Scientific Resources
-
文献M. Li\'egeois et al. (aug 2022) American journal of respiratory cell and molecular biology 67 2 241--252
Airway Macrophages Encompass Transcriptionally and Functionally Distinct Subsets Altered by Smoking.
Alveolar macrophages (AMs) are functionally important innate cells involved in lung homeostasis and immunity and whose diversity in health and disease is a subject of intense investigations. Yet,it remains unclear to what extent conditions like smoking or chronic obstructive pulmonary disease (COPD) trigger changes in the AM compartment. Here,we aimed to explore heterogeneity of human AMs isolated from healthy nonsmokers,smokers without COPD,and smokers with COPD by analyzing BAL fluid cells by flow cytometry and bulk and single-cell RNA sequencing. We found that subpopulations of BAL fluid CD206+ macrophages could be distinguished based on their degree of autofluorescence in each subject analyzed. CD206+ autofluorescenthigh AMs were identified as classical,self-proliferative AM,whereas autofluorescentlow AMs were expressing both monocyte and classical AM-related genes,supportive of a monocytic origin. Of note,monocyte-derived autofluorescentlow AMs exhibited a functionally distinct immunoregulatory profile,including the ability to secrete the immunosuppressive cytokine IL-10. Interestingly,single-cell RNA-sequencing analyses showed that transcriptionally distinct clusters of classical and monocyte-derived AM were uniquely enriched in smokers with and without COPD as compared with healthy nonsmokers. Of note,such smoking-associated clusters exhibited gene signatures enriched in detoxification,oxidative stress,and proinflammatory responses. Our study independently confirms previous reports supporting that monocyte-derived macrophages coexist with classical AM in the airways of healthy subjects and patients with COPD and identifies smoking-associated changes in the AM compartment that may favor COPD initiation or progression. View Publication -
文献F. Qian et al. (may 2022) FASEB journal : official publication of the Federation of American Societies for Experimental Biology 36 5 e22328
Interleukin-4 treatment reduces leukemia burden in acute myeloid leukemia.
Interleukin-4 (IL-4) is a signature cytokine pivotal in Type 2 helper T cell (Th2) immune response,particularly in allergy and hypersensitivity. Interestingly,IL-4 increases endogenous levels of prostaglandin D2 (PGD2 ) and its metabolites,$\Delta$12 -prostaglandin J2 ($\Delta$12 -PGJ2 ) and 15-deoxy-$\Delta$12,14 -prostaglandin J2 (15d-PGJ2 ),collectively called cyclopentenone PGs (CyPGs). However,the therapeutic role of IL-4 in hematologic malignancies remains unclear. Here,we employed a murine model of acute myeloid leukemia (AML),where human MLL-AF9 fusion oncoprotein was expressed in hematopoietic progenitor cells,to test the effect of IL-4 treatment in vivo. Daily intraperitoneal treatment with IL-4 at 60 µg/kg/d significantly alleviated the severity of AML,as seen by decreased leukemia-initiating cells (LICs). The effect of IL-4 was mediated,in part,by the enhanced expression of hematopoietic- PGD2 synthase (H-PGDS) to effect endogenous production of CyPGs,through autocrine and paracrine signaling mechanisms. Similar results were seen with patient-derived AML cells cultured ex vivo with IL-4. Use of GW9662,a peroxisome proliferator-activated receptor gamma (PPAR$\gamma$) antagonist,suggested endogenous CyPGs-PPAR$\gamma$ axis mediated p53-dependent apoptosis of LICs by IL-4. Taken together,our results reveal a beneficial role of IL-4 treatment in AML suggesting a potential therapeutic regimen worthy of clinical trials in patients with AML. View Publication -
文献C. Mirabelli et al. (apr 2022) mBio 13 2 e0017522
Human Norovirus Triggers Primary B Cell Immune Activation In Vitro.
