attenuates its activity and toxicity•Engineered SA-IL-33 exhibits prolonged residence in the secondary lymphoid organs (SLOs)•SA-IL-33 treatment both prevents the onset of and reduces established neuroinflammation in mice•Cytokine therapy suppresses TH17 cells in the CNS and promotes immunoregulation in the SLOs The clinical utility of interleukin-33 is hindered by poor pharmacokinetics and toxicity. Budina et al. develop a fusion of serum albumin and interleukin-33 (SA-IL-33) with reduced toxicity and prolonged lymph node residence. SA-IL-33 prevents the onset of and suppresses established inflammation-mediated paralysis in mice,demonstrating promise as a therapeutic for neuroinflammatory diseases.
View Publication