Pages
Products
Human SLC2A2 Stable Cell Line - CHO-K1

Human SLC2A2 Stable Cell Line - CHO-K1

Cat.No. :  CSC-SC014484-1 Host Cell:  CHO-K1

Inquire for Price

Cell Line Information

Cell Culture Information

Safety and Packaging

Cat. No. CSC-SC014484-1
Description This cell line is engineered to stably express human solute carrier family 2 member 2 (SLC2A2) in CHO-K1 cells.
Gene SLC2A2
Gene Species Homo sapiens (Human)
Host Cell CHO-K1
Host Cell Species Cricetulus griseus (Chinese hamster)
Stability Validated for at least 10 passages
Application

1. Gene expression studies

2. Signaling pathway research

3. Drug screening and toxicology

4. Disease research

Quality Control Negative for bacteria, yeast, fungi and mycoplasma.
Shipping Dry ice
Storage Liquid nitrogen
Revival Rapidly thaw cells in a 37°C water bath. Transfer contents into a tube containing pre-warmed media. Centrifuge cells and seed into a 25 cm2 flask containing pre-warmed media.
Mycoplasma Negative
Format One frozen vial containing millions of cells
Storage Liquid nitrogen
Safety Considerations

The following safety precautions should be observed.

1. Use pipette aids to prevent ingestion and keep aerosols down to a minimum.

2. No eating, drinking or smoking while handling the stable line.

3. Wash hands after handling the stable line and before leaving the lab.

4. Decontaminate work surface with disinfectant or 70% ethanol before and after working with stable cells.

5. All waste should be considered hazardous.

6. Dispose of all liquid waste after each experiment and treat with bleach.

Ship Dry ice
Quick Inquiry

Background

Case Study

Applications

Publications

Q & A

Customer Reviews

SLC2A2, also known as the facilitated glucose transporter member 2 (GLUT2), is a pivotal transmembrane protein involved in cellular glucose transport. Initially discovered in 1991 by Thorens et al., SLC2A2 plays a crucial role in glucose homeostasis, particularly in pancreatic β-cells and hepatocytes. The identification of SLC2A2's significance in glucose sensing and insulin secretion underscores its importance in metabolic regulation. To facilitate further research, stable cell lines expressing human SLC2A2, such as the CHO-K1 cell line, have been developed. The generation of these stable cell lines involves transfecting host cells with a vector containing the human SLC2A2 gene, followed by selection and propagation of cells expressing high levels of SLC2A2. These cell lines serve as invaluable tools for investigating the molecular mechanisms underlying glucose transport and metabolism, as well as for screening potential therapeutic agents targeting glucose-related disorders. "FLT3 (Fms-like tyrosine kinase 3) plays a critical role as a receptor tyrosine kinase in the differentiation and proliferation of hematopoietic stem and progenitor cells, essential processes in maintaining a healthy blood cell population. The groundbreaking discovery of internal tandem duplication (ITD) mutations within FLT3 in 1996 revolutionized our understanding of leukemia pathogenesis. These ITD mutations, predominantly located in the juxtamembrane domain of FLT3, induce constitutive activation of the receptor, rendering it insensitive to normal regulatory mechanisms. Consequently, downstream signaling pathways are aberrantly activated, driving uncontrolled cell proliferation and inhibiting apoptosis, ultimately contributing to the development of acute myeloid leukemia (AML). Furthermore, the identification of the D835V mutation within the tyrosine kinase domain of FLT3 unveiled another layer of complexity in FLT3-driven leukemogenesis. This specific mutation, observed in a subset of AML patients, confers resistance to conventional FLT3 inhibitors by altering the ATP-binding pocket of the kinase domain, thereby impeding the binding of these inhibitors and compromising treatment efficacy. The emergence of resistance underscores the urgent need for alternative therapeutic strategies targeting FLT3-ITD-D835V mutations. The multifaceted nature of FLT3 mutations underscores the importance of comprehensive research efforts aimed at unraveling the intricate molecular mechanisms underlying leukemogenesis. Through elucidating these mechanisms, novel therapeutic targets can be identified and innovative treatment modalities developed, ultimately improving clinical outcomes for AML patients." "The technique of Transcriptional Promoter Reporter (TPR) and its subsequent modifications, such as the Multicolor Expression Technology (MET), have revolutionized the field of cellular biology research. Originally conceived in the latter part of the 20th century, TPR marked a pivotal advancement in our ability to study gene expression dynamics within living cells. By integrating specific promoter regions with reporter genes, TPR facilitated the direct visualization and precise quantification of transcriptional activity, offering invaluable insights into the intricacies of gene regulation. Building upon the foundation laid by TPR, the introduction of Multicolor Expression Technology (MET) represented a significant leap forward. MET's innovation lies in its capability to simultaneously monitor multiple gene promoters within a single cell, providing researchers with a comprehensive view of transcriptional events occurring within complex cellular environments. This breakthrough has greatly expanded our understanding of gene regulatory networks and their functional implications. The development of the Human TPR-MET Stable Cell Line - BaF3 exemplifies the continual refinement of these methodologies. By integrating human-specific transcriptional regulatory elements with the MET technology, this stable cell line offers researchers a reliable and physiologically relevant model system for investigating gene expression dynamics in human cells. The availability of such a tool is instrumental in unraveling the complexities of human biology and has profound implications for various fields, including drug discovery, disease modeling, and regenerative medicine." "The discovery of EGFR mutations, particularly EGFR-Del19, T790M, and C797S, has revolutionized the management of non-small cell lung cancer (NSCLC), offering critical insights into tumor biology and therapeutic strategies. EGFR-Del19, first identified in the early 2000s, involves the deletion of amino acids 747 to 750 in exon 19 of the EGFR gene. This alteration leads to increased kinase activity and heightened sensitivity to EGFR tyrosine kinase inhibitors (TKIs) like gefitinib and erlotinib. However, the emergence of resistance remains a significant challenge in EGFR-mutant NSCLC. The T790M mutation, occurring in approximately half of patients who develop resistance to first-generation EGFR TKIs, involves a substitution of threonine with methionine at position 790 within the EGFR kinase domain. This alteration impairs TKI binding while maintaining EGFR activation, rendering tumors resistant to therapy. Subsequently, third-generation EGFR TKIs, such as osimertinib, were developed to target T790M-positive tumors, yet the emergence of additional mutations, notably C797S, poses further obstacles. C797S mutation arises within the kinase domain of EGFR and disrupts the covalent binding of third-generation TKIs, leading to treatment resistance. Its identification underscores the dynamic nature of tumor evolution and the necessity for ongoing therapeutic innovation. In response, researchers have utilized sophisticated genetic engineering techniques to establish cell line models harboring EGFR-Del19, T790M, and C797S mutations, such as the Human EGFR-Del19/T790M/C797S Stable Cell Line - PC-9. These models serve as invaluable tools for elucidating mechanisms of drug resistance, evaluating novel treatment modalities, and guiding personalized therapeutic approaches in EGFR-mutant NSCLC patients." "The Receptor Tyrosine Kinase-like Orphan Receptor 2 (ROR2) is a member of the ROR family of receptor tyrosine kinases. It was discovered in the late 1990s as a transmembrane protein involved in embryonic skeletal development and tissue morphogenesis. ROR2 plays a crucial role in the regulation of cell polarity, migration, and differentiation pathways through its interaction with Wnt signaling cascades. HEK293T cells, derived from human embryonic kidney cells, have been widely utilized as a host for gene expression studies and protein production due to their high transfection efficiency and robust growth characteristics. These cells have been instrumental in elucidating the molecular mechanisms underlying ROR2-mediated signaling pathways and functional activities. The establishment of Human ROR2 Stable Cell Line - HEK293T has facilitated detailed investigations into the role of ROR2 in various cellular processes, including its involvement in cancer progression, immune response modulation, and developmental disorders. This cell line serves as a valuable tool for exploring the therapeutic potential of targeting ROR2 in various disease contexts." "The KIT proto-oncogene, also known as c-Kit or CD117, encodes a receptor tyrosine kinase (RTK) critical for various cellular processes, including proliferation, survival, and differentiation of hematopoietic progenitor cells and melanocytes. Aberrant activation of this gene is closely associated with the occurrence and progression of multiple tumors. The Ba/F3 cell line, initially derived from murine lymphoid cells, has been extensively utilized in the functional characterization of oncogenes and signaling pathways due to its dependency on interleukin-3 (IL-3) for growth. Its characteristics render it an ideal cellular model for investigating novel oncogenes. The establishment of the human KIT stable cell line-Ba/F3 represents a significant advancement. By stably transfecting human KIT cDNA into Ba/F3 cells, the study of KIT-mediated signaling and its implications in cellular physiology and pathology became feasible. This cell line's establishment has provided valuable insights into the molecular mechanisms underlying KIT-associated diseases, such as gastrointestinal stromal tumors (GISTs) and acute myeloid leukemia (AML). Through these studies, we can better comprehend the role of KIT in normal cellular functions and disease development, thereby laying a theoretical foundation for the diagnosis and treatment of related diseases." "STAT5 (Signal Transducer and Activator of Transcription 5) is a key member of the STAT transcription factor family, influencing vital cellular processes including proliferation, differentiation, and apoptosis. Identified in 1994, STAT5 plays a central role in cellular signaling cascades, particularly in response to cytokines like interleukin-2 (IL-2) and erythropoietin (EPO), marking a significant shift in understanding cellular responses to external stimuli. The introduction of Luciferase reporter gene assays in the late 1970s transformed molecular biology, enabling real-time monitoring of transcriptional activity and offering insights into gene expression regulation. Coupling these assays with STAT5 studies provided unprecedented insights into the transcriptional regulation of STAT5-responsive genes, unraveling complexities in cellular signaling networks. Ba/F3, a murine pro-B cell line responsive to interleukin-3 (IL-3), has become pivotal in hematopoietic research due to its genetic manipulability and robust proliferation, serving as an ideal model for studying hematopoiesis signaling pathways. Introduction of stable luciferase reporter constructs controlled by STAT5-responsive promoters into Ba/F3 cells facilitated the creation of the STAT5-Luc Reporter Cell Line-Ba/F3, aiding in dissecting STAT5 signaling dynamics under various conditions." "The development of GFP reporter stable cell lines, particularly in the context of HEK293 cells, traces back to the early 1990s with the discovery of the green fluorescent protein (GFP) from the jellyfish Aequorea victoria by Shimomura et al. in 1962. The utilization of GFP as a molecular marker gained prominence following the elucidation of its structure and the subsequent demonstration of its utility as a versatile reporter molecule for cellular processes. Subsequent advancements in molecular biology techniques facilitated the engineering of GFP variants with improved fluorescence properties and expanded applications. In the case of HEK293 cells, the establishment of stable cell lines expressing GFP reporters involved the transfection of plasmids encoding GFP constructs into these cells, followed by selection and clonal expansion of cells expressing the reporter gene. Over the years, optimization of transfection protocols, selection markers, and expression vectors has enhanced the efficiency and stability of GFP reporter cell line generation in HEK293 cells. These stable cell lines have become invaluable tools in various research fields, including cell biology, drug discovery, and disease modeling, enabling the visualization and quantification of cellular processes in real-time." The development of the RFP Reporter Stable Cell Line-HEK293 traces its origins to the early 1970s with the discovery and establishment of the Human Embryonic Kidney 293 (HEK293) cell line by Graham et al. in 1977. Initially derived from human embryonic kidney cells, HEK293 has since served as a cornerstone in biomedical research due to its robust growth characteristics and versatility in molecular biology applications. The subsequent emergence of Red Fluorescent Protein (RFP) as a vital tool for cellular imaging and protein localization further catalyzed the fusion of RFP with HEK293 cells, resulting in the creation of the RFP Reporter Stable Cell Line. This innovative lineage, born at the intersection of cell biology and fluorescence microscopy, has revolutionized the visualization and tracking of cellular processes, offering researchers unparalleled insights into dynamic intracellular events. The synergy between HEK293's reliability and RFP's fluorescence properties underscores the significance of this cell line in elucidating complex biological phenomena, spanning from gene expression analysis to drug discovery endeavors. The development of GFP Stable Cell Line-B16-F10(Mouse) stems from the groundbreaking discovery of green fluorescent protein (GFP) in the 1960s. GFP, initially identified in the jellyfish Aequorea victoria, found application as a vital tool in cellular and molecular biology due to its inherent fluorescence. The fusion of GFP to proteins of interest enables visualization and tracking within living cells, revolutionizing biological research. Over time, advancements in molecular biology techniques facilitated the creation of stable cell lines expressing GFP, enhancing experimental capabilities. The GFP Stable Cell Line-B16-F10(Mouse) specifically refers to a murine B16-F10 melanoma cell line engineered to express GFP continuously. This development amalgamates the intrinsic properties of GFP with the versatile B16-F10 model system, offering a platform for studying cellular processes, tumor biology, and therapeutic interventions in vivo. The continuous refinement and utilization of GFP Stable Cell Line-B16-F10(Mouse) underscore its significance in elucidating intricate biological mechanisms. The GFP Stable Cell Line-HCT-116 originates from the development of stable cell lines expressing Green Fluorescent Protein (GFP) within the HCT-116 cell line background. This cell line has been instrumental in elucidating various cellular processes and pathways due to its ability to visualize protein localization, dynamics, and interactions in live cells. The establishment of GFP-expressing stable cell lines began with the discovery of GFP as a naturally occurring fluorescent protein in the jellyfish Aequorea victoria in the 1960s. Subsequent advancements in molecular biology and recombinant DNA technology facilitated the cloning and expression of GFP in mammalian cells. This breakthrough allowed for the creation of GFP Stable Cell Lines, including those derived from the HCT-116 cell line, which is widely used in cancer research. By tracking GFP-tagged proteins, researchers gain insights into cellular behavior, gene expression, signal transduction, and drug responses, enhancing our understanding of various physiological and pathological processes. The Luc/GFP reporter cell line HT-29 has emerged as a vital tool in molecular biology research, facilitating the study of gene expression dynamics and cellular processes. Its development traces back to the early 2000s when researchers sought to enhance the precision and efficiency of gene expression analysis. By fusing luciferase (Luc) and green fluorescent protein (GFP) genes to regulatory elements of interest, such as promoters or enhancers, scientists could monitor gene activity in real-time with both luminescent and fluorescent readouts. This dual-reporter system offers distinct advantages, including high sensitivity, non-invasiveness, and compatibility with live-cell imaging techniques. Over the years, refinements in vector design, cell culture techniques, and imaging technologies have further optimized the utility and versatility of Luc/GFP reporter cell lines like HT-29. Today, these cell lines continue to underpin investigations spanning diverse fields such as oncology, developmental biology, and drug discovery. The Luc/GFP reporter cell line-B16F10 has emerged as a pivotal tool in biomedical research, facilitating the elucidation of various cellular processes. Its inception traces back to the seminal discovery of luciferase as a bioluminescent enzyme in the early 20th century. Subsequent advancements in molecular biology, particularly the advent of recombinant DNA technology in the 1970s, catalyzed the development of reporter gene systems. Notably, the discovery and engineering of green fluorescent protein (GFP) in the 1960s and its subsequent refinement in the 1990s by Roger Y. Tsien revolutionized cellular imaging techniques. The fusion of luciferase and GFP genes into a single expression cassette paved the way for dual-reporter systems, offering complementary readouts for cellular activities. The derivation of the Luc/GFP reporter cell line-B16F10, specifically from murine melanoma cells, epitomizes the synergy between fundamental discoveries and applied biomedical research. Its utilization in various experimental paradigms underscores its significance in elucidating molecular mechanisms underlying cancer progression, drug efficacy, and therapeutic interventions. The Luc Reporter Cell Line-HCT116, developed over time through a meticulous process, represents a significant advancement in cellular research. Originally derived from the human colorectal carcinoma cell line HCT116, this cell line was engineered to incorporate luciferase reporter gene constructs. These constructs enable the monitoring of specific cellular processes and signaling pathways through the emission of bioluminescence upon activation. The development of Luc Reporter Cell Line-HCT116 stems from the imperative to elucidate intricate molecular mechanisms underlying various biological phenomena. This innovative tool provides researchers with a robust platform for real-time, non-invasive assessment of gene expression, protein-protein interactions, and cellular responses to stimuli. The integration of luciferase reporter technology into the HCT116 cell line, pioneered in the early 21st century, has since revolutionized biomedical research, offering unparalleled insights into fundamental cellular processes. The RFP Reporter Cell Line-MC3T3, derived from murine calvaria, has been genetically engineered to express a red fluorescent protein (RFP), facilitating real-time visualization and tracking of cellular processes. The developmental trajectory of this cell line involves meticulous optimization of transfection techniques and selection methodologies to ensure stable and reliable expression of the RFP reporter gene. Furthermore, its integration into various experimental paradigms has demonstrated its utility in elucidating dynamic cellular behaviors, such as proliferation, differentiation, and morphogenesis. The establishment of the RFP Reporter Cell Line-MC3T3 underscores its pivotal role in advancing our understanding of fundamental biological processes and holds immense promise for applications in drug discovery, tissue engineering, and regenerative medicine. The NALM6 cell line, established in 1979 from the bone marrow of a pediatric patient with