Human norovirus (HNoV) is a global health and socioeconomic burden,estimated to infect every individual at least five times during their lifetime. The underlying mechanism for the potential lack of long-term immune protection from HNoV infections is not understood and prompted us to investigate HNoV susceptibility of primary human B cells and its functional impact. Primary B cells isolated from whole blood were infected with HNoV-positive stool samples and harvested at 3??days postinfection (dpi) to assess the viral RNA yield by reverse transcriptase quantitative PCR (RT-qPCR). A 3- to 18-fold increase in the HNoV RNA yield was observed in 50 to 60% of donors. Infection was further confirmed in B cells derived from splenic and lymph node biopsy specimens. Next,we characterized infection of whole-blood-derived B cells by flow cytometry in specific functional B cell subsets (naive CD27- IgD+,memory-switched CD27+ IgD-,memory-unswitched CD27+ IgD+,and double-negative CD27- IgD- cells). While the susceptibilities of the subsets were similar,changes in the B cell subset distribution upon infection were observed,which were also noted after treatment with HNoV virus-like particles and the predicted recombinant NS1 protein. Importantly,primary B cell stimulation with the predicted recombinant NS1 protein triggered B cell activation and induced metabolic changes. These data demonstrate that primary B cells are susceptible to HNoV infection and suggest that the NS1 protein can alter B cell activation and metabolism in vitro,which could have implications for viral pathogenesis and immune responses in vivo. IMPORTANCE Human norovirus (HNoV) is the most prevalent causative agent of gastroenteritis worldwide. Infection results in a self-limiting disease that can become chronic and severe in the immunocompromised,the elderly,and infants. There are currently no approved therapeutic and preventative strategies to limit the health and socioeconomic burdens associated with HNoV infections. Moreover,HNoV does not elicit lifelong immunity as repeat infections are common,presenting a challenge for vaccine development. Given the importance of B cells for humoral immunity,we investigated the susceptibility and impact of HNoV infection on human B cells. We found that HNoV replicates in human primary B cells derived from blood,spleen,and lymph node specimens,while the nonstructural protein NS1 can activate B cells. Because of the secreted nature of NS1,we put forward the hypothesis that HNoV infection can modulate bystander B cell function with potential impacts on systemic immune responses. View Publication -
文献A. Revenko et al. (apr 2022) Journal for immunotherapy of cancer 10 4
Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer.
BACKGROUND The Regulatory T cell (Treg) lineage is defined by the transcription factor FOXP3,which controls immune-suppressive gene expression profiles. Tregs are often recruited in high frequencies to the tumor microenvironment where they can suppress antitumor immunity. We hypothesized that pharmacological inhibition of FOXP3 by systemically delivered,unformulated constrained ethyl-modified antisense oligonucleotides could modulate the activity of Tregs and augment antitumor immunity providing therapeutic benefit in cancer models and potentially in man. METHODS We have identified murine Foxp3 antisense oligonucleotides (ASOs) and clinical candidate human FOXP3 ASO AZD8701. Pharmacology and biological effects of FOXP3 inhibitors on Treg function and antitumor immunity were tested in cultured Tregs and mouse syngeneic tumor models. Experiments were controlled by vehicle and non-targeting control ASO groups as well as by use of multiple independent FOXP3 ASOs. Statistical significance of biological effects was evaluated by one or two-way analysis of variance with multiple comparisons. RESULTS AZD8701 demonstrated a dose-dependent knockdown of FOXP3 in primary Tregs,reduction of suppressive function and efficient target downregulation in humanized mice at clinically relevant doses. Surrogate murine FOXP3 ASO,which efficiently downregulated Foxp3 messenger RNA and protein levels in primary Tregs,reduced Treg suppressive function in immune suppression assays in vitro. FOXP3 ASO promoted more than 70% reduction in FOXP3 levels in Tregs in vitro and in vivo,strongly modulated Treg effector molecules (eg,ICOS,CTLA-4,CD25 and 4-1BB),and augmented CD8+ T cell activation and produced antitumor activity in syngeneic tumor models. The combination of FOXP3 ASOs with immune checkpoint blockade further enhanced antitumor efficacy. CONCLUSIONS Antisense inhibitors of FOXP3 offer a promising novel cancer immunotherapy approach. AZD8701 is being developed clinically as a first-in-class FOXP3 inhibitor for the treatment of cancer currently in Ph1a/b clinical trial (NCT04504669). View Publication -
文献D. J. Friedman et al. (apr 2022) Journal of immunology (Baltimore,Md. : 1950) 208 8 1845--1850
Cutting Edge: Enhanced Antitumor Immunity in ST8Sia6 Knockout Mice.
