Product Information
Items 2065 to 2076 of 13914 total
- Reference(May 2025) Brain Communications 7 3
Evidence that minocycline treatment confounds the interpretation of neurofilament as a biomarker
AbstractNeurofilament light (NfL) concentration in CSF and blood serves as an important biomarker in neurology drug development. Changes in NfL are generally assumed to reflect changes in neuronal damage, while little is known about the clearance of NfL from biofluids. In a study of asymptomatic individuals at risk for prion disease, both blood and CSF NfL spiked in one participant following a 6-week course of minocycline, absent any other biomarker changes and without subsequent onset of symptoms. We subsequently observed high NfL after minocycline treatment in discarded clinical plasma samples from inpatients, in mouse plasma and in conditioned media from neuron–microglia co-cultures. The specificity and kinetics of NfL response lead us to hypothesize that minocycline does not cause or exacerbate neuronal damage, but instead affects NfL by inhibiting its clearance, posing a potential confounder for the interpretation of this important biomarker. Gentile et al. report that treatment with the drug minocycline may cause neurofilament light concentration to rise in CSF and blood. This effect appears mediated by changes in clearance of the protein, rather than release from tissue, confounding this biomarker normally held to report on neuronal health. Graphical Abstract Graphical AbstractCatalog #: Product Name: 85850 ³¾°Õ±ð³§¸éâ„¢1 34811 ´¡²µ²µ°ù±ð°Â±ð±ô±ôâ„¢800 Catalog #: 85850 Product Name: ³¾°Õ±ð³§¸éâ„¢1 Catalog #: 34811 Product Name: ´¡²µ²µ°ù±ð°Â±ð±ô±ôâ„¢800 Reference(Apr 2025) PLOS One 20 4A human iPSC-derived midbrain neural stem cell model of prenatal opioid exposure and withdrawal: A proof of concept study
A growing body of clinical literature has described neurodevelopmental delays in infants with chronic prenatal opioid exposure and withdrawal. Despite this, the mechanism of how opioids impact the developing brain remains unknown. Here, we developed an in vitro model of prenatal morphine exposure and withdrawal using healthy human induced pluripotent stem cell (iPSC)-derived midbrain neural progenitors in monolayer. To optimize our model, we identified that a longer neural induction and regional patterning period increases expression of canonical opioid receptors mu and kappa in midbrain neural progenitors compared to a shorter protocol (OPRM1, two-tailed t-test, p =? 0.004; OPRK1, p =? 0.0003). Next, we showed that the midbrain neural progenitors derived from a longer iPSC neural induction also have scant toll-like receptor 4 (TLR4) expression, a key player in neonatal opioid withdrawal syndrome pathophysiology. During morphine withdrawal, differentiating neural progenitors experience cyclic adenosine monophosphate overshoot compared to cell exposed to vehicle (p =? 0.0496) and morphine exposure conditions (p, =? 0.0136, 1-way ANOVA). Finally, we showed that morphine exposure and withdrawal alters proportions of differentiated progenitor cell fates (2-way ANOVA, F =? 16.05, p <? 0.0001). Chronic morphine exposure increased proportions of nestin positive progenitors (p =? 0.0094), and decreased proportions of neuronal nuclear antigen positive neurons (NEUN) (p =? 0.0047) compared to those exposed to vehicle. Morphine withdrawal decreased proportions of glial fibrillary acidic protein positive cells of astrocytic lineage (p =? 0.044), and increased proportions of NEUN-positive neurons (p <? 0.0001) compared to those exposed to morphine only. Applications of this paradigm include mechanistic studies underscoring neural progenitor cell fate commitments in early neurodevelopment during morphine exposure and withdrawal.Catalog #: Product Name: 05854 ³¾¹ó°ù±ð³§¸éâ„¢ 05872 ¸é±ð³¢±ð³§¸éâ„¢ 100-0276 mTeSRâ„¢ Plus Catalog #: 05854 Product Name: ³¾¹ó°ù±ð³§¸éâ„¢ Catalog #: 05872 Product Name: ¸é±ð³¢±ð³§¸éâ„¢ Catalog #: 100-0276 Product Name: mTeSRâ„¢ Plus Reference(Oct 2024) bioRxiv 170 2Independent control of neurogenesis and dorsoventral patterning by NKX2-2