acute lymphoblastic leukemia (ALL), serves as a crucial tool in leukemia research. Its origin in B-cell precursor ALL renders it valuable for studying B-lineage leukemogenesis and therapeutic interventions. Over decades, NALM6 has been extensively utilized to investigate molecular mechanisms underlying leukemic progression, drug sensitivity, and resistance. Additionally, the integration of luciferase and GFP reporter genes into NALM6 cells facilitates real-time monitoring of cellular events such as proliferation, apoptosis, and response to therapeutic agents. This modification enhances the utility of NALM6 in high-throughput drug screening assays and in vivo imaging studies. Thus, the NALM6-Luc/GFP reporter cell line amalgamates the robustness of NALM6 with the versatility of luciferase/GFP technology, offering a powerful platform for elucidating leukemia pathogenesis and evaluating novel therapeutic strategies. "The K562 cell line, established in 1970 from a patient with chronic myelogenous leukemia (CML), has since served as a fundamental tool in hematological research. Its origin, characterized by the Philadelphia chromosome, facilitated elucidation of the BCR-ABL fusion gene's role in CML pathogenesis. K562's versatility extends to drug screening and gene expression studies due to its stable karyotype and ease of cultivation. The luciferase/green fluorescent protein (Luc/GFP) reporter system, pioneered in the late 20th century, enables real-time monitoring of gene expression. This technology relies on luciferase enzymes derived from fireflies or other organisms, coupled with GFP, allowing researchers to visualize and quantify transcriptional activity accurately. Combining these resources, the K562 Luc/GFP reporter cell line was developed to investigate regulatory elements and signaling pathways governing hematopoietic gene expression dynamics. This cell line amalgamates the strengths of both K562's physiological relevance and the Luc/GFP reporter system's sensitivity and versatility. Its establishment marks a pivotal advancement in hematological research, enhancing our understanding of gene regulatory mechanisms within the context of leukemia and beyond." "The Raji cell line, originating from Burkitt's lymphoma, was established in 1961 by Epstein and colleagues. Raji cells are characterized by their B lymphocyte origin and carry the Epstein-Barr virus genome. Over the years, Raji cells have served as a fundamental model in immunology and oncology research due to their ability to express various cell surface markers and cytokines relevant to B cell biology. Luciferase Stable Cell Lines refer to cell lines engineered to express luciferase, a light-emitting enzyme derived from fireflies. Luciferase assays are widely utilized in molecular biology for monitoring gene expression, cell viability, and protein-protein interactions. The fusion of luciferase with target genes allows for real-time, non-invasive monitoring of cellular processes in vitro and in vivo. The creation of Raji-Luciferase Stable Cell Lines involves transfection of Raji cells with luciferase expression vectors followed by selection and maintenance of stable clones expressing luciferase. This technology enables researchers to investigate signaling pathways, drug efficacy, and disease progression in the context of B cell biology." "Colon 26 is a murine colon carcinoma cell line established in the mid-1970s. Its development marked a significant milestone in cancer research, providing a robust model system for studying colorectal cancer progression, metastasis, and therapeutic interventions. Colon 26 cells exhibit high tumorigenicity in syngeneic mouse models, making them valuable tools for preclinical studies. The GFP/Luc Reporter Cell Line derived from Colon 26 integrates green fluorescent protein and luciferase reporter genes, enhancing its utility for in vivo imaging and monitoring tumor growth and metastasis. This modification enables non-invasive longitudinal tracking of tumor dynamics and response to treatment in live animals, facilitating the evaluation of experimental therapeutics and elucidation of underlying biological mechanisms. The combination of Colon 26 and reporter gene technologies has revolutionized cancer research by providing a platform for real-time visualization and quantification of tumor behavior in preclinical models. This model system has greatly contributed to our understanding of colorectal cancer biology and has facilitated the development of novel therapeutic strategies." "The Jeko-1 cell line originated from the peripheral blood of a patient diagnosed with mantle cell lymphoma (MCL), making it a crucial resource for MCL research. Through meticulous characterization, Jeko-1 exhibited characteristic features of MCL, including the t(11;14)(q13;q32) translocation and overexpression of cyclin D1. This faithful representation of MCL pathophysiology positions Jeko-1 as an indispensable tool for unraveling the disease's etiology and exploring therapeutic strategies. The creation of the GFP/Luc reporter cell line involved transfecting Jeko-1 cells with plasmids encoding green fluorescent protein (GFP) and luciferase. This innovative modification enables dynamic visualization and quantification of cellular processes and drug responses both in vitro and in vivo. Such advancements not only offer unprecedented insights into the underlying biology of MCL but also facilitate the assessment of novel therapeutic interventions and their efficacy. The establishment of the Jeko-1 cell line marked a seminal milestone in MCL research, catalyzing deeper investigations into its molecular intricacies and therapeutic modalities. Subsequent advancements, exemplified by the creation of the GFP/Luc reporter cell line, have further expanded the utility of Jeko-1, providing a sophisticated platform for conducting intricate experiments and high-throughput drug screenings tailored specifically to the context of MCL." "The HL60 cell line, initially derived in 1977 from a patient afflicted with acute promyelocytic leukemia, has emerged as a cornerstone in hematological research. Its establishment marked a pivotal moment in understanding leukemia biology, facilitating investigations into cellular differentiation, apoptosis, and leukemic pathophysiology. Over decades of study, HL60 cells have provided invaluable insights into the molecular mechanisms underlying leukemia development and progression. Moreover, the advent of GFP reporter technology has revolutionized cellular imaging and gene expression analysis. The creation of GFP Reporter Cell Lines, including the HL60 GFP Reporter Cell Line, has empowered researchers to probe gene expression dynamics with unprecedented precision. By harnessing the inherent fluorescence of GFP, these engineered cells enable real-time visualization and quantification of gene activity within living cells. This technological breakthrough has catalyzed groundbreaking discoveries, allowing for the elucidation of intricate signaling cascades and regulatory networks governing hematopoietic differentiation and leukemogenesis. In essence, the convergence of HL60 cells and GFP reporter technology has ushered in a new era of cellular and molecular hematological research, driving advancements in our understanding of leukemia and offering promising avenues for therapeutic intervention." "The MCF10DCIS.com cell line is a widely utilized model system in breast cancer research, derived from the MCF10A immortalized mammary epithelial cell line. Established in 2001 by Miller et al., this cell line recapitulates key features of ductal carcinoma in situ (DCIS), a non-invasive form of breast cancer. Notably, MCF10DCIS.com cells exhibit characteristics akin to early-stage breast cancer, such as the formation of duct-like structures and the ability to progress to invasive phenotypes upon appropriate stimuli. The GFP Reporter Cell Line is engineered to express green fluorescent protein (GFP) under the control of specific regulatory elements, facilitating real-time visualization of gene expression dynamics. The coupling of GFP reporter technology with MCF10DCIS.com cells enables precise monitoring of molecular events implicated in breast cancer progression. This synergy offers researchers a powerful tool for elucidating the molecular mechanisms underlying DCIS pathogenesis and the transition to invasive breast cancer. Established at a later date, the GFP Reporter Cell Line augments the utility of MCF10DCIS.com as a robust model system for investigating breast cancer biology." "Since its discovery in 1951 by Dr. Margaret Lewis, the LLC (Lewis Lung Carcinoma) cell line has played a pivotal role in oncological research. Initially derived from a lung carcinoma in a C57BL mouse strain, LLC cells have emerged as a cornerstone model for investigating various aspects of cancer biology, metastasis, and therapeutic strategies. Through meticulous characterization efforts spanning several decades, researchers have meticulously unraveled the molecular and phenotypic intricacies of LLC cells, substantially advancing our comprehension of tumor progression and immunotherapeutic responses. Concurrently, the advent of GFP (Green Fluorescent Protein) reporter cell lines has heralded a revolution in cellular imaging and molecular biology. Integration of GFP into LLC cells has revolutionized the visualization and tracking of intricate cellular processes, encompassing gene expression, protein localization, and cell migration dynamics. This groundbreaking technique, spearheaded by Shimomura, Chalfie, and Tsien in the 1990s, has exponentially augmented the applicability of LLC cells in dissecting complex biological mechanisms. The amalgamation of LLC cells' robustness with GFP reporter technology has empowered researchers to probe deeper into the dynamic interplay between tumor cells and their microenvironment." "The A20 cell line, established in 1971 by Hamburger et al., originates from a spontaneous reticulum cell sarcoma in a BALB/cAnN mouse. Initially utilized as a tool to study lymphoma pathogenesis, A20 cells exhibit characteristics typical of B lymphocytes, including surface immunoglobulins and the ability to undergo class switching. Over time, researchers have extensively employed A20 cells to elucidate various signaling pathways and immune responses, owing to their amenability to genetic manipulation and stable propagation. Subsequent advancements in molecular biology techniques facilitated the development of reporter cell lines incorporating fluorescent or luminescent markers to monitor gene expression and cellular activities. The fusion of A20 cells with reporter genes such as mCherry and Luciferase has revolutionized the study of molecular mechanisms underlying immune responses, tumorigenesis, and therapeutic interventions. The mCherry reporter, a red fluorescent protein derived from Discosoma sp., enables visual tracking of A20 cells in vivo and in vitro with exceptional brightness and photostability. In parallel, the incorporation of Luciferase, a light-emitting enzyme from fireflies, into A20 cells provides a sensitive and quantitative means to assess transcriptional activity and cellular viability." "OVCAR8, a cell line derived from human ovarian adenocarcinoma, has been instrumental in advancing cancer research since its discovery in the early 1980s. Developed by J. C. Soucie and colleagues at the National Cancer Institute, OVCAR8 cells exhibit characteristics typical of high-grade serous ovarian carcinoma, making them valuable tools for studying ovarian cancer biology, progression, and therapeutic responses. OVCAR8 cells have been extensively characterized for their genetic and phenotypic features, including their ability to form tumors in xenograft models. Moreover, the Luciferase Reporter Cell Line based on OVCAR8 provides an enhanced platform for studying gene expression regulation and signaling pathways implicated in ovarian cancer. This reporter system incorporates a luciferase reporter gene under the control of specific promoters or response elements, enabling quantitative assessment of transcriptional activity in OVCAR8 cells. The integration of luciferase technology with the OVCAR8 cell line enhances the sensitivity and specificity of detecting molecular events associated with ovarian cancer progression and treatment responses." "U2OS, a human osteosarcoma cell line, has garnered significant attention in biomedical research owing to its robustness, ease of cultivation, and genomic stability. Established in 1964 by J. Pontén and colleagues, U2OS cells were derived from a 15-year-old female patient suffering from osteosarcoma. The utility of U2OS cells in elucidating various cellular processes stems from their epithelial morphology, diploid karyotype, and functional characteristics resembling primary osteoblasts. Over the years, researchers have extensively employed U2OS cells to investigate diverse biological phenomena including cell cycle regulation, DNA repair mechanisms, and signal transduction pathways. The advent of luciferase reporter assays revolutionized the study of gene expression dynamics and cellular signaling events. The luciferase enzyme, isolated from the firefly Photinus pyralis, catalyzes the oxidation of luciferin, resulting in the emission of bioluminescent light. This phenomenon, exploited for monitoring gene expression, enables real-time, non-invasive assessment of transcriptional activity in living cells. Combining the versatility of U2OS cells with luciferase reporter technology has facilitated the development of U2OS luciferase reporter cell lines, empowering researchers to dissect intricate molecular mechanisms underlying various physiological and pathological processes. These engineered cell lines typically incorporate luciferase reporter constructs under the control of specific promoters or response elements, allowing for precise monitoring of gene expression changes in response to environmental stimuli or pharmacological interventions." "The REH cell line, originating from a patient with acute lymphoblastic leukemia, has been extensively utilized in biomedical research due to its relevance in the study of hematopoietic malignancies. Established in the early 1970s, REH cells have served as a fundamental tool in elucidating various aspects of leukemia biology, including molecular mechanisms underlying disease progression and therapeutic responses. Over the years, researchers have harnessed the potential of REH cells to investigate genetic alterations, signaling pathways, and drug sensitivities pertinent to leukemia pathogenesis. The development of REH cells stably expressing Green Fluorescent Protein (GFP) reporter has significantly enhanced their utility in cellular imaging studies and high-throughput screening assays. The GFP reporter system enables real-time visualization and quantification of cellular processes, facilitating the assessment of dynamic cellular behaviors, such as proliferation, migration, and differentiation. This innovative approach has greatly expedited the identification of novel therapeutic targets and evaluation of drug efficacy in leukemia research. The REH cell line, established decades ago, continues to be a cornerstone in leukemia research, providing valuable insights into disease mechanisms and serving as a platform for drug discovery endeavors. The integration of GFP reporter technology further augments the versatility of REH cells, enabling precise and comprehensive investigations into leukemia biology." "The KU-812E cell line is a notable hematopoietic cell line derived from a patient with acute myelogenous leukemia (AML). It was originally established in 1976 by Toru Itoh and colleagues at the Kawasaki Medical School in Japan. The cell line was derived from the KU-812 cell line, which was established from the peripheral blood of a patient with chronic myelogenous leukemia (CML). The transition to the KU-812E subline was achieved by continuous culture in the presence of phorbol ester, leading to the acquisition of characteristics resembling those of immature cells in AML. In the context of molecular biology research, the KU-812E cell line has been extensively utilized due to its relevance in studying hematopoietic differentiation, leukemogenesis, and the molecular mechanisms underlying leukemia pathogenesis. Furthermore, its sensitivity to various pharmacological agents has rendered it a valuable tool for drug screening and therapeutic development in the field of leukemia research. The GFP (Green Fluorescent Protein) reporter cell line, established using the KU-812E cells, incorporates a GFP expression cassette into the cellular genome. This reporter system allows for the visualization and quantification of gene expression or cellular processes through the detection of GFP fluorescence. The introduction of the GFP reporter into the KU-812E cell line provides a versatile platform for studying gene regulation, signal transduction pathways, and drug responses in the context of leukemia." "The MV4-11 cell line is a widely utilized model in leukemia research, particularly in studies focusing on acute myeloid leukemia (AML). Originally established in the early 1970s, MV4-11 cells were derived from the bone marrow of a 32-year-old male patient diagnosed with AML. These cells have been extensively characterized and are recognized for their high proliferative capacity and relevance to AML biology. Over the decades, MV4-11 cells have served as an invaluable tool in elucidating the molecular mechanisms underlying AML pathogenesis and drug response. Researchers have employed MV4-11 cells to investigate various aspects of leukemia biology, including cellular signaling pathways, genetic mutations, and drug resistance mechanisms. Furthermore, the availability of the MV4-11 cell line has facilitated the development and evaluation of novel therapeutic strategies for AML treatment. In conjunction with MV4-11 cells, Luciferase Reporter Cell Lines have emerged as indispensable tools for studying gene regulation and expression dynamics. By incorporating luciferase reporter constructs into the genome of MV4-11 cells, researchers can monitor the activity of specific promoters or regulatory elements in real-time. This enables the quantitative assessment of transcriptional activity and the evaluation of drug effects on gene expression profiles." "The SU-DHL-4 cell line, established in 1982, is a well-characterized model of diffuse large B-cell lymphoma (DLBCL). Initially derived from the lymph node biopsy of a patient with DLBCL, SU-DHL-4 cells have been extensively utilized in research aimed at understanding the pathogenesis and therapeutic responses of DLBCL. These cells exhibit characteristic features of DLBCL, including the expression of B-cell markers and the presence of chromosomal abnormalities commonly observed in this malignancy. The development of the SU-DHL-4 GFP reporter cell line represents a significant advancement in DLBCL research. This reporter cell line incorporates a green fluorescent protein (GFP) reporter gene into the SU-DHL-4 cellular genome, allowing for the visualization and tracking of specific cellular processes or molecular events in real-time. By exploiting the unique fluorescence properties of GFP, researchers can monitor cellular behaviors, such as proliferation, migration, and apoptosis, in response to various experimental conditions or therapeutic interventions. Additionally, the SU-DHL-4 GFP reporter cell line serves as a valuable tool for high-throughput screening assays and drug discovery efforts targeting DLBCL." "The FOLH1 gene, also known as folylpolyglutamate synthetase, is a protein-coding gene in monkeys. The protein encoded by this gene has two main functions. It plays an important role in folate metabolism and is involved in the synthesis of foylpolyglutamic acid, a compound necessary for the synthesis of purines and pyrimidines. FOLH1 protein is expressed in multiple tissues, including kidney, liver, intestine, and placenta, demonstrating its widespread importance in metabolic processes. Mutations in the FOLH1 gene are associated with a variety of cancers. The protein encoded by this gene is also a highly specific biomarker for prostate cancer and is used in prognosis and imaging of the disease. Furthermore, it has recently been used as a target for immunotherapeutic strategies for prostate cancer treatment. The monkey FOLH1 stable cell line is a specially