Inhibitory receptors have a critical role in the regulation of immunity. Siglecs are a family of primarily inhibitory receptors expressed by immune cells that recognize specific sialic acid modifications on cell surface glycans. Many tumors have increased sialic acid incorporation. Overexpression of the sialyltransferase ST8Sia6 on tumors led to altered immune responses and increased tumor growth. In this study,we examined the role of ST8Sia6 on immune cells in regulating antitumor immunity. ST8Sia6 knockout mice had an enhanced immune response to tumors. The loss of ST8Sia6 promoted an enhanced intratumoral activation of macrophages and dendritic cells,including upregulation of CD40. Intratumoral regulatory T cells exhibited a more inflammatory phenotype in ST8Sia6 knockout mice. Using adoptive transfer studies,the change in regulatory T cell phenotype was not cell intrinsic and depended on the loss of ST8Sia6 expression in APCs. Thus,ST8Sia6 generates ligands for Siglecs that dampen antitumor immunity. View Publication -
文献S. Tahtinen et al. (apr 2022) Nature immunology 23 4 532--542
IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines.
The use of lipid-formulated RNA vaccines for cancer or COVID-19 is associated with dose-limiting systemic inflammatory responses in humans that were not predicted from preclinical studies. Here,we show that the 'interleukin 1 (IL-1)-interleukin 1 receptor antagonist (IL-1ra)' axis regulates vaccine-mediated systemic inflammation in a host-specific manner. In human immune cells,RNA vaccines induce production of IL-1 cytokines,predominantly IL-1$\beta$,which is dependent on both the RNA and lipid formulation. IL-1 in turn triggers the induction of the broad spectrum of pro-inflammatory cytokines (including IL-6). Unlike humans,murine leukocytes respond to RNA vaccines by upregulating anti-inflammatory IL-1ra relative to IL-1 (predominantly IL-1$\alpha$),protecting mice from cytokine-mediated toxicities at >1,000-fold higher vaccine doses. Thus,the IL-1 pathway plays a key role in triggering RNA vaccine-associated innate signaling,an effect that was unexpectedly amplified by certain lipids used in vaccine formulations incorporating N1-methyl-pseudouridine-modified RNA to reduce activation of Toll-like receptor signaling. View Publication -
文献R. Bitsch et al. (mar 2022) Journal for immunotherapy of cancer 10 3
STAT3 inhibitor Napabucasin abrogates MDSC immunosuppressive capacity and prolongs survival of melanoma-bearing mice.
BACKGROUND Myeloid-derived suppressor cells (MDSCs) represent a negative prognostic factor in malignant melanoma. These cells are generated under chronic inflammatory conditions typical of cancer. The transcription factor signal transducer and activator of transcription 3 (STAT3) orchestrates MDSC accumulation and acquisition of immunosuppressive properties. Here we studied STAT3 inhibition by Napabucasin as a way to block MDSC accumulation and activity and its potential to treat malignant melanoma. METHODS In vitro generated murine MDSC and primary MDSC from melanoma-bearing mice were used to investigate the effects of Napabucasin on MDSC in vitro. The RET transgenic mouse model of malignant melanoma was used to examine Napabucasin therapy efficiency and its underlying mechanisms in vivo. Furthermore,STAT3 activation and its correlation with survival were explored in MDSC from 19 patients with malignant melanoma and human in vitro generated monocytic myeloid-derived suppressor cell (M-MDSC) were used to evaluate the effects of Napabucasin. RESULTS Napabucasin was able to abrogate the capacity of murine MDSC to suppress CD8+ T-cell proliferation. The STAT3 inhibitor induced apoptosis in murine MDSC,significantly increased expression of molecules associated with antigen processing and presentation,as well as slightly decreased expression of immunosuppressive factors on these cells. RET transgenic mice treated with Napabucasin showed prolonged survival accompanied by a strong accumulation of tumor-infiltrating antigen-presenting cells and activation of CD8+ and CD4+ T cells. Interestingly,patients with malignant melanoma with high expression of activated STAT3 in circulating M-MDSC showed significantly worse progression-free survival (PFS) than patients with low levels of activated STAT3. In addition,Napabucasin was able to abrogate suppressive capacity of human in vitro generated M-MDSC. CONCLUSION Our findings demonstrate that STAT3 inhibitor Napabucasin completely abrogated the immunosuppressive capacity of murine MDSC and human M-MDSC and improved melanoma-bearing mouse survival. Moreover,patients with malignant melanoma with high expression levels of activated STAT3 in M-MDSC displayed shorter PFS,indicating its role as a promising therapeutic target in patients with malignant melanoma and a predictive marker for their clinical outcome. View Publication -
文献Y. Shen et al. (mar 2022) Journal for immunotherapy of cancer 10 3
Cancer cell-intrinsic resistance to BiTE therapy is mediated by loss of CD58 costimulation and modulation of the extrinsic apoptotic pathway.