SUMMARYHuman neurogenesis is disproportionately protracted, lasting >10 times longer than in mouse, allowing neural progenitors to undergo more rounds of self-renewing cell divisions and generate larger neuronal populations. In the human spinal cord, expansion of the motor neuron lineage is achieved through a newly evolved progenitor domain called vpMN (ventral motor neuron progenitor) that uniquely extends and expands motor neurogenesis. This behavior of vpMNs is controlled by transcription factor NKX2-2, which in vpMNs is co-expressed with classical motor neuron progenitor (pMN) marker OLIG2. In this study, we sought to determine the molecular basis of NKX2-2-mediated extension and expansion of motor neurogenesis. We found that NKX2-2 represses proneural gene NEUROG2 by two distinct, Notch-independent mechanisms that are respectively apparent in rodent and human spinal progenitors: in rodents (and chick), NKX2-2 represses Olig2 and the motor neuron lineage through its tinman domain, leading to loss of Neurog2 expression. In human vpMNs, however, NKX2-2 represses NEUROG2 but not OLIG2, thereby allowing motor neurogenesis to proceed, albeit in a delayed and protracted manner. Interestingly, we found that ectopic expression of tinman-mutant Nkx2-2 in mouse pMNs phenocopies human vpMNs, repressing Neurog2 but not Olig2, and leading to delayed and protracted motor neurogenesis. Our studies identify a Notch- and tinman-independent mode of Nkx2-2-mediated Neurog2 repression that is observed in human spinal progenitors, but is normally masked in rodents and chicks due to Nkx2-2’s tinman-dependent repression of Olig2.Catalog #: Product Name: 100-0276 mTeSR™ Plus Catalog #: 100-0276 Product Name: mTeSR™ Plus Reference(Jan 2025) Nature Communications 16Mitotic chromatin marking governs the segregation of DNA damage
The faithful segregation of intact genetic material and the perpetuation of chromatin states through mitotic cell divisions are pivotal for maintaining cell function and identity across cell generations. However, most exogenous mutagens generate long-lasting DNA lesions that are segregated during mitosis. How this segregation is controlled is unknown. Here, we uncover a mitotic chromatin-marking pathway that governs the segregation of UV-induced damage in human cells. Our mechanistic analyses reveal two layers of control: histone ADP-ribosylation, and the incorporation of newly synthesized histones at UV damage sites, that both prevent local mitotic phosphorylations on histone H3 serine residues. Functionally, this chromatin-marking pathway controls the segregation of UV damage in the cell progeny with consequences on daughter cell fate. We propose that this mechanism may help preserve the integrity of stem cell compartments during asymmetric cell divisions. The transmission of unrepaired DNA lesions through mitosis needs tight control. Here, the authors uncover a damaged chromatin marking mechanism driving the segregation of UV damage through mitosis with potential consequences on daughter cell fate.Catalog #: Product Name: 100-0276 mTeSRâ„¢ Plus Catalog #: 100-0276 Product Name: mTeSRâ„¢ Plus Reference(Jan 2025) Burns & Trauma 13The empowering influence of air-liquid interface culture on skin organoid hair follicle development