designed cell line expressing the monkey-derived FOLH1 gene. This cell line is critical for a variety of biomedical and genetic research efforts, primarily due to its stable expression of the FOLH1 gene. The Monkey FOLH1 stable cell line is a reliable and consistent tool for conducting a wide range of laboratory studies related to these conditions. It enables researchers to study the mechanisms of these diseases at the cellular level, making a significant contribution to the development of potential treatments and treatment strategies." "Avaren-Fc (AvFc) is a unique and innovative fusion protein composed of a variant of the actinomycete lectin actinomycin (Avaren) and the Fc (crystallizable fragment) region of human immunoglobulin G1 (IgG1). Avaren is a natural sugar-binding protein produced by actinomycetes. This lectin is noteworthy because of its high specificity for binding to high-mannose-type sugar chains, which are often found on the surfaces of various types of viruses and glycosylated proteins. Due to this property, Avaren is considered for its great potential as an effective antiviral agent in the field of virology against particularly dangerous and widespread viruses (such as HIV). However, in order for Avaren to be used effectively in vivo, modifications were required, resulting in Avaren-Fc. The Fc region of an antibody (such as human IgG1) is responsible for triggering the immune response. By merging this part of the antibody with Avaren, two significant benefits are achieved. First, the fusion molecule inherits the long metabolic half-life of the Fc fragment, thereby enhancing the duration of Avaren's therapeutic effects in the body. Second, because the human Fc region is readily recognized by the body, it enhances the potential of Avaren-Fc to trigger a more robust immune response against affected cells displaying high mannose chains." "Spectrin beta, non-erythrocytic 1 (SPTBN1) is a gene encoding a member of the spectrin family. Spectrin is widely distributed in cells and forms a key component of the cytoskeletal system, giving cells their shape and ability to function efficiently. The SPTBN1 protein plays a critical role in maintaining cell shape, cytokinesis, and cell stability. It is also involved in intracellular membrane trafficking and binding to various cellular proteins. Mutations in this gene can lead to a variety of health conditions, including neurodegenerative diseases. Furthermore, SPTBN1 is considered to be a key link between the cell membrane and the underlying cytoskeleton, essential for maintaining the integrity of the plasma membrane and cell morphology. Furthermore, SPTBN1 has been reported to be associated with tumor progression and metastasis in various cancers. Human SPTBN1 Stable Cell Line - HEK293 is a cell line engineered to stably express the human SPTBN1 gene. The SPTBN1 gene was encoded into the HEK293 cell line, a specific line of human embryonic kidney cells. These kidney cells are grown in vitro and are commonly used in cell biology research due to their ease of culture and vigorous growth. The stability of the SPTBN1 gene in this cell line ensures continued expression of the gene, making these cells an extremely reliable tool for studying gene function using advanced molecular techniques." "Mouse Fap, also known as fibroblast-activating protein alpha, is the protein encoded by the Fap gene in mice. This protein is part of the serine protease family and plays a role in a variety of physiological and pathological processes. Fap is expressed at low levels in normal tissues but is often highly expressed in cancer-associated fibroblasts (CAFs) of epithelial cancers, including more than 90% of human breast, colorectal, and pancreatic cancers. It plays an important role in tissue remodeling, wound healing, and tumor growth. Because Fap is associated with cancer disease progression, it has been the target of various pharmacological development programs aimed at reducing its activity in vivo. It is also used as a marker in diagnostic tests to identify the presence of certain cancers or other related conditions. Mouse Fap Stable Cell Line - TrampC2 is a scientifically developed cell line specifically designed to stably overexpress mouse Fap in TrampC2 cells. The engineered cell line was derived from a mouse prostate epithelial cell line called transgenic mouse adenocarcinoma of the prostate (TRAMP). This particular cell line is therefore a valuable tool to study and understand the complexity and behavior of cellular processes associated with prostate epithelial cells." "Mouse Fap refers to the Fap gene present in mice. FAP (Fibroblast Activating Protein) is a serine protease used in a variety of biological functions and processes. FAP is commonly overexpressed during wound healing, tissue remodeling, inflammation, and fibrosis. However, it is best known for being highly expressed in more than 90% of epithelial cancers, including breast, colorectal, pancreatic, lung, and ovarian cancers, and is found exclusively in the tumor stroma. The unique expression pattern of FAP is primarily restricted to the tumor stroma in adults, making it an attractive target for novel cancer therapies. It is speculated that FAP exerts tumor-promoting effects by remodeling the extracellular matrix, promoting tumor growth, enhancing angiogenesis, and inhibiting anti-tumor immune responses. NIH_3T3 fibroblasts derived from mouse embryonic tissue. Their ability to continuously grow and multiply makes them ideal for creating large numbers of cells. The mouse Fap stable cell line – NIH_3T3 will be valuable to researchers interested in studying cancer, specifically the role of stromal cells and the function of fibroblast activating protein in tumor development. They can also be used to develop and test potential cancer treatments and explore the molecular pathways involved in tumor growth." "Mouse Agtr1a, also known as angiotensin II receptor type 1, is a receptor protein encoded by the mouse Agtr1a gene. This gene is part of the angiotensin receptor family, and the protein it encodes plays a key role in the renin-angiotensin system, helping to regulate blood pressure and fluid balance in the body. Once this receptor is activated, it stimulates various intracellular pathways, leading to physiological responses such as vasoconstriction, aldosterone synthesis and secretion, increased vasopressin secretion, cardiac hypertrophy, and extracellular matrix synthesis. Defects in the Agtr1a gene can lead to various health-related problems, such as hypertension, vascular remodeling, inflammation, and end-organ damage. Therefore, the Agtr1a gene and its protein product play an important role in maintaining cardiovascular homeostasis. It may become an effective target for the treatment of cardiovascular diseases. CHO-K1 cells are widely used cell lines in biology, and they can be genetically modified to produce a variety of protein products. This CHO-K1 cell line has been modified to stably express the mouse Agtr1a receptor. By introducing the gene encoding the receptor into CHO-K1 cells, the cells were able to produce the receptor and present it on their cell surface." "CYP7A1, also known as cytochrome P450 7A1, is a key enzyme in the human body and plays an important role in cholesterol metabolism. It is primarily responsible for converting cholesterol into bile acids, a critical step in maintaining cholesterol balance in the body. CYP7A1 is particularly important in the liver, where most cholesterol metabolism occurs. Its activity can be affected by a range of factors, from dietary cholesterol intake to various hormones and medications. Mutations in the CYP7A1 gene can cause certain inherited diseases, most notably cerebrotendinous xanthomatosis, a disease characterized by the accumulation of cholesterol in abnormal areas, causing a variety of symptoms. Due to its importance in cholesterol regulation, this enzyme is also a potential target for drugs aimed at lowering cholesterol levels. HeLa cells are a commonly used lab resource, derived from an immortal line of human cervical cancer cells originally taken from Henrietta Lacks in 1951. They are known for their robustness and adaptability, which can add an extra degree of reliability to studies using the Human CYP7A1 Stable Cell Line. Human CYP7A1 Stable Cell Line - HeLa is a cell line engineered to stably express CYP7A1. The stability of CYP7A1 expression in these cells makes them a reliable and consistent model for studying a variety of topics related to cholesterol metabolism and bile acid synthesis." "Coagulation factor III, also known as tissue factor (F3), is the protein encoded by the human F3 gene. F3 is normally found in subendothelial tissue and plays an important role in the body's blood coagulation process. When vascular integrity is compromised, F3 is exposed to the blood and initiates complex formation with coagulation factor VII or VIIa, thereby activating the coagulation pathway and promoting blood coagulation to prevent excessive bleeding. In addition to coagulation, F3 has other biological effects. Studies have found that it is also involved in angiogenesis, the formation of new blood vessels, which may contribute to tumor progression. Because of this correlation, F3 has also been studied in cancer research as a potential therapeutic target. Human F3 Stable Cell Line - HEK293 is a genetically engineered cell line specifically designed to express human F3. The cell line is derived from HEK293 cells, a specific type of cell commonly used in various forms of scientific research due to their ease of growth and transformation. By utilizing advanced genetic engineering techniques, these cells have been engineered to continuously produce human clotting factor III." "MGMT, or O-6-methylguanine-DNA methyltransferase, is an important enzyme that repairs DNA. Specifically, it repairs the DNA molecule at the O6-methylguanine site by transferring the methyl group at the lesion to the cysteine residue in its active site. This enzyme plays a vital role in the stability and integrity of genetic material within cells and is critical for preventing mutations that can lead to diseases such as cancer. Its primary function involves DNA repair, specifically the repair of alkylated DNA. Studies have shown that loss of MGMT function sensitizes cells to alkylating agents, forming the basis for one of the chemotherapy strategies. Mutations in this gene are associated with defects in DNA repair and increased susceptibility to certain types of cancer. Therefore, MGMT is a potential target for cancer therapy. Human MGMT Stable Cell Line - GL261 is a carefully engineered cell line that stably overexpresses human MGMT. GL261 refers to a specific glioma cell line known for its consistent and reproducible behavior in experimental settings. The human MGMT stable cell line - GL261 provides researchers with a powerful and reliable platform to study MGMT function, DNA repair mechanisms and related disease processes." "TAS2R39 is the protein encoded by the TAS2R39 gene in humans. It is located on chromosome 12. This gene is part of the taste receptor type 2 (TAS2R) gene family, which is a bitter taste receptor gene. The TAS2R gene encodes a G protein-coupled receptor that is part of a large family of human taste receptors. Functionally, these receptors are responsible for our ability to perceive the bitter taste of substances. Because of its responsibility for bitter taste reception, TAS2R39 is thought to play a crucial role in our ability to recognize and avoid harmful toxins in food. Research shows that the TAS2R39 gene plays an important role in food preferences, and that certain variants of the gene are associated with a dislike of bitter foods. Additionally, variations in this gene have been linked to certain health conditions, such as abnormal cholesterol levels and alcohol dependence. The human TAS2R39 stable cell line - HEK293 is a product specifically designed for biological and medical research, especially in the field of taste and other sensory research. HEK293 cells are very easy to grow in culture and have high transfection efficiency, making them ideal for creating stable cell lines. HEK293 cells are one of the most commonly used cell lines in biological and medical research, primarily due to their reliable growth and transfection propensity. The human TAS2R39 stable cell line - HEK293 has a wide range of research applications, such as pharmacological studies, drug screening, and taste studies." "SLC26A4, also known as Pendrin, is a gene located on chromosome 7q22.3 and is part of solute carrier family 26. This gene codes for a protein that acts as an anion exchanger. This protein is expressed in many tissues, such as the thyroid and kidneys, and its function is to transport chloride and iodide ions, playing a crucial role in the synthesis of thyroid hormones. It is also expressed in the inner ear and contributes to the formation and regulation of endolymph, a fluid necessary for normal hearing and balance. Mutations in the SLC26A4 gene are associated with several genetic disorders, such as Pendred syndrome and DFNB4 nonsyndromic hearing loss. Research also suggests that certain variations in the SLC26A4 gene may increase susceptibility to environmental goitrogens, which may increase the risk of thyroid disease in some individuals. Of note, genetic testing for this gene can be used to identify carriers of these mutations and can provide important clinical information for the treatment of patients with hearing loss and related conditions. The human SLC26A4 stable cell line - HEK293 provides a consistent and reliable biological system for research and development in a variety of applications. This cell line was developed from the parent cell line HEK293, known for its high transfection efficiency and broad range of growth properties. This cell line is critical for studying pathologies associated with dysfunctional SLC26A4 genes, such as Pendred syndrome and DFNB4 hearing loss. It also facilitates drug screening assays and pharmacological studies, helping researchers identify potential treatments for related conditions." "TRPV1 (transient receptor potential vanilloid 1) is a protein encoded by the TRPV1 gene in humans. It is a member of the TRPV group of the transient receptor potential family of ion channels. This protein is best known as a receptor for capsaicin, the active ingredient in chili peppers, and as a heat receptor in mammalian sensory systems. TRPV1 is a nonselective cation channel that is activated by a variety of exogenous and endogenous physical and chemical stimuli. TRPV1 is involved in the transmission and modulation of pain, as well as the integration of multiple pain stimuli. TRPV1 sensitivity to noxious stimuli, such as high temperatures, is not static but can be modulated by a variety of substances and conditions. Current research focuses on targeting this receptor to relieve pain in conditions such as neuropathic pain, osteoarthritis and cancer pain. The human TRPV1 stable cell line-HEK293 is a model specially developed for scientific research, especially in the fields of medical and biological research. Developed from human embryonic kidney 293 cells (commonly known as HEK293 cells), this cell line is easy to grow and transfect, making it popular in cell biology research. The human TRPV1 stable cell line - HEK293 has high expression levels of the human TRPV1 receptor, making it particularly suitable for experimental studies requiring high receptor signaling. It is proven and validated to ensure stable and long-term expression of TRPV1 receptors and consistently demonstrates robust responses to the TRPV1 agonists capsaicin and protons." "Angiotensin-converting enzyme 2 (ACE2) is an enzyme attached to the membranes of cells in the lungs, arteries, heart, kidneys and intestines. Its main function is to convert angiotensin II, a protein that constricts blood vessels, thereby raising blood pressure, into angiotensin-(1-7), a less potent molecule that relaxes blood vessels, thereby lowering blood pressure. Therefore, ACE2 plays a crucial role in blood pressure regulation. Additionally, since 2003, ACE2 has been recognized as a receptor for coronavirus entry into cells. Due to its important role in body function and infectious disease processes, the study of ACE2 is critical not only for developing drugs to treat high blood pressure and heart disease, but also for treating and preventing viral infections like COVID-19. CHO-K1 is a Chinese hamster ovary cell line that is a major cell type used in biological and medical research due to its rapid growth and high protein production capabilities. Human ACE2 Stable Cell Line - CHO-K1 is a stable cell line expressing human angiotensin-converting enzyme 2 (ACE2). This cell line has been genetically engineered to stably express human ACE2 protein, ensuring consistent and reliable experimental results." "The androgen receptor (AR) is a nuclear receptor that is activated by binding to androgens, testosterone, or dihydrotestosterone. AR is located in the cytoplasm and plays a crucial role in the function of the male reproductive system and the development of male secondary sexual characteristics. The AR gene is located on the X chromosome, and mutations in this gene can cause a variety of problems, such as androgen insensitivity syndrome. This receptor also plays a key role in the development and progression of prostate cancer. Overexpression or mutations in the AR gene may increase the risk of prostate cancer and determine its response to treatment. Human AR Knockdown Cell Line - HEK293 is a scientifically engineered cell line in which the AR (androgen receptor) gene has been significantly reduced or ""knocked down."" This cell line is derived from human embryonic kidney 293 cells, commonly referred to as HEK293. HEK293 is a special cell line that is widely used in cell biology and molecular biology research due to its ease of maintenance and robust growth in culture. AR knockdown in this cell line was achieved using small hairpin RNA (shRNA) technology. shRNA is designed to bind to AR messenger RNA (mRNA), preventing its translation into protein. This results in a significant reduction in AR protein expression, which helps elucidate the specific role and function of AR in a controlled laboratory setting." "Poly(ADP-ribose) polymerase 2 (PARP2) is a protein-coding gene responsible for post-translational modification of proteins through the addition of an ADP-ribose moiety. This includes a range of cellular processes such as DNA repair, cell cycle progression and cell death. It is part of the ADP-ribosyltransferase (ART) family and exists in different sized forms due to alternative splicing events within the gene. PARP2 plays a crucial role in maintaining genome stability and preventing mutations. It is essential for base excision repair (BER), a key pathway to repair damaged DNA throughout the cell cycle. It is also a promising therapeutic target, particularly in the treatment of cancer, since inhibition of its effects leads to synthetic lethality in certain types of cancer cells. Human PARP2 knockdown cell line - HeLa is a cell line specifically designed to study PARP2 gene function and behavior. It is an improved version of the widely studied HeLa cell line, a human cell line first isolated in 1951 from cervical cancer patient Henrietta Lacks. Specific changes have been made to this cell line to reduce the expression or function of the PARP2 gene, allowing scientists to better understand its role and impact in cellular processes as well as certain medical conditions such as cancer and neurodegenerative diseases." "Hep G2 is an immortal cell line that was isolated in 1975 from the liver tissue of a 15-year-old white Argentinian male with well-differentiated hepatocellular carcinoma. These cells are epithelial in morphology and are not tumorigenic in nude mice. Cells secrete several major plasma proteins, such as albumin and the acute-phase protein fibrinogen, α2-macroglobulin, α1-antitrypsin, transferrin, and plasminogen. The HepG2 cell line has the advantages of unlimited proliferation, stable phenotype, easy acquisition, and easy operation. Because Hep G2 cells are highly morphologically and functionally differentiated in vitro, they are a suitable model to study the intracellular trafficking and dynamics of bile canaliculi, sinusoidal membrane proteins, and lipids in human hepatocytes in