BACKGROUND Bispecific T-cell engager (BiTE) molecules induce redirected lysis of cancer cells by T cells and are an emerging modality for solid tumor immunotherapy. While signs of clinical activity have been demonstrated,efficacy of T-cell engagers (TCEs) in solid tumors settings,molecular determinants of response,and underlying mechanisms of resistance to BiTE therapy require more investigation. METHODS To uncover cancer cell-intrinsic genetic modifiers of TCE-mediated cytotoxicity,we performed genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) loss-of-function and CRISPRa (CRISPR activation) gain-of-function screens using TCEs against two distinct tumor-associated antigens (TAAs). By using in vitro T-cell cytotoxicity assays and in vivo efficacy studies,we validated the roles of two common pathways identified in our screen,T-cell costimulation pathway and apoptosis pathway,as key modifiers of BiTE activity. RESULTS Our genetic screens uncovered TAAs-independent cancer cell-intrinsic genes with functions in autophagy,T-cell costimulation,the apoptosis pathway,chromatin remodeling,and cytokine signaling that altered responsiveness to BiTE-mediated killing. Notably,loss of CD58 (the ligand of the CD2 T-cell costimulatory receptor),a gene frequently altered in cancer,led to decreased TCE-mediated cytotoxicity,T-cell activation and antitumor efficacy in vitro and in vivo. Moreover,the effects of CD58 loss were synergistically compounded by concurrent loss of CD80/CD86 (ligands for the CD28 T-cell costimulatory receptor),whereas joint CD2 and CD28 costimulation additively enhanced TCE-mediated killing,indicating non-redundant costimulatory mechanisms between the two pathways. Additionally,loss of CFLAR (Caspase-8 and FADD Like Apoptosis Regulator),BCL2L1,and BID (BH3 Interacting Domain Death Agonist) induced profound changes in sensitivity to TCEs,indicating that key regulators of apoptosis,which are frequently altered in cancer,impact tumor responsiveness to BiTE therapy. CONCLUSIONS This study demonstrates that genetic alterations central to carcinogenesis and commonly detected in cancer samples lead to significant modulation of BiTE antitumor activity in vitro and in vivo,findings with relevance for a better understanding of patient responses to BiTE therapy and novel combinations that enhance TCE efficacy. View Publication -
文献A. Goral et al. ( 2022) Frontiers in immunology 13 781364
A Specific CD44lo CD25lo Subpopulation of Regulatory T Cells Inhibits Anti-Leukemic Immune Response and Promotes the Progression in a Mouse Model of Chronic Lymphocytic Leukemia.