AbstractBackgroundRodent models have been widely used to investigate skin development, but do not account for significant differences in composition compared to human skin. On the other hand, two-dimensional and three-dimensional engineered skin models still lack the complex features of human skin such as appendages and pigmentation. Recently, hair follicle containing skin organoids (SKOs) with a stratified epidermis, and dermis layer have been generated as floating spheres from human-induced pluripotent stem cells (hiPSCs).MethodsThe current study aims to investigate the generation of hiPSCs-derived SKOs using an air-liquid interface (ALI) model on transwell membranes (T-SKOs) and compares their development with conventional floating culture in low-attachment plates (F-SKOs).ResultsMature SKOs containing an epidermis, dermis, and appendages are created in both T-SKO and F-SKO conditions. It was found that the hair follicles are smaller and shorter in the F-SKO compared with T-SKOs. Additionally, the ALI conditions contribute to enhanced hair follicle numbers than conventional floating culture.ConclusionsTogether, this study demonstrates the significant influence of transwell culture on the morphogenesis of hair follicles within SKOs and highlights the potential for refinement of skin model engineering for advancing dermatology and skin research.Catalog #: Product Name: 100-0276 mTeSRâ„¢ Plus Catalog #: 100-0276 Product Name: mTeSRâ„¢ Plus Reference(Jul 2024) bioRxiv 4 3Cryopreservation of neuroectoderm on a pillar plate and
Cryopreservation in cryovials extends cell storage at low temperatures, and advances in organoid cryopreservation improve reproducibility and reduce generation time. However, cryopreserving human organoids presents challenges due to the limited diffusion of cryoprotective agents (CPAs) into the organoid core and the potential toxicity of these agents. To overcome these obstacles, we developed a cryopreservation technique using a pillar plate platform. To illustrate cryopreservation application to human brain organoids (HBOs), early-stage HBOs were produced by differentiating induced pluripotent stem cells (iPSCs) into neuroectoderm (NEs) in an ultralow atachement (ULA) 384-well plate. These NEs were transferred and encapsulated in Matrigel on the pillar plate. The early-stage HBOs on the pillar plate were exposed to four commercially available CPAs, including PSC cryopreservation kit, CryoStor CS10, 3dGRO, and 10% DMSO, before being frozen overnight at ?80°C and subsequently stored in a liquid nitrogen dewar. We examined the impact of CPA type, organoid size, and CPA exposure duration on cell viability post-thaw. Additionally, the differentiation of early-stage HBOs on the pillar plate was assessed using RT-qPCR and immunofluorescence staining. The PSC cryopreservation kit proved to be the least toxic for preserving these HBOs on the pillar plate. Notably, smaller HBOs showed higher cell viability post-cryopreservation than larger ones. An incubation period of 80 minutes with the PSC kit was essential to ensure optimal CPA diffusion into HBOs with a diameter of 400 – 600 ?m. These cryopreserved early-stage HBOs successfully matured over 30 days, exhibiting gene expression patterns akin to non-cryopreserved HBOs. The cryopreserved early-stage HBOs on the pillar plate maintained high viability after thawing and successfully differentiated into mature HBOs. This on-chip cryopreservation method could extend to other small organoids, by integrating cryopreservation, thawing, culturing, staining, rinsing, and imaging processes within a single system, thereby preserving the 3D structure of the organoids.Catalog #: Product Name: 100-0276 mTeSR™ Plus Catalog #: 100-0276 Product Name: mTeSR™ Plus Reference(Dec 2024) Life Science Alliance 8 2RuvBL1/2 reduce toxic dipeptide repeat protein burden in multiple models of C9orf72-ALS/FTD