vitro. Cas9 Stable Cell Line - HepG2 is derived from the human HepG2 liver cancer cell line and genetically engineered to stably express CRISPR-associated protein 9 (Cas9). Cas9 is a nuclease, an enzyme that cuts a DNA strand at a specific location, allowing scientists to accurately and efficiently edit or modify genes by adding, removing, or changing parts of the DNA sequence. Cas9 is an important component of the clustered regularly interspaced short palindromic repeats (CRISPR) system, a commonly used tool in gene editing. This cell line will benefit researchers who wish to use the CRISPR/Cas9 system for specific genetic manipulations, including gene knockout, insertion of specific genes or gene fragments, and precise gene editing or mutation in disease-related research." "Jurkat cells, derived from the peripheral blood of a 14-year-old patient with T-cell acute lymphoblastic leukemia (T-ALL), are a well-known human T lymphocyte cell line commonly used in cell biology research, particularly in cancer research and immune system disorder investigations. Jurkat cells produce interleukin-2 and are used in research involving cancer's sensitivity to drugs and radiation. Jurkat cells are commonly used in HIV research because they express CD4 receptors on their cell membranes. The CD4 receptor is the primary receptor used by HIV to enter host cells. Because Jurkat cells express this receptor, they can be infected by HIV, making them a useful model for studying HIV's interaction with human T cells, the virus's primary target in the body. The use of Jurkat cells in studies of HIV activation and the life cycle of HIV infection has greatly contributed to the understanding of viral interactions with human cells and helped to identify potential targets for antiretroviral therapy. Cas9 Stable Cell Line-Jurkat is a cell line that has been genetically engineered to stably express Cas9 endonuclease. The enzyme is a key component of the CRISPR/Cas9 system, a widely used genome-editing tool that allows genes to be added, deleted or changed within an organism's cells. The use of stable cell lines ensures consistent Cas9 expression, allowing for more reliable and efficient gene editing compared to traditional transient transfection methods. Additionally, this stable cell line eliminates the need for continuous transfection of cells with Cas9, increasing the efficiency and cost savings of genome editing experiments." "THP1 cells are a spontaneously immortalized monocyte-like cell line derived from the peripheral blood of a 1-year-old patient with monocytic leukemia and are an important model in immunology and cancer research. The THP-1 monocyte cell line is known for its ability to differentiate into mature macrophages and dendritic cells and is critical for studying the function and properties of these immune cells in vitro, including adipose tissue macrophages and M2 mononuclear phagocytes. THP-1 macrophages can polarize into M1 or M2 macrophages, which is critical for the study of immunity and inflammation, innate immunity, and inflammatory responses. Cas9 Stable Cell Line - THP1 is a genetically engineered cell line that is highly durable and efficient for gene editing applications. The cell line contains the Cas9 protein, a key component of the CRISPR-Cas9 gene editing system, stably integrated into its genome. The Cas9 protein is an RNA-guided DNA endonuclease that enables highly specific gene editing. Due to the stability of Cas9 integration, these cells can perform robust and reliable gene editing operations. The Cas9 stable cell line-THP1 was cultivated to ensure consistent expression of Cas9 protein, which greatly facilitates the CRISPR-Cas9 gene editing process. These cells are valuable tools for researchers aiming to study gene function, create disease models, or develop new treatment strategies." "CD274, also known as programmed death ligand 1 (PD-L1), is a protein encoded by the CD274 gene in humans. This protein is a member of the B7 family of cell surface ligands that regulate T cell activation and immune responses. CD274 is primarily expressed on antigen-presenting cells such as dendritic cells and macrophages and can be upregulated in response to inflammatory signals. It can also be expressed by a variety of other cell types, including T cells, B cells, endothelial cells, and various tumor cells. CD274 binds to its receptor PD-1 on T cells, inhibiting T cell receptor signaling, reducing cytokine production and T cell proliferation. This phenomenon is a key immune evasion mechanism exploited by tumor cells. This has led to the development of PD-1/PD-L1 blockade therapy in cancer treatment, which aims to enhance anti-tumor immune responses by blocking the interaction between PD-1 and CD274. Human CD274 Stable Cell Line - HEK293T is a highly advanced, efficient and stable cell line that can be used for a variety of biotechnology and scientific research purposes. HEK293T cells are derived from human embryonic kidney cells, are easy to grow and maintain, and are highly transfectable, making them easy to manipulate and introduce foreign genes. The CD274 protein is overexpressed in this cell line, allowing researchers to easily obtain large amounts of the protein for their studies." "CD40 is a costimulatory protein found on antigen-presenting cells and is critical for mediating multiple immune and inflammatory responses. It was first discovered in B cells and is a member of the tumor necrosis factor receptor superfamily. Binding of the CD40 protein ligand (CD40L) leads to numerous immune and inflammatory responses. The interaction between CD40 and its ligand (CD40L) is critical for the activation of B cells, dendritic cells, and macrophages, which in turn can lead to the production of specific antibodies against a variety of pathogens. In addition, due to its widespread expression and key role in immune activation and regulation, CD40 is considered a potential therapeutic target for a variety of diseases. Several CD40 antagonists have been developed and are in clinical trials for various autoimmune diseases and cancers. Human CD40 Stable Cell Line - HEK293T is a cell line that stably expresses the human CD40 protein. These stable cell lines can become excellent tools for drug discovery and development, providing a reliable platform to study the function of CD40, study its role in various immune responses, and screen and test potential drugs targeting CD40." "CD40LG, also known as CD40 ligand, is the protein encoded by the CD40LG gene in humans. It is a member of the TNF receptor superfamily. This protein is expressed on the surface of T cells and regulates B cell function by binding to CD40 on the surface of B cells. This protein is critical for mediating multiple immune and inflammatory responses, including T cell-dependent immunoglobulin class switching, memory B cell development, and germinal center formation. Mutations in this gene cause X-linked hyperIgM syndrome, a rare immunodeficiency characterized by defects in immunoglobulin class switch recombination. CD40LG has also been found to play an important role in various diseases including cancer and autoimmunity. Human CD40LG Stable Cell Line - CHO-K1 is a cell line genetically engineered to stably express the CD40 ligand (CD40LG) protein found in humans. This particular cell line is derived from Chinese Hamster Ovary (CHO-K1) cells. The CD40LG-CHO-K1 cell line is particularly valuable for scientific research in immunology, inflammation, and cancer. Researchers can use this cell line to study the function of the CD40/CD40LG interaction, which is important for immune regulation and homeostasis." "GPC3, also known as glypican 3, is a protein-coding gene belonging to the glypican family. Glypicans are heparan sulfate proteoglycans that are linked to the cell surface through glycosylphosphatidylinositol anchors. They are involved in the regulation of cell division and growth regulation. GPC3 mutations are associated with Simpson-Golabi-Behmel syndrome, a rare congenital disorder characterized by overgrowth and other abnormalities. Additionally, elevated levels of GPC3 were detected in the serum of patients with hepatocellular carcinoma, a type of liver cancer. GPC3 is expressed primarily in lung, ovary, and kidney cells and has the potential to interact with several other proteins, including members of the Wnt signaling pathway. Therefore, it plays an important role in cell communication and embryonic development. The human GPC3 stable cell line - CT26 is a powerful and reliable biological tool specifically designed for the purpose of studying GPC3 expression in a variety of studies and experiments. This stable cell line is based on the CT26 cell line, a murine colon cancer cell line. Although derived from a mouse model, it efficiently expresses human GPC3 and serves as a valuable model for understanding the function and impact of GPC3 in human biological processes. Therefore, the human GPC3 stable cell line - CT26 is an important resource for researchers in oncology, cell biology, and related fields." "NECTIN4, also known as Nectin cell adhesion molecule 4, is the protein encoded by the NECTIN4 gene in humans. This protein belongs to a family of cell adhesion molecules (CAMs) called nectins, which are involved in a range of biological processes such as cell migration, proliferation and differentiation. Nectin is best known for its role in the formation of adherens junctions, which are essential for maintaining tissue structure and function. Specifically, NECTIN4 has been implicated in the development and function of the nervous and immune systems. Recent studies have also suggested that NECTIN4 may function as a tumor marker, as it is frequently overexpressed in various types of cancer, including breast, ovarian, and lung cancers. Therefore, NECTIN4 is considered a potential target for cancer treatment. The CHO-K1 (Chinese Hamster Ovary-K1) host cell line is derived from the ovaries of Chinese hamsters and is specifically designed for protein production. Due to their efficient gene expression mechanism, they are widely used in biological and medical research. Human NECTIN4 Stable Cell Line - CHO-K1 is a cell line genetically engineered to stably express the human NECTIN4 gene. These cell lines are important tools for biomedical research, particularly in studying the role of the NECTIN4 gene in cancer initiation and progression and potential drug development." "Trem2 (myeloid cell-expressed triggering receptor 2) and Tyrobp (TYRO protein tyrosine kinase-binding protein) are two important proteins involved in the brain's immune response. Both proteins are most prominently expressed in microglia, the main immune cells of the central nervous system. Trem2 is a receptor that initiates protective microglial responses to injury, infection, and other threats. When Trem2 binds to its ligand, it triggers intracellular signaling pathways that lead to microglial activation, proliferation, and survival. Tyrobp, also known as DAP12, functionally interacts with Trem2 and plays a crucial role in immune responses. Tyrobp acts as an adapter molecule that transduces signals from Trem2 into the cell interior. Upon activation of Trem2, Tyrobp is phosphorylated and downstream signaling processes are initiated, leading to microglial activation. Mouse Trem2-Tyrobp Stable Cell Line - HEK293T is a scientifically engineered cell line in which human embryonic kidney cells (HEK293T) are stably modified to express mouse Trem2 and Tyrobp. This cell line can be effectively used for various research or drug discovery purposes. The expression and interaction between Trem2 and Tyrobp can be further examined in this cell line, providing important insights into their function and involvement in health and disease, particularly in age-related neurodegenerative diseases such as Alzheimer's disease." "CD274, also known as programmed death ligand 1 (PD-L1), is the protein encoded by the CD274 gene in humans. It is an immune protein or immunoglobulin found on the surface of cells. CD274 works by binding to the PD-1 receptor present on immune T cells, thereby suppressing the immune response. This process is essential for maintaining the balance of the immune system because it prevents the immune system from overreacting, which could damage the body's own cells. In diagnostic pathology, CD274 is of great interest as it serves as a biomarker for different types of cancer, including lung cancer, breast cancer, and melanoma. Therapeutic strategies involving blocking the interaction between CD274 and PD-1 are promising approaches in cancer immunotherapy. Studies have shown that using drugs that target the CD274 pathway improves survival in a variety of cancers. Human CD274/Luc Stable Cell Line - SHP-77 is a biological line scientifically engineered to possess and express the CD274 gene in a stable and consistent manner. Additionally, it contains a luciferase reporter gene that facilitates simple and easy tracking and visualization of proteins produced by the CD274 gene. The SHP-77 cell line is a cell line derived from human small cell lung cancer and is commonly used to study and understand the behavior of this type of cancer at the cellular level. The human CD274/Luc stable cell line - SHP-77 is a valuable tool in cancer biology research, helping to explore the role of PD-L1 in lung cancer and investigate potential therapeutic strategies targeting this pathway." "Epithelial cell adhesion molecule (EpCAM), originally identified as a cancer marker, is a transmembrane glycoprotein that mediates Ca2+-independent homogeneous intercellular adhesion in epithelial cells. It is expressed on the basolateral cell surface of most human epithelial cells. In addition to functioning as an adhesion molecule, EpCAM actively interferes with cellular processes such as proliferation, signaling, and migration. EpCAM has been reported to be involved in various complex events of cell adhesion, migration, and proliferation during carcinogenesis. EpCAM is also a recognized marker of cancer stem cells (CSCs) in many types of tumors, and detection of EpCAM+ circulating tumor cells (CTCs) in a patient's blood, bone marrow, or lymph nodes indicates a poor prognosis. Therefore, EpCAM is considered a promising target for cancer therapy. Human EPCAM Stable Cell Line - HEK293 was created from a specific human cell line called HEK293, which stands for human embryonic kidney 293. These cells are genetically modified to stably express EPCAM. The stable expression of EPCAM in the human EPCAM stable cell line - HEK293 makes these cells a useful tool for drug screening and development and also allows scientists to study the downstream signaling pathways in which EPCAM participates. Furthermore, this cell line could prove to be a valuable resource for developing potential EPCAM therapies for various diseases." "EML4-ALK, short for echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase, is a fusion gene that has been identified as a driver mutation in a subgroup of non-small cell lung cancer (NSCLC). It was first described in 2007 and has since been found in approximately 3-7% of NSCLC patients, making it a relatively rare mutation. The fusion gene is the result of a chromosomal inversion within the short arm of chromosome 2, resulting in a chimeric gene that combines part of the EML4 gene with part of the ALK gene. The product of this fusion gene is a constitutively active tyrosine kinase that promotes cell proliferation and survival, thereby promoting cancer development. The human EML4-ALK stable cell line-Ba/F3 is an important tool for cancer research. This cell line is essentially a disorder of recombinant constructs. The Ba/F3 cell line is an interleukin-3-dependent pre-B cell line derived from mice that maintains the growth, proliferation, and survival of these modified cells. It expresses human microtubule-associated proteins such as the abnormal fusion protein of four anaplastic lymphoma kinases (EML4-ALK). The human EML4-ALK stable cell line Ba/F3 plays an important role in cancer research in understanding the characteristics of this oncogene and in developing targeted therapeutics against this oncogene." "The epidermal growth factor receptor (EGFR) gene plays a crucial role in controlling cell growth, division, and replication because it provides instructions for the synthesis of proteins that span cell membranes and is involved in pathways that promote cell division. EGFR gene mutations, such as DEL19, mainly occur in patients with non-small cell lung cancer (NSCLC). These genetic mutations tend to be more common in individuals who have never smoked, and they often respond favorably to targeted treatments such as tyrosine kinase inhibitors. Essentially, EGFR-DEL19 mutations indicate a subtype of lung cancer with specific characteristics and treatment options. Human EGFR-DEL19 stable cell line-Ba/F3 is a cell line developed by modifying the parent cell Ba/F3 to stably express human epidermal growth factor receptor (EGFR) with exon 19 deletion mutation (DEL19) . It is mainly used in scientific research, especially in the field of cancer research, and plays an important role in the study of the pathogenesis of NSCLC. Exon 19 deletion mutations are associated with NSCLC, and tumors harboring this mutation often respond to tyrosine kinase inhibitors. Therefore, the human EGFR-DEL19 stable cell line-Ba/F3 provides a powerful tool to explore the molecular mechanisms of EGFR signaling, thereby facilitating the development of targeted therapies for the treatment of EGFR-driven cancers." "The epidermal growth factor receptor (EGFR) gene encodes a protein that spans cell membranes and is essential for regulating cell growth and division. In healthy states, the EGFR protein contributes to the normal cycle of cell death and renewal. However, mutations within this gene can cause cells to grow uncontrolled and form malignant tumors. The EGFR-L858R mutation is caused by a substitution of amino acid 858 from leucine (L) to arginine (R) in the protein. This mutation increases the sensitivity of the receptor to EGF, leading to uncontrolled cell division. However, the T790M mutation is caused by a substitution of threonine (T) to methionine (M) at position 790 in the EGFR protein. This mutation leads to resistance to first- and second-generation tyrosine kinase inhibitor (TKI) drugs, posing a major challenge in the treatment of EGFR-positive lung cancer. Human EGFR-L858R/T790M Stable Cell Line-Ba/F3 provides an ideal in vitro model to study the biology of these two important EGFR mutations and to develop therapeutic strategies for EGFR-mutated lung cancer. This cell line was maintained under stable conditions and grew in an interleukin-3 (IL-3)-independent manner. It exhibits high sensitivity to EGFR tyrosine kinase inhibitors in proliferation assays." "FLT3 (or FMS-related tyrosine kinase 3) is a protein that is critical for the maturation of hematopoietic progenitor cells (cells that eventually develop into blood cells). FLT3 plays a crucial role in cell development, particularly in the production and development of blood cells in the bone marrow. In a healthy body, the FLT3 gene helps control how quickly and how many cells the body produces. However, mutations in the FLT3 gene can lead to the overproduction of immature blood cells, a condition that can lead to acute myeloid leukemia (AML). Due to its association with cancer, FLT3 has become an important area of research in cancer research. There are currently several therapies and drugs that target FLT3 in development with the goal of treating or preventing leukemia and other similar diseases. Human FLT3 Stable Cell Line-Ba/F3 is engineered to stably integrate the human FLT3 gene into Ba/F3 cells. This specific feature of the human FLT3 stable cell line-Ba/F3 is particularly useful for the study of AML, as well as drug screening studies to identify potential therapeutic targets in this process. This cell line provides researchers with an important tool to understand the function of FLT3 and the effects of mutations." "EGFR-A763_Y764insFQEA is a mutation found in the epidermal growth factor receptor (EGFR) gene. This is a specific type of insertional mutation, which means that extra DNA base pairs (in this case, FQEA) are added to the coding sequence at positions 763 and 764. In normal circumstances, the EGFR gene produces a protein that helps cells, particularly skin cells, to grow and