Regulatory T cells (Tregs) are capable of inhibiting the proliferation,activation and function of T cells and play an important role in impeding the immune response to cancer. In chronic lymphocytic leukemia (CLL) a dysfunctional immune response and elevated percentage of effector-like phenotype Tregs have been described. In this study,using the Eµ-TCL1 mouse model of CLL,we evaluated the changes in the Tregs phenotype and their expansion at different stages of leukemia progression. Importantly,we show that Tregs depletion in DEREG mice triggered the expansion of new anti-leukemic cytotoxic T cell clones leading to leukemia eradication. In TCL1 leukemia-bearing mice we identified and characterized a specific Tregs subpopulation,the phenotype of which suggests its role in the formation of an immunosuppressive microenvironment,supportive for leukemia survival and proliferation. This observation was also confirmed by the gene expression profile analysis of these TCL1-specific Tregs. The obtained data on Tregs are consistent with those described so far,however,above all show that the changes in the Tregs phenotype described in CLL result from the formation of a specific,described in this study Tregs subpopulation. In addition,functional tests revealed the ability of Tregs to inhibit T cells that recognize model antigens expressed by leukemic cells. Moreover,inhibition of Tregs with a MALT1 inhibitor provided a therapeutic benefit,both as monotherapy and also when combined with an immune checkpoint inhibitor. Altogether,activation of Tregs appears to be crucial for CLL progression. View Publication -
文献E. Berenice Mart\'inez-Shio et al. (may 2022) Clinical and experimental immunology 208 1 83--94
Differentiation of circulating monocytes into macrophages with metabolically activated phenotype regulates inflammation in dyslipidemia patients.
Macrophages are mediators of inflammation having an important role in the pathogenesis of cardiovascular diseases. Recently,a pro-inflammatory subpopulation,known as metabolically activated macrophages (MMe),has been described in conditions of obesity and metabolic syndrome where they are known to release cytokines that can promote insulin resistance. Dyslipidemia represents an important feature in metabolic syndrome and corresponds to one of the main modifiable risk factors for the development of cardiovascular diseases. Circulating monocytes can differentiate into macrophages under certain conditions. They correspond to a heterogeneous population,which include inflammatory and anti-inflammatory subsets; however,there is a wide spectrum of phenotypes. Therefore,we decided to investigate whether the metabolic activated monocyte (MoMe) subpopulation is already present under dyslipidemia conditions. Secondly,we assessed whether different levels of cholesterol and triglycerides play a role in the polarization towards the metabolic phenotype (MMe) of macrophages. Our results indicate that MoMe cells are found in both healthy and dyslipidemia patients,with cells displaying the following metabolic phenotype: CD14varCD36+ABCA1+PLIN2+. Furthermore,the percentages of CD14++CD68+CD80+ pro-inflammatory monocytes are higher in dyslipidemia than in healthy subjects. When analysing macrophage differentiation,we observed that MMe percentages were higher in the dyslipidemia group than in healthy subjects. These MMe have the ability to produce high levels of IL-6 and the anti-inflammatory cytokine IL-10. Furthermore,ABCA1 expression in MMe correlates with LDL serum levels. Our study highlights the dynamic contributions of metabolically activated macrophages in dyslipidemia,which may have a complex participation in low-grade inflammation due to their pro- and anti-inflammatory function. View Publication -
文献D. Wang et al. (jun 2022) Immunology 166 2 169--184
CTLA4Ig/VISTAIg combination therapy selectively induces CD4+ T cell-mediated immune tolerance by targeting the SOCS1 signaling pathway in porcine islet xenotransplantation.