Enhancing RuvBL1, but particularly RuvBL2 expression, reduces toxic dipeptide repeat proteins in vitro and in vivo models of C9orf72-linked ALS/FTD, suggesting that modulating RuvBL1/2 levels could be a promising therapeutic approach for C9ALS/FTD. A G4C2 hexanucleotide repeat expansion in C9orf72 is the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Bidirectional transcription and subsequent repeat-associated non-AUG (RAN) translation of sense and antisense transcripts leads to the formation of five dipeptide repeat (DPR) proteins. These DPRs are toxic in a wide range of cell and animal models. Therefore, decreasing RAN-DPRs may be of therapeutic benefit in the context of C9ALS/FTD. In this study, we found that C9ALS/FTD patients have reduced expression of the AAA+ family members RuvBL1 and RuvBL2, which have both been implicated in aggregate clearance. We report that overexpression of RuvBL1, but to a greater extent RuvBL2, reduced C9orf72-associated DPRs in a range of in vitro systems including cell lines, primary neurons from the C9-500 transgenic mouse model, and patient-derived iPSC motor neurons. In vivo, we further demonstrated that RuvBL2 overexpression and consequent DPR reduction in our Drosophila model was sufficient to rescue a number of DPR-related motor phenotypes. Thus, modulating RuvBL levels to reduce DPRs may be of therapeutic potential in C9ALS/FTD.Catalog #: Product Name: 05872 ¸é±ð³¢±ð³§¸éâ„¢ 100-0276 mTeSRâ„¢ Plus Catalog #: 05872 Product Name: ¸é±ð³¢±ð³§¸éâ„¢ Catalog #: 100-0276 Product Name: mTeSRâ„¢ Plus Reference(Apr 2024) The Journal of Experimental Medicine 221 6A de novo dominant-negative variant is associated with OTULIN-related autoinflammatory syndrome
This study describes an OTULIN-related autoinflammatory syndrome (ORAS) patient with two rare heterozygous variants of OTULIN (p.P152L and p.R306Q); the latter is a de novo variant that acts in a dominant-negative manner to cause ORAS. OTULIN-related autoinflammatory syndrome (ORAS), a severe autoinflammatory disease, is caused by biallelic pathogenic variants of OTULIN, a linear ubiquitin-specific deubiquitinating enzyme. Loss of OTULIN attenuates linear ubiquitination by inhibiting the linear ubiquitin chain assembly complex (LUBAC). Here, we report a patient who harbors two rare heterozygous variants of OTULIN (p.P152L and p.R306Q). We demonstrated accumulation of linear ubiquitin chains upon TNF stimulation and augmented TNF-induced cell death in mesenchymal stem cells differentiated from patient-derived iPS cells, which confirms that the patient has ORAS. However, although the de novo p.R306Q variant exhibits attenuated deubiquitination activity without reducing the amount of OTULIN, the deubiquitination activity of the p.P152L variant inherited from the mother was equivalent to that of the wild-type. Patient-derived MSCs in which the p.P152L variant was replaced with wild-type also exhibited augmented TNF-induced cell death and accumulation of linear chains. The finding that ORAS can be caused by a dominant-negative p.R306Q variant of OTULIN furthers our understanding of disease pathogenesis. Graphical AbstractCatalog #: Product Name: 85850 ³¾°Õ±ð³§¸éâ„¢1 05230 STEMdiffâ„¢ Trilineage Differentiation Kit Catalog #: 85850 Product Name: ³¾°Õ±ð³§¸éâ„¢1 Catalog #: 05230 Product Name: STEMdiffâ„¢ Trilineage Differentiation Kit Reference(Sep 2024) Biology Open 13 9High mobility group box 1 (HMGB1) is a potential disease biomarker in cell and mouse models of Duchenne muscular dystrophy
ABSTRACTDuchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder affecting 1:3500 male births and is associated with myofiber degeneration, regeneration, and inflammation. Glucocorticoid treatments have been the standard of care due to immunomodulatory/immunosuppressive properties but novel genetic approaches, including exon skipping and gene replacement therapy, are currently being developed. The identification of additional biomarkers to assess DMD-related inflammatory responses and the potential efficacy of these therapeutic approaches are thus of critical importance. The current study uses RNA sequencing of skeletal muscle from two mdx mouse models to identify high mobility group box 1 (HMGB1) as a candidate biomarker potentially contributing to DMD-related inflammation. HMGB1 protein content was increased in a human iPSC-derived skeletal myocyte model of DMD and microdystrophin treatment decreased HMGB1 back to control levels. In