divide. However, the A763_Y764insFQEA mutation causes the gene to function abnormally, which often leads to uncontrolled cell growth and the development of potential cancer, most commonly lung cancer. This type of mutation is typically discovered through DNA sequencing or genomic testing and, although rare, may be important in personalized cancer treatment, particularly in predicting the likelihood of response or resistance to certain targeted therapies. The human EGFR-A763_Y764insFQEA stable cell line - BaF3 is a powerful research tool designed to study specific forms of EGFR involved in tumor proliferation and resistance to EGFR-targeted therapies. The BaF3 cell line is a mouse pro-B cell line that lacks expression of functional EGFR, making it a suitable background for expressing mutant forms of EGFR. The human EGFR-A763_Y764insFQEA stable cell line could help researchers develop and test new treatments for cancers caused by this specific EGFR mutation. This cell line is ready-to-use and guaranteed to provide consistent research results." "The KIT gene is located on the long arm of chromosome 4 and is a proto-oncogene that plays a key role in cell survival, proliferation and differentiation. It contains 21 exons encoding a 976-amino-acid long protein named c-Kit, which belongs to the tyrosine kinase receptor family. KIT-D816V is a mutation commonly associated with mastocytosis or systemic mastocytosis. The KIT-D816V mutation is present in the oncogene KIT and causes a change in amino acid position 816 from aspartate (D) to valine (V). The presence of the KIT-D816V mutation, which is used as a diagnostic tool in systemic mastocytosis, may also influence treatment decisions, as targeted therapies against the KIT receptor have shown efficacy in treating this disease. Human KIT-D816Y Stable Cell Line - BaF3 is derived from the BaF3 cell line, a mouse pro-B cell line engineered to stably express the human KIT D816Y mutation. The D816Y mutation in the KIT gene is commonly associated with certain chronic and acute myeloid leukemias, systemic mastocytosis, and gastrointestinal stromal tumors. The KIT-D816Y stable cell line - BaF3 provides a useful model for studying KIT pathway-related diseases, allowing researchers to test potential therapeutic strategies for patients with this mutation." "FLT3 (or FMS-like tyrosine kinase 3) is a protein encoded by the human FLT3 gene. It is a cell surface receptor for cytokines and is involved in the regulation of hematopoiesis (the process of blood cell formation). FLT3 is expressed on the surface of many hematopoietic progenitor cells. Activating mutations in the FLT3 gene are one of the most common genetic abnormalities in acute myeloid leukemia (AML), and internal tandem duplications (ITDs) of the FLT3 gene are particularly associated with poor prognosis. The FLT3-ITD-D835Y mutation means a change in amino acid 835, replacing aspartate (D) with tyrosine (Y). The presence of this mutation affects treatment options for patients with AML. Therefore, early detection of this mutation is crucial for optimal patient management. Human FLT3-ITD-D835Y stable cell line - BaF3 is a cell line genetically engineered to express the FLT3-ITD-D835Y mutation. The specific mutations expressed in this cell line, FLT3-ITD-D835Y, involve the insertion of a repeat sequence (ITD) and a point mutation that changes the amino acid at position 835 from aspartate to tyrosine (D835Y). This BaF3 cell line is derived from mouse pro-B cells and is widely used in drug screening and biomedical research. It is a stable cell line, which means that the quality and properties of the cells remain unchanged even after many generations of growth. These cells provide a valuable model system for studying the role of FLT3 mutations in hematological malignancies and can be used to test novel therapies targeting the FLT3-ITD-D835Y mutation." "Insulin-like growth factor 1 receptor (IGF1R) is a member of the receptor tyrosine kinase (RTK) family and plays a critical role in regulating cellular functions, including cell cycle progression and energy metabolism. This receptor primarily binds to one of its ligands, insulin-like growth factor 1 (IGF1), and triggers a series of downstream signaling pathways that promote cell growth, proliferation, and survival. Dysregulation of the IGF1R signaling pathway is associated with different diseases, especially cancer. IGF1R has been reported to be overexpressed in several types of solid tumors, including breast, lung, and prostate cancer, suggesting that it may serve as a potential target for cancer therapy. In addition to cancer, alterations in IGF1R signaling have been associated with diabetes, cardiovascular disease, and neurological disease. Human IGF1R Stable Cell Line - BaF3 is a cell line engineered to express the insulin-like growth factor 1 receptor (IGF1R). The host for this stable expression system is the BaF3 cell line, derived from mouse bone marrow. These cells are particularly important for researchers to study the role and function of IGF1R in human health and disease, as well as during drug development. They can provide insights into the pathophysiology of diseases related to insulin signaling, including diabetes and various cancers." "Epidermal growth factor receptor (EGFR) is a protein found on the surface of certain cells and plays a key role in cell growth and survival. When a molecule called a ligand binds to EGFR, it triggers a series of cellular events that promote cell growth and division. This binding stimulates the intrinsic protein tyrosine kinase activity of the receptor, resulting in autophosphorylation of several tyrosine residues in the C-terminal domain. EGFR-T790M/C797S/L858R refers to three specific mutations in the epidermal growth factor receptor (EGFR) gene. These mutations are often associated with non-small cell lung cancer. The EGFR-T790M mutation is often referred to as a “gatekeeper” mutation because of its role in the development of resistance to first- and second-generation tyrosine kinase inhibitors (TKIs), a common treatment for lung cancer. This mutation changes the structure of EGFR, thereby reducing the drug's ability to bind to and inhibit the receptor. The EGFR-C797S mutation often occurs after treatment with third-generation TKIs, leading to resistance to these drugs. The C797S mutation changes the shape of the EGFR protein and reduces the efficacy of TKIs. EGFR-L858R mutation is a common EGFR activating mutation. It makes cells more likely to grow and divide in an uncontrolled manner, which often leads to the development of cancer. Human EGFR-T790M/C797S/L858R Stable Cell Line - NCI-H1975 is a unique form of cell line genetically engineered to express three specific mutations in the epidermal growth factor receptor (EGFR) gene. Derived from the NCI-H1975 human lung adenocarcinoma cell line, this cell line provides an exciting tool for researchers seeking to explore the complex dynamics of lung cancer pathology, drug resistance, and potential targeted therapies." "MERTK, also known as Mer receptor tyrosine kinase, is the protein encoded by the MERTK gene in humans. This gene is a member of the MER/AXL/TYRO3 receptor kinase family and encodes a transmembrane protein with two fibronectin type-III domains, two Ig-like C2-type (immunoglobulin-like) domains, and two tyrosine kinase domains. This receptor transduces signals from the extracellular matrix to the cytoplasm by binding to vitamin D3 metabolites or integrins. It is involved in normal cellular processes as well as immune responses, blood clotting, and maintaining the integrity of the eye's photoreceptors. Dysfunction or absence of MERTK leads to diseases such as retinitis pigmentosa and cone dystrophy. The human MERTK stable cell line - THP-1 is a unique tool for researchers studying the Mer proto-oncogene tyrosine kinase (MERTK). This stable cell line is based on the human THP-1 monocyte cell line, which has been genetically modified to overexpress the MERTK gene. The THP-1 cell line is derived from human acute monocytic leukemia patients and is a widely used in vitro model of monocyte/macrophage differentiation and function. They are particularly useful for drug discovery and development, allowing researchers to study the biological effects of potential therapeutic compounds on MERTK expression and function in a controlled, reproducible environment." "EML4-ALK, also known as echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase, is a fusion gene that combines part of the EML4 gene with part of the ALK gene. This fusion is often caused by chromosomal rearrangements, specifically a type of translocation that is most common in certain types of non-small cell lung cancer (NSCLC). EML4 is a gene that codes for a protein involved in the organization of microtubules, components of the cytoskeleton required for cell shape, function, and division. ALK, on the other hand, stands for anaplastic lymphoma kinase, an enzyme that in its normal state is active primarily in cells of the nervous system during early development. When it becomes active in cells where it is not normally present, it can promote cell growth and division in an uncontrolled manner. The G1202R-L1196M point mutation refers to a specific change in the DNA sequence of the EML4-ALK fusion gene. This mutation further increases the activity of the EML4-ALK protein, making it more effective in driving cancer progression. The G1202R mutation occurs at position 1202 of the ALK domain, in which glycine (G) is replaced by arginine (R), while the L1196M mutation occurs at position 1196, in which leucine (L) is replaced by methionine (M) replace. Research is currently underway to develop drugs that effectively target and inhibit the EML4-ALK fusion protein harboring the G1202R-L1196M mutation, with the goal of improving and personalizing cancer treatment options for patients harboring these genetic alterations." "The HRE-Luc Reporter Cell Line-HeLa is a unique tool that aids researchers by providing a simple and accurate method to study the hypoxic response of cancer cells. This cell line is derived from human cervical cancer cells (HeLa cells) and is genetically engineered to express luciferase, a bioluminescent enzyme, under the control of a hypoxia response element (HRE). HRE is a regulatory DNA sequence that is activated under low oxygen or hypoxic conditions. The key feature of the HRE-Luc reporter cell line HeLa is its ability to emit light in response to hypoxic conditions. When cells are starved of oxygen, it activates specific genes that allow the cells to adapt and survive. One of these genes is regulated by hypoxia-inducible factor (HIFA), which binds to the HRE and initiates the transcription of various adaptive genes. In this engineered cell line, the presence of luciferase under HRE control allows real-time monitoring of the hypoxic response by measuring the amount of light produced." "NFAT-Luc/KDR Stable Cell Line - HEK293T is a unique and powerful tool designed for analyzing NFAT (nuclear factor of activated T cells) activity and studying KDR (kinase insert domain receptor) related functions. This cell line was genetically engineered from the human embryonic kidney 293T line to stably integrate a luciferase reporter gene under the control of an NFAT response element. The NFAT transcription factor plays a crucial role in immune response, heart valve development, blood vessel development, and various other physiological processes. KDR, also known as VEGFR-2, is a major transducer of VEGF-mediated signals within the cardiovascular system, making this cell line highly beneficial for research in these areas. The presence of a luciferase reporter gene in this cell line allows for convenient detection and measurement of intracellular NFAT-dependent transcriptional activity. This makes the NFAT-Luc/KDR stable cell line – HEK293T very useful for high-throughput screening applications, drug discovery and development, and for fundamental understanding of NFAT and KDR biology." "Luciferase is the general term for a class of oxidative enzymes that produce bioluminescence. It is extracted from fluorescein, and with the help of oxygen, it undergoes a series of reactions that cause it to glow. Luciferase is the basis of scientific research in various fields, especially in the field of genetics, where it is often used as a reporter gene to examine the activity of other genes. Luciferase reporter genes are paired with target signaling pathways or specific gene promoters to indicate the activity of the target pathway or gene expression through luminescence levels. The luminescence signal is easy to measure, greatly aiding researchers in drug screening applications or exploring gene function. The luciferase reporter stable cell line-SKOV-3 has the luciferase gene exogenously integrated into the SKOV-3 cell genome. SKOV-3 is a human ovarian cancer cell line first isolated in 1973 from the malignant ascites of a 64-year-old white woman with ovarian adenocarcinoma. SK-OV-3 cells are commonly used in cancer research to study ovarian cancer biology, drug resistance mechanisms and explore potential therapeutic targets. The luciferase reporter stable cell line-SKOV-3 is a reliable tool to study the activity of specific genes or signaling pathways and provides a powerful platform for drug screening applications." "BT-474 is a human breast cancer cell line established from primary tumors of breast cancer patients. The BT474 cell line has several properties that make it a useful tool in cancer research. It can grow in culture and exhibit many of the same properties as breast cancer cells found in patients. Researchers used the BT-474 cell line to study the molecular mechanisms of breast cancer, including genes and pathways involved in cancer cell growth, invasion and metastasis. The BT-474 cell line is also used to test the efficacy of various cancer treatments, including chemotherapy, radiation therapy and targeted therapy. The GFP stable cell line-BT474 is generated by transforming BT474 cells with a plasmid carrying the GFP gene, resulting in stable integration of the gene into the genome of the BT474 cells. The GFP stable cell line-BT474 enables researchers to monitor the in vitro and in vivo behavior of BT474 subtype breast cancer cells under a variety of experimental conditions. GFP fluorescence provides a real-time, non-invasive indication of cell status, allowing for continuous monitoring and analysis. It can be used to study proliferation, migration, invasion and protein localization as well as other cellular functions in living organisms or populations of living cells in real time. Therefore, the GFP stable cell line-BT474 is a valuable tool in various areas of biomedical research, including cancer biology, drug discovery and development, cell biology, and preclinical research." "HT-29 is a human colon cancer cell line widely used in biological and cancer research. HT-29 cells, originally extracted from a 44-year-old white woman in 1964 by Jorgen Fogh, form a compact monolayer while displaying similarities to the intestinal epithelial cells of the small intestine. In preclinical studies, HT-29 cells were studied for their ability to differentiate to mimic real colon tissue in vitro, a property that makes HT-29 useful for epithelial cell studies. This cell line is a suitable transfection host and has applications in cancer and toxicology research. Green fluorescent protein (GFP) is a versatile biomarker used to monitor physiological processes, visualize protein localization, and detect transgene expression in vivo. When excited by blue light (maximum wavelength 395 nm), it can emit green light (maximum wavelength 509 nm). By combining the advantages of GFP and HT-29 cell lines, GFP stable cell line-HT-29 provides researchers with a valuable tool. It can be used in bioimaging studies, studying the cellular behavior of cancer cells, and testing the effectiveness of drugs against colorectal cancer." "NCI-H1299, also known as H1299, is a cell line established from lung lymph node metastases of a 43-year-old white male cancer patient. H1299 and H292 are non-small cell lung cancer (NSCLC) cell lines. Regarding their genetic characteristics, H1299 cells have a homozygous partial deletion of the p53 protein and lack expression of the p53 protein. These cells are widely used to study lung cancer biology, drug resistance, and potential treatment options. Luc/GFP reporter cell line-NCI-H1299 is a genetically engineered lung cancer cell line used in biomedical research. This cell line has been modified to express two proteins: luciferase (Luc) and green fluorescent protein (GFP). Luciferase is an enzyme that produces light through a chemical reaction. This bioluminescence can be used to measure the activity of specific cellular processes. GFP is a protein that emits green light when exposed to light in the blue to ultraviolet range. This fluorescence can be used to visualize cellular structures and processes, track the movement of individual cells, and monitor gene expression. The presence of luciferase and GFP allows dual monitoring of cellular activity, providing the opportunity to obtain more detailed and comprehensive data." "4T1 is a breast cancer cell line derived from mouse BALB/c strain mammary tissue. 4T1 cells are epithelial cells that are resistant to 6-thioguanine. In preclinical studies, 4T1 cells have been used to study breast cancer metastasis because they can metastasize to the lungs, liver, lymph nodes, brain and bone. 4T1 is similar to human metastatic triple-negative breast cancer (TNBC) and lacks expression of the estrogen receptor (ER), progesterone receptor (PR), and epidermal growth factor receptor 2 (HER2). Furthermore, this cell line is poorly immunogenic in mice, consistent with the characteristics of human breast cancer, resulting in higher tumorigenicity and invasiveness in this breast cancer model. Luc/RFP Reporter Cell Line-4T1 is engineered to stably overexpress Luc (luciferase) and RFP (red fluorescent protein). Luciferase is an enzyme that produces bioluminescence, allowing cells in living animals to be easily detected through bioluminescence imaging. Red fluorescent protein, on the other hand, helps observe the location and spread of cancer cells through fluorescence microscopy. Under the influence of certain substances or conditions, the intensity of fluorescence and bioluminescence produced by these labeled cells increases or decreases. This allows researchers to monitor the effects of various treatments or stimuli on cancer cells in real time." "A549 cells are adenocarcinoma human alveolar basal epithelial cells, a cell line first developed by D. J. Giard et al. in 1972. Cancerous lung tissue was removed and cultured from a transplanted tumor in a 58-year-old white man. A549 cells are found in the lung tissue from which they originate, are squamous, and are responsible for the diffusion of certain substances, such as water and electrolytes, into the alveoli. These cells are capable of synthesizing lecithin and contain high levels of unsaturated fatty acids, which are important for maintaining membrane phospholipids. A549 cells are used as a model for lung cancer research and to develop drug therapies for lung cancer. Luc Reporter Cell Line-A549 is a transgenic human lung cancer cell line that stably expresses the luciferase gene, making it a valuable tool in a variety of scientific research settings. Luciferase is an enzyme that produces light when it oxidizes a substrate. There are different types of luciferase, such as firefly luciferase and Renilla luciferase, each with its own unique characteristics such as emission wavelength and reaction speed. Luciferase has many applications in scientific research, including gene expression assays, bioluminescence imaging, and bacterial detection. This specific cell line type is designed for convenient high-throughput screening of therapeutic agents as well as in vivo monitoring of tumor growth in xenograft models." "SK-BR-3 cells were derived from pleural effusion caused by breast cancer in a 43-year-old white female patient, and overexpressed the HER2/neu product, which is believed to be related to various breast cancer proliferation pathways. SK-BR-3 cells are known to grow in grape-like clusters and have an invasive phenotype similar to cells in vivo. SK-BR-3 cells have been used in studies seeking to overcome trastuzumab resistance in breast cancer that