T cell inhibitory receptors can regulate the proliferation or function of T cells by binding to their ligands and present a unique opportunity to manage destructive immune responses during porcine islet xenotransplantation. We applied ex vivo porcine islet xenotransplantation and in vitro mixed lymphocyte-islet reaction models to assess immune checkpoint receptor expression profiles in recipient T cells,investigated whether CTLA4 or VISTA immunoglobulin (Ig) combination therapy alone could suppress porcine islet xenograft rejection and further analyzed its potential immune tolerance mechanism. Recipient T cells expressed moderate to high levels of CTLA4,PD-1,TIGIT and VISTA,and the frequency of CTLA4+ CD4+,TIGIT+ CD4+,VISTA+ CD4+ and VISTA+ CD8+ T cells was positively correlated with porcine islet xenograft survival time in xenotransplant recipients. Combined treatment with CTLA4Ig and VISTAIg selectively inhibited recipient CD4+ T cell hyper-responsiveness and proinflammatory cytokine production and significantly delayed xenograft rejection. SOCS1 deficiency in CD4+ T cells stimulated by xenogeneic islets facilitated hyper-responsiveness and abolished the suppressive effect of combination therapy on recipient T cell-mediated porcine islet damage in vivo and in vitro. Further mechanistic studies revealed that combined treatment significantly induced SOCS1 expression and inhibited the Jak-STAT signalling pathway in wild-type recipient CD4+ T cells stimulated by xenogeneic islets,whereas SOCS1 deficiency resulted in Jak-STAT signalling pathway activation in recipient CD4+ T cells. We demonstrated a major role for CTLA4 and VISTA as key targets in CD4+ T cell hyper-responsiveness and porcine islet xenograft rejection. The selective inhibition of CD4+ T cell immunity by CTLA4Ig/VISTAIg is based on SOCS1-dependent signalling. View Publication -
文献Y. Hong et al. (mar 2022) JCI insight 7 5
Cure of syngeneic carcinomas with targeted IL-12 through obligate reprogramming of lymphoid and myeloid immunity.
Therapeutic IL-12 has demonstrated the ability to reduce local immune suppression in preclinical models,but clinical development has been limited by severe inflammation-related adverse events with systemic administration. Here,we show that potent immunologic tumor control of established syngeneic carcinomas can be achieved by i.t. administration of a tumor-targeted IL-12 antibody fusion protein (NHS-rmIL-12) using sufficiently low doses to avoid systemic toxicity. Single-cell transcriptomic analysis and ex vivo functional assays of NHS-rmIL-12-treated tumors revealed reinvigoration and enhanced proliferation of exhausted CD8+ T lymphocytes,induction of Th1 immunity,and a decrease in Treg number and suppressive capacity. Similarly,myeloid cells transitioned toward inflammatory phenotypes and displayed reduced suppressive capacity. Cell type-specific IL-12 receptor-KO BM chimera studies revealed that therapeutic modulation of both lymphoid and myeloid cells is required for maximum treatment effect and tumor cure. Study of single-cell data sets from human head and neck carcinomas revealed IL-12 receptor expression patterns similar to those observed in murine tumors. These results describing the diverse mechanisms underlying tumor-directed IL-12-induced antitumor immunity provide the preclinical rationale for the clinical study of i.t. NHS-IL-12. View Publication