vivo, HMGB1 protein levels were increased in vehicle treated B10-mdx skeletal muscle compared to B10-WT and significantly decreased in B10-mdx animals treated with adeno-associated virus (AAV)-microdystrophin. However, HMGB1 protein levels were not increased in D2-mdx skeletal muscle compared to D2-WT, demonstrating a strain-specific difference in DMD-related immunopathology. Summary: Duchenne muscular dystrophy is a devastating that currently has limited treatment options. RNA sequencing and downstream analysis in iSkM and mdx samples revealed HMGB1 may be a relevant treatment biomarker.Catalog #: Product Name: 85850 ³¾°Õ±ð³§¸éâ„¢1 Catalog #: 85850 Product Name: ³¾°Õ±ð³§¸éâ„¢1 Reference(Jan 2025) Nature Communications 16Engineering synthetic signaling receptors to enable erythropoietin-free erythropoiesis
Blood transfusion plays a vital role in modern medicine, but frequent shortages occur. Ex vivo manufacturing of red blood cells (RBCs) from universal donor cells offers a potential solution, yet the high cost of recombinant cytokines remains a barrier. Erythropoietin (EPO) signaling is crucial for RBC development, and EPO is among the most expensive media components. To address this challenge, we develop highly optimized small molecule-inducible synthetic EPO receptors (synEPORs) using design-build-test cycles and genome editing. By integrating synEPOR at the endogenous EPOR locus in O-negative induced pluripotent stem cells, we achieve equivalent erythroid differentiation, transcriptomic changes, and hemoglobin production using small molecules compared to EPO-supplemented cultures. This approach dramatically reduces culture media costs. Our strategy not only addresses RBC production challenges but also demonstrates how protein and genome engineering can introduce precisely regulated cellular behaviors, potentially improving scalable manufacturing of a wide range of clinically relevant cell types. Shortages of donor blood for transfusions can have severe medical consequences, and ex vivo production of red blood cells offers a potential solution. Here authors developed synthetic EPO receptors, which allow erythropoiesis (red blood cell production) without the need for expensive EPO.Catalog #: Product Name: 05872 ¸é±ð³¢±ð³§¸éâ„¢ 09600 StemSpanâ„¢ SFEM 85850 ³¾°Õ±ð³§¸éâ„¢1 05310 STEMdiffâ„¢ Hematopoietic Kit Catalog #: 05872 Product Name: ¸é±ð³¢±ð³§¸éâ„¢ Catalog #: 09600 Product Name: StemSpanâ„¢ SFEM Catalog #: 85850 Product Name: ³¾°Õ±ð³§¸éâ„¢1 Catalog #: 05310 Product Name: STEMdiffâ„¢ Hematopoietic Kit Reference(Mar 2025) Communications Biology 8A chromatin-focused CRISPR screen identifies USP22 as a barrier to somatic cell reprogramming
Cell-autonomous barriers to reprogramming somatic cells into induced pluripotent stem cells (iPSCs) remain poorly understood. Using a focused CRISPR-Cas9 screen, we identified Ubiquitin-specific peptidase 22 (USP22) as a key chromatin-based barrier to human iPSC derivation. Suppression of USP22 significantly enhances reprogramming efficiency. Surprisingly, this effect is likely to be independent of USP22’s deubiquitinase activity or its association with the SAGA complex, as shown through module-specific knockouts, and genetic rescue experiments. USP22 is not required for iPSC derivation or maintenance. Mechanistically, USP22 loss during reprogramming downregulates fibroblast-specific genes while activating pluripotency-associated genes, including DNMT3L, LIN28A, SOX2, and GDF3. Additionally, USP22 loss enhances reprogramming efficiency under naïve stem cell conditions. These findings reveal an unrecognized role for USP22 in maintaining somatic cell identity and repressing pluripotency genes, highlighting its potential as a target to improve reprogramming efficiency. Ubiquitin-specific peptidase 22 (USP22) is identified as a key chromatin-based barrier to human iPSC derivation through a chromatin-focused CRISPR-Cas9 screen.Catalog #: Product Name: 05872 ¸é±ð³¢±ð³§¸éâ„¢ 100-0276 mTeSRâ„¢ Plus Catalog #: 05872 Product Name: ¸é±ð³¢±ð³§¸éâ„¢ Catalog #: 100-0276 Product Name: mTeSRâ„¢ Plus Reference(Apr 2025) Molecular Neurodegeneration 20 2Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model