overexpresses human epidermal growth factor receptor 2. The RFP reporter cell line – SK-BR-3 is a valuable tool for researchers studying cancer biology, specifically the progression and mechanisms of breast cancer. One of the key features of this cell line is the incorporation of red fluorescent protein (RFP), which enables live cell imaging and real-time tracking of cellular processes. RFP, like GFP, maintains its fluorescence without the need for added cofactors or substrates, making it particularly convenient for use in a variety of experimental settings. The RFP reporter gene is stably integrated into the SK-BR-3 cell line, ensuring its consistent expression and subsequent generation of bright red fluorescence - a feature that can be exploited in a variety of experimental settings. These cells exhibit strong RFP signals that allow easy detection and rapid data collection." "The HuH-7 cell line was established in 1982 from a well-differentiated hepatocyte-derived carcinoma cell line originally derived from a liver tumor of a 57-year-old Japanese man. This cell line was established by Nakabayshi, H. and Sato, J.. Huh-7 is an immortal cell line composed of epithelioid tumorigenic cells. Most Huh-7 cells have chromosome numbers between 55 and 63 (pattern 60) and are highly heterogeneous. Huh7 cells have played an important role in hepatitis C research. Until 2005, it was impossible to culture hepatitis C in the laboratory. The introduction of the Huh7 cell line allows screening of drug candidates against laboratory-grown hepatitis C virus and allows the development of new drugs against hepatitis C. The GFP reporter cell line - Huh7 is a powerful tool for monitoring gene expression in real time. The cells, derived from the human liver cancer cell line Huh7, were genetically engineered to express green fluorescent protein (GFP) when a specific promoter is activated. After transfection with a specific gene of interest, the GFP reporter in these cells fluoresces green when exposed to light in the blue to ultraviolet range. This characteristic luminescence can serve as an indicator of gene expression, allowing researchers to visually monitor the transcriptional activity of genes of interest qualitatively and quantitatively." "A20 cells are a cell line originally derived from B-cell lymphoma in aged BALB/c mice. A20 cells are suitable for in vitro and in vivo experiments. These cells will form tumors and spontaneous metastases in immunocompromised and syngeneic Balb/c mice. Depending on the route of vaccination, implanted A20 cells can transfer to many sites, including bone marrow, liver, spleen, lymph nodes, ovaries, and peritoneal cavity. A20 cells are used in medical research such as drug screening or vaccine target selection. A20 cells are also highly sensitive to immunomodulatory antibodies and are therefore often used in immunotherapeutic drug research. Luciferase Reporter Cell Line - A20 is a genetically engineered cell line designed to express luciferase, a bioluminescent enzyme, under specific biological conditions. The luciferase gene in this reporter cell line acts as a reporter gene, meaning it produces a measurable phenotype when a specific biological process is activated or inactivated. Because luciferase produces light when it is enzymatically active, luciferase expression levels in the A20 cell line can be easily measured using bioluminescence imaging or a photometer." "MIA PaCa-2 is a human pancreatic cancer cell line widely used in pancreatic cancer research and therapeutic development. In 1977, MIA PaCa-2 cells were derived from cancer in a 65-year-old man. These cells display CK5.6, AE1/AE3, E-cadherin, vimentin, chromogranin A, synaptophysin, SSTR2, and NTR1, but not CD56. This cell line is commonly used as a model system in cancer research to study the properties and behavior of pancreatic cancer cells and for drug screening and development. GFP Stable Cell Line - MIA PaCa-2 is engineered to stably express GFP. Originally isolated from Aequorea victoria, GFP is a luminescent protein that exhibits bright green fluorescence when exposed to light in the blue to ultraviolet range. When inserted into cells, the protein essentially makes the cells glow, which helps them be tracked more easily under a fluorescence microscope. The MIA PaCa-2 GFP stable cell line combines the unique qualities of the parent cell line, such as high levels of transfection efficiency and growth robustness, with the unparalleled advantages of fluorescent proteins. GFP integration provides a visual aid that researchers can use to scrutinize cellular components, track the dynamic movement of cells, and study genetic regulation within cells." "Colon-26 is a mouse colon adenocarcinoma cell line derived from the tumor tissue of Balb/c mice bearing Colon-26 carcinoma, induced by a single rectal application of N-Nitroso-N-Methyl-urethan (NMU). Colon-26 cells are used to create a mouse model of cachexia for the study of cancer cachexia, a syndrome of progressive loss of skeletal muscle mass and fat tissue that results in weight loss and weakness. Additionally, the colon-26 cell line can be used for metastasis studies. Injection of the colon-26 cell line into the spleen of wild-type mice induces tumor necrosis factor alpha (TNF-α) expression and subsequent liver metastasis formation, which may be mediated by tumor necrosis factor receptor p55 signaling. Luciferase Stable Cell Line - Colon26 is a genetically engineered cell line modified to express luciferase. The introduction of the luciferase gene into the Colon26 cell line allows measurement of cellular activity through luminescence. This is particularly useful in the field of biomedical research, where it is often used in in vivo imaging studies to track tumor growth and metastasis, monitor gene expression, or evaluate the effectiveness of therapeutic interventions." "Huh7 is an immortalized cell line that can be grown in the laboratory for research purposes. Huh7 is a well-differentiated hepatocyte-derived cancer cell line originally derived from a liver tumor in a 57-year-old Japanese man in 1982. These cells adhere to the surface of a flask or plate and typically grow as a 2D monolayer. Although containing many mutations and INDELS, it is noteworthy that Huh7 cells harbor point mutations in the p53 gene. Huh-7 is highly sensitive to hepatitis C virus (HCV) and is often used as a model to study liver cancer and HCV. This cell line can be used in HCV replicon systems, allowing the production of infectious HCV particles in vitro and the development of drugs against HCV. GFP/Luc Reporter Cell Line - Huh7 is engineered to stably express the genes encoding GFP and luciferase. The main advantage of this dual reporter system is that detection of GFP can be immediately visualized under a fluorescence microscope for cell sorting and tracking, while the luciferase allows non-invasive imaging of cells and animals in the bioluminescent mode, enabling long-term monitoring. Additionally, it provides the ability to generate quantitative data, making it a powerful tool for high-throughput screening applications." "MC38 is a mouse colon adenocarcinoma cell line originally derived from colon tumors induced by the chemical carcinogen azomethane in C57BL/6 mice. These cells have been widely used as a model system to study various aspects of colon cancer biology, including the molecular mechanisms of tumor initiation and progression, and to identify potential therapeutic targets for colon cancer treatment. MC38 cells have a variety of properties. First, they are relatively easy to cultivate in the laboratory and can be grown in large quantities. Second, they express markers of colon cancer, such as carcinoembryonic antigen (CEA) and cytokeratin, and exhibit some characteristics of colon cancer, including uncontrolled proliferation, invasion, and metastasis. Third, they respond to various stimuli, such as cytokines and growth factors, making them useful model systems to study the effects of these factors on colon cancer cell growth and survival. Luciferase Reporter Cell Line - MC38 is a genetically engineered variant of the widely studied MC38 murine colon cancer cell line. In this enhanced cell line, cells are used to express the gene for luciferase, a bioluminescent protein originally derived from fireflies. With this modification, cells are able to produce light in response to certain stimuli, which can then be measured using bioluminescence imaging techniques. This property makes the luciferase reporter cell line - MC38 an important tool for a variety of biomedical research, including studies of cancer progression, cellular immune responses, and the effectiveness of potential new drugs." "PC-9, formerly known as PC-14, is a human non-small cell lung cancer (NSCLC) cell line harboring a deletion mutation in exon 19 of the EGFR gene. It originates from undifferentiated human lung tissue adenocarcinoma and consists of a heterogeneous mixture of round and spindle-shaped cells. PC-9 cells exhibit adherent and suspension growth properties. They are used to study the effect of alpha-tocopherol on cytotoxic drugs, investigating whether it enhances or diminishes their effectiveness against NSCLC. In addition, PC-9 cells can be used as a model to study the resistance of lung cancer to tyrosine kinase inhibitors (TKIs) and help to understand the related mechanisms. GFP/Luc Reporter Cell Line - PC-9 is a bioengineered cell line designed for biomedical research and drug testing purposes. It is derived from the human lung cancer cell line PC-9. This cell line has been genetically engineered to stably express green fluorescent protein (GFP) and luciferase (Luc) reporter genes. GFP is a protein that fluoresces green when exposed to light in the blue to ultraviolet range, making it easy to visualize and monitor in real time. Luc, on the other hand, is a luminescent enzyme found in fireflies that produces a detectable bioluminescent signal in the presence of a luciferin substrate." "The MC38 adenocarcinoma colorectal cell line is a well-established and frequently used tumor model for preclinical research on neoantigens and immunotherapeutic approaches. This transplantable cell line was established in 1975 by repeatedly injecting mice with the carcinogen dimethylhydrazine and thus has the characteristics of tumors with a high mutational load. MC38 cells exhibit similar characteristics to human colon adenocarcinoma, making them valuable for studying this disease. They have an epithelial morphology and are capable of forming tumors when implanted in animal models. Researchers are using MC38 to study genetic alterations, signaling pathways, immune checkpoint regulation and drug sensitivity in colorectal cancer. It has been widely used to evaluate the efficacy of various treatments, including chemotherapy, immunotherapy, and combination therapies. DsRedExpress Reporter Cell Line - MC38 is a unique and innovatively developed research tool that is widely used in a variety of scientific research. This specially engineered cell line is derived from MC38 cells. The main feature that distinguishes this cell line is the stable expression of the DsRedExpress2 fluorescent protein. The reporter emits red fluorescence, allowing easy and precise detection and analysis of cells. Scientists frequently exploit this capability for in vivo imaging of tumors and other biological structures, as well as for tracking cell proliferation, migration, and protein expression in real time. In addition, unlike green fluorescent protein (GFP), the red fluorescence of DsRedExpress does not cause phototoxic effects, making it safer and less damaging to cells during experiments." "A20 is a murine B lymphoma cell line derived from BALB/c mice with spontaneous reticulum tumors. It is commonly used as a model to study B-cell lymphoma and its response to various treatments. The A20 cell line has several properties that make it a useful tool in cancer research. It was able to grow in culture and was tumorigenic when injected into mice. Researchers used the A20 cell line to study the molecular mechanisms of B-cell lymphoma, including genes and pathways involved in cancer cell growth and survival. The A20 cell line is also used to test the efficacy of various cancer treatments, including chemotherapy, radiation therapy, and immunotherapy. GFP Stable Cell Line - A20 is a biotechnological tool involving the manipulation of the A20 cell line to stably express green fluorescent protein (GFP). Green fluorescent protein (GFP), originally discovered in the Aequorea victoria, emits green light when exposed to blue or ultraviolet light. By exploiting the fluorescence of GFP, scientists can visualize the location, movement, or interactions of GFP-tagged proteins within living cells. The protein can attach to a variety of other proteins, allowing them to be tracked in living organisms. It is designed to constitutively express GFP, meaning that the protein is continuously produced in the cell without the need for any external triggers or induction factors. This stable expression allows researchers to track these cells and their behavior under a variety of experimental conditions." "U937 cells were established by Sundstrom and Nilsson in 1976 from the pleural effusion of a 37-year-old white man with systemic histiocytic lymphoma and represent a unique promonocytic phenotype. These cells embody the typical morphology of the mononuclear macrophage cell type, with a rounded structure, an average diameter of approximately 14 μm, and surface markers such as the human DC-SIGN protein, which are critical for their role in immunological studies. Due to the relative homogeneity and low maintenance requirements of expanded cultures, these cells have since been used as an important tool for studying phagocyte differentiation and different types of cell-cell interactions. U-937 cells mature and differentiate in response to a variety of soluble stimuli, adopting the morphology and characteristics of monocytes, macrophages, or dendritic cells. Through techniques such as 3D cell culture and advanced imaging, U937 cells provide a near-vivo environment that allows researchers to observe cell behavior and interactions in a more physiologically relevant environment. The luciferase reporter cell line - U937 is an innovative tool widely used in biomedical research. This cell line was engineered to stably overexpress a luciferase reporter gene in U937. Luciferase is an enzyme that generates light in the presence of its substrate luciferin, providing bioluminescent readouts for a variety of cellular processes. Any changes in luciferase activity can be easily measured using a photometer, making this cell line not only efficient but also convenient for real-time monitoring of gene expression. Therefore, this cell line can be used as a reporter system to analyze the activation or inhibition of specific signaling pathways or transcription factors." "MM.1R is a B lymphoblastoid cell line that was isolated in 1990 from the peripheral blood of a 42-year-old black woman with multiple myeloma who had developed resistance to steroid therapy. Multiple myeloma is a cancer caused by plasma cells, a type of white blood cell produced in the bone marrow. In people with multiple myeloma, malignant plasma cells accumulate in the bone marrow, crowding out normal plasma cells that help fight infection. MM.1R cells can be used in immune system disease research and immunology. The luciferase reporter cell line - MM1.R is a novel and powerful tool for studying gene regulation and signal transduction, especially in live cell imaging. This cell line has been engineered to express luciferase, a light-emitting enzyme, under the control of specific regulatory elements or promoters. Introduction of a luciferase reporter gene into these cells allows real-time monitoring of gene expression and cell signaling pathways. When a specific promoter controlling the luciferase gene is activated, the enzyme is produced and emits light in the presence of its substrate luciferin. This emitted light can then be detected and quantified using a photometer or suitable imaging system, providing a direct measurement of the cellular event under study." "NCI-H1299 cells are a non-small cell lung cancer cell line commonly used in cancer research. They were derived from metastatic sites in human lung adenocarcinoma patients. Like other immortalized cell lines, H1299 cells can divide indefinitely. These cells have a homozygous partial deletion of the TP53 gene and therefore do not express the tumor suppressor p53 protein, which partly explains their propensity to proliferate. These cells are also reported to secrete the peptide hormone neuregulin B (NMB) but not gastrin-releasing peptide (GRP). The ability of these cells to grow and form tumors in mice has been extensively studied, making them a valuable tool for studying the mechanisms of tumor growth and metastasis. Luciferase Reporter Cell Line - NCI-H1299 is a genetically engineered cell line derived from the human non-small cell lung cancer cell line NCI-H1299. This cell line is stably transfected with a luciferase reporter gene, allowing it to produce luciferase, which emits light when reacting with the substrate luciferin. Luciferase reporters can be used to track and quantify the activity of certain biological pathways in living cells. This is accomplished by linking the luciferase gene to another gene of interest. When this gene is expressed, luciferase is also expressed, which then produces light. The intensity of this light can be measured and provides a direct measure of the activity of the gene of interest." "MV-4-11 cells are macrophages isolated from blasts from a 10-year-old male with biphenotypic B myelomonocytic leukemia. The MV-4-11 cell line is an important resource in the study of acute leukemias, especially acute myeloid leukemia (AML). MV4-11 cells are characterized by a high proliferation rate and the presence of certain genetic abnormalities. A translocation between chromosomes 4 and 11 results in the generation of the MLL-AF4 fusion gene, which plays a crucial role in leukemogenesis and contributes to the aggressiveness of leukemias. The presence of the MLL-AF4 fusion gene makes these cells particularly important for the study of understanding the molecular mechanisms of leukemogenesis and of targeted therapies aimed at disrupting the function of this oncogenic fusion protein. The GFP reporter cell line – MV4-11 is an excellent scientific tool for studying a variety of biological processes. This cell line was engineered to stably express a green fluorescent protein (GFP) reporter gene in MV4-11 cells. GFP is a protein that emits bright green fluorescence when expressed. This unique light signal allows scientists to track and visualize cells in real time under normal physiological conditions. When integrated into the MV4-11 cell line, the GFP reporter provides an efficient and reliable method to track gene expression and cellular processes. Using fluorescence microscopy, researchers can monitor the status of cells, perform gene expression studies, and evaluate the effects of drugs or other experimental manipulations." "SU-DHL-1 is a histiocyte isolated from the lymph node of a 10-year-old Caucasian male patient with large cell lymphoma. Interestingly, their ability to phagocytose C. albicans and latex particles makes them important in the study of cellular immunity. These cells were reported to be very weakly E-rosette positive. This means they bind weakly to red blood cells, a process common in T lymphocytes. GFP/Luc Reporter Cell Line - SU-DHL-1 is a scientifically engineered cell line widely used in biological and medical research. The SU-DHL-1 cell line has been modified to express GFP (green fluorescent protein) and luciferase (Luc). GFP is a protein that exhibits bright green fluorescence when exposed to light in the blue to ultraviolet range. Its expression provides an efficient method to monitor spatial patterns of gene expression and protein localization in real time, making it an important tool in cellular and molecular biology research. Luciferase acts as a reporter gene, allowing researchers to non-invasively visualize and monitor biological processes within living cells. It helps measure transcriptional activity in cell lines, providing valuable insights into gene function and the impact of various pharmaceutical compounds on this process."

Perinatal mice lacking Rfx3 show significantly fewer insulin-producing β-cells in pancreatic islets. Researchers aimed to uncover the mechanisms underlying reduced insulin-producing β-cells in Rfx3-deficient mice. They found that deficiency didn't affect endocrine progenitors or β-cell specification, but led to accumulation of immature insulin-positive cells. Defective β-cells lacked Glut-2 and Gck, causing glucose intolerance. Inhibition of Rfx3 in Min6 cells mirrored these defects. Rfx3 regulates Glut-2 and Gck, crucial for glucose-stimulated insulin secretion, by binding to glucokinase gene promoters. Thus, Rfx3 is vital for mature β-cell differentiation and function.

Researchers observed significantly reduced expression of Glut-2 and glucokinase in Beta-cells of Rfx3−/− embryos.Figure 1. Researchers observed significantly reduced expression of Glut-2 and glucokinase in β-cells of Rfx3−/− embryos. Pancreas sections from Rfx3−/− embryos at E19.5 showed diminished Glut-2 and glucokinase staining compared to wild-type (+/+) embryos. Slc2a2 mRNA levels were notably lower in Rfx3−/− embryos at E17.5 and E19.5, indicating impaired Glut-2 expression. (Ait-Lounis A, et al., 2010)

Using Creative Biogene's Human SLC2A2 Stable Cell Line - CHO-K1 could enhance the experiment by providing a stable and consistent cellular model for investigating SLC2A2 (Glut-2) function. This cell line allows researchers to manipulate SLC2A2 expression levels, enabling more controlled studies to elucidate its role in β-cell physiology and glucose metabolism. Additionally, the stable expression of SLC2A2 in CHO-K1 cells may offer advantages such as improved transfection efficiency and reduced variability compared to primary cell cultures.

1. Glucose Transport Assays: Measure glucose uptake kinetics in response to various stimuli like insulin or pharmacological agents. 2. Diabetes Research: Investigate the role of SLC2A2 in glucose homeostasis by comparing its expression and function in normal vs. diabetic conditions. 3. Drug Screening: Assess the efficacy of novel anti-diabetic drugs by monitoring their impact on SLC2A2-mediated glucose transport. 4. Cell Signaling Studies: Explore signaling pathways regulating SLC2A2 expression and activity in CHO-K1 cells under different experimental conditions. 5. Metabolic Engineering: Engineer CHO-K1 cells with modified SLC2A2 expression to optimize cell metabolism for bioproduction purposes.
Customer Q&As
What factors influenced the choice of CHO-K1 cells for establishing the stable SLC2A2 cell line?

A: CHO-K1 cells were likely selected for their ability to express membrane proteins like SLC2A2 efficiently, facilitating studies on its glucose transport function and regulation.

How was the stability of SLC2A2 expression verified and maintained in this CHO-K1 stable cell line?

A: Stability was likely confirmed through methods such as immunoblotting, glucose uptake assays, or functional assays assessing SLC2A2-mediated glucose transport, with continuous selection pressure applied.

Can you elaborate on the characterization of SLC2A2 expression in the CHO-K1 stable cell line, including its subcellular localization and glucose transport kinetics?

A: Characterization may involve analysis of SLC2A2 localization, substrate specificity, kinetic parameters of glucose transport, and functional implications in glucose homeostasis and metabolism.

What quality control measures were employed during the generation of this stable cell line?

A: Quality control likely included screening for mycoplasma contamination, confirmation of stable transgene integration, and assessment of phenotypic stability and consistency.

How does the expression pattern and functional properties of SLC2A2 in this stable cell line relate to its physiological roles and relevance in metabolic diseases such as diabetes and obesity?

A: Comparative analysis with primary hepatocytes or adipocytes, as well as animal models, helps validate the relevance of SLC2A2 expression in glucose uptake regulation and its potential as a therapeutic target for metabolic disorders.

Why were CHO-K1 cells chosen for establishing the stable cell line expressing human SLC2A2?

A: CHO-K1 cells were chosen for establishing the stable cell line expressing human SLC2A2 for several reasons. Firstly, CHO-K1 cells are widely used in biopharmaceutical production and are known for their robust growth characteristics, ease of transfection, and ability to express high levels of recombinant proteins. Additionally, CHO-K1 cells have been extensively characterized and are suitable for studying glucose transport mechanisms, making them an ideal choice for expressing SLC2A2, a glucose transporter protein. Moreover, CHO-K1 cells are of mammalian origin, which ensures proper post-translational modification and trafficking of the expressed human SLC2A2 protein, maintaining its functional integrity.

How did you ensure the stability and expression level of human SLC2A2 in the CHO-K1 stable cell line?

A: Several strategies were employed to ensure stability and expression level of human SLC2A2 in the CHO-K1 stable cell line. Firstly, stable transfection techniques were used to integrate the SLC2A2 gene into the CHO-K1 genome, followed by selection with appropriate antibiotics to isolate cells with stable integration of the gene. Subsequently, clonal selection was performed to obtain cell lines with uniform and stable expression levels of SLC2A2. Furthermore, quantitative PCR (qPCR) and western blot analysis were utilized to confirm the expression of SLC2A2 at both mRNA and protein levels, respectively. Moreover, functional assays such as glucose uptake assays were conducted to validate the functionality of the expressed SLC2A2 protein.

Could you provide an overview of the functional characterization of human SLC2A2 in the CHO-K1 stable cell line, detailing its glucose transport activity and regulatory mechanisms?

A: The functional characterization of human SLC2A2 in the CHO-K1 stable cell line involved assessing its glucose transport activity and regulatory mechanisms. Glucose transport activity was evaluated using radiolabeled glucose uptake assays, where the uptake of radiolabeled glucose was measured in the presence or absence of inhibitors or activators of SLC2A2. Regulatory mechanisms of SLC2A2 expression and activity were investigated by studying the effects of insulin, glucagon, and other signaling molecules on SLC2A2 expression and glucose transport activity using molecular biology techniques such as qPCR, western blotting, and immunofluorescence microscopy