过滤器
筛选结果
品牌
- ALDECOUNT 9 项目
- CellPore 8 项目
- CellSTACK 1 项目
- Corning 1 项目
- EasyPick 2 项目
- ELISA 2 项目
- ErythroClear 3 项目
- ES-Cult 95 项目
- Falcon 1 项目
- GloCell 2 项目
- GyneCult 2 项目
- HetaSep 2 项目
- iCell 14 项目
- Maestro 4 项目
- Matrigel 3 项目
- MegaCult 38 项目
- STEMgrid 1 项目
- STEMprep 2 项目
- ALDEFLUOR 231 项目
- AggreWell 68 项目
- ArciTect 35 项目
- BloodStor 2 项目
- BrainPhys 49 项目
- CellAdhere 2 项目
- ClonaCell 93 项目
- CloneR 9 项目
- CryoStor 78 项目
- EC-Cult 3 项目
- EasySep 741 项目
- EpiCult 13 项目
- HemaTox 7 项目
- HepatiCult 23 项目
- ImmunoCult 54 项目
- IntestiCult 128 项目
- Lymphoprep 24 项目
- MammoCult 55 项目
- MesenCult 105 项目
- MethoCult 518 项目
- MyeloCult 80 项目
- MyoCult 9 项目
- NaïveCult 1 项目
- NeuroCult 360 项目
- NeuroFluor 4 项目
- PBS-MINI 11 项目
- PancreaCult 19 项目
- PneumaCult 86 项目
- RSeT 10 项目
- ReLeSR 5 项目
- RoboSep 99 项目
- RosetteSep 281 项目
- STEMdiff 189 项目
- STEMscript 1 项目
- STEMvision 27 项目
- SepMate 47 项目
- SmartDish 11 项目
- StemSpan 327 项目
- TeSR 1676 项目
- ThawSTAR 10 项目
- mFreSR 35 项目
产品类型
- Antibodies 2 项目
- Cell Culture Media and Supplements 350 项目
- Cell Dyes and Detection Assay Kits 11 项目
- Cell Engineering and Molecular Tools 22 项目
- Cell Isolation Products 106 项目
- Cell Storage Media 2 项目
- Contract Services 6 项目
- Cultureware and General Supplies 2 项目
- Cytokines and Proteins 3 项目
- Density Gradient Media 1 项目
- Instruments and Software 11 项目
- Laboratory Equipment 2 项目
- Matrices and Substrates 1 项目
- Primary and Cultured Cells 33 项目
- Small Molecules 1 项目
- Standardization Tools 5 项目
- Tissue and Cell Culture Dissociation Reagents 12 项目
- Training and Education 29 项目
- ELISAs 1 项目
资源类别
细胞类型
- B 细胞 182 项目
- Cardiomyocytes 21 项目
- CD4+ 121 项目
- CD8+ 92 项目
- CHO细胞 3 项目
- Endoderm 18 项目
- Endothelial Cells 12 项目
- Epithelial Cells 29 项目
- HEK-293细胞(人胚肾293细胞) 1 项目
- Hematopoietic Cells 22 项目
- Hepatic Cells 13 项目
- HUVEC细胞(人脐静脉内皮细胞) 1 项目
- Mesenchymal Cells 18 项目
- Mesoderm 18 项目
- Neural Cells 89 项目
- NK 细胞 121 项目
- Other Subsets 21 项目
- PSC-Derived 128 项目
- PSC衍生 27 项目
- Regulatory 34 项目
- T Cells 102 项目
- T 细胞 352 项目
- 上皮细胞 106 项目
- 中胚层 1 项目
- 乳腺细胞 74 项目
- 先天性淋巴细胞 23 项目
- 全血 6 项目
- 内皮细胞 8 项目
- 内皮集落形成细胞(ECFCs) 3 项目
- 前列腺细胞 8 项目
- 单个核细胞 73 项目
- 单核细胞 142 项目
- 多巴胺能神经元 3 项目
- 多能干细胞 1859 项目
- 小胶质细胞 3 项目
- 巨噬细胞 25 项目
- 巨核细胞 8 项目
- 心肌细胞 15 项目
- 成骨细胞 6 项目
- 星形胶质细胞 2 项目
- 杂交瘤细胞 83 项目
- 树突状细胞(DCs) 91 项目
- 气道细胞 73 项目
- 淋巴细胞 33 项目
- 癌细胞及细胞系 130 项目
- 白细胞单采样本 12 项目
- 白血病/淋巴瘤细胞 14 项目
- 真皮细胞 2 项目
- 神经元 165 项目
- 神经干/祖细胞 420 项目
- 神经细胞 6 项目
- 粒细胞及其亚群 76 项目
- 红系细胞 9 项目
- 肌源干/祖细胞 9 项目
- 肝细胞 25 项目
- 肠道细胞 61 项目
- 肾细胞 3 项目
- 肾脏细胞 4 项目
- 肿瘤细胞 11 项目
- 胰腺细胞 12 项目
- 脂肪细胞 6 项目
- 脑肿瘤干细胞 87 项目
- 血小板 4 项目
- 血浆 16 项目
- 血管生成细胞 2 项目
- 调节性细胞 9 项目
- 软骨细胞 7 项目
- 造血干/祖细胞 875 项目
- 间充质基质细胞 13 项目
- 间充质干/祖细胞 156 项目
- 间充质细胞 1 项目
- 骨髓基质细胞 2 项目
- 骨髓瘤细胞 4 项目
- 髓系细胞 116 项目
- 鼠胚胎成纤维细胞 1 项目
- 白细胞 9 项目
- 其它细胞系 5 项目
- 红细胞 10 项目
研究方向
种属