BackgroundThe epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in A? mouse models of Alzheimer’s disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood.MethodsHere we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions.ResultssEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures.ConclusionThese findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.Supplementary InformationThe online version contains supplementary material available at 10.1186/s13024-025-00844-x.Catalog #: Product Name: 100-0276 mTeSR™ Plus Catalog #: 100-0276 Product Name: mTeSR™ Plus Items 2065 to 2076 of 13914 total
Shop ByFilter Results- Resource Type
-
- Product Information Sheet 2907 items
- Reference 7892 items
- Safety Data Sheet 3052 items
- Technical Manual 63 items
- Product Type
-
- 35 items
- Cell Culture Media and Supplements 27 items
- Cell Engineering and Molecular Tools 3 items
- Cell Isolation Products 5 items
- Instruments and Software 4 items
- Tissue and Cell Culture Dissociation Reagents 2 items
- Training and Education 1 item
- Area of Interest
-
- 29 items
- Angiogenic Cell Research 49 items
- Antibody Development 1 item
- Cancer 601 items
- Cell Line Development 137 items
- Cell Therapy Development 1 item
- Chimerism 5 items
- Cord Blood Banking 25 items
- Disease Modeling 4 items
- Drug Discovery and Toxicity Testing 182 items
- Endothelial Cell Biology 2 items
- Epithelial Cell Biology 158 items
- HIV 52 items
- HLA 8 items
- Hybridoma Generation 1 item
- Immunology 743 items
- Infectious Diseases 4 items
- Neuroscience 491 items
- Organoids 1 item
- Respiratory Research 1 item
- Stem Cell Biology 2493 items
- Transplantation Research 54 items
- Brand
-
- 0 20 items
- ALDECOUNT 7 items
- ALDEFLUOR 216 items
- AggreWell 55 items
- ArciTect 1 item
- BrainPhys 45 items
- CellPore 1 item
- ClonaCell 84 items
- CryoStor 65 items
- ES-Cult 77 items
- EasyPick 1 item
- EasySep 752 items
- EpiCult 12 items
- HepatiCult 1 item
- ImmunoCult 7 items
- IntestiCult 142 items
- Lymphoprep 9 items
- MammoCult 45 items
- MegaCult 34 items
- MesenCult 133 items
- MethoCult 444 items
- MyeloCult 64 items
- MyoCult 2 items
- NeuroCult 353 items
- NeuroFluor 1 item
- PancreaCult 3 items
- PneumaCult 78 items
- RSeT 7 items
- ReLeSR 1 item
- RoboSep 23 items
- RosetteSep 253 items
- STEMdiff 54 items
- STEMvision 3 items
- SepMate 29 items
- StemSpan 219 items
- TeSR 1456 items
- ThawSTAR 1 item
- mFreSR 3 items
- Cell and Tissue Source
-
- 24 items
- Cell Line
-
- 24 items
- Cell Type
-
- 27 items
- Airway Cells 41 items
- B Cells 134 items
- Brain Tumor Stem Cells 81 items
- Cancer Cells and Cell Lines 116 items
- Cardiomyocytes, PSC-Derived 8 items
- Dendritic Cells 59 items
- Dermal Cells 1 item
- Endoderm, PSC-Derived 1 item
- Endothelial Cells 1 item
- Endothelial Cells, PSC-Derived 1 item
- Epithelial Cells 49 items
- Granulocytes and Subsets 61 items
- Hematopoietic Stem and Progenitor Cells 777 items
- Hepatic Cells 2 items
- Hybridomas 75 items
- Innate Lymphoid Cells 3 items
- Intestinal Cells 13 items
- Kidney Cells 1 item
- Leukemia/Lymphoma Cells 8 items
- Leukopaks 1 item
- Mammary Cells 68 items
- Mesenchymal Stem and Progenitor Cells 132 items
- Monocytes 105 items
- Mononuclear Cells 33 items
- Myeloid Cells 99 items
- NK Cells 80 items
- Neural Cells, PSC-Derived 17 items
- Neural Stem and Progenitor Cells 381 items
- Neurons 135 items
- Plasma 3 items
- Pluripotent Stem Cells 1689 items
- Prostate Cells 7 items
- Renal Cells 2 items
- T Cells 179 items
- T Cells, CD4+ 85 items
- T Cells, CD8+ 49 items
- T Cells, Regulatory 18 items
- Species
-
- 41 items
Loading...Copyright © 2025 º£½ÇÆÆ½â°æ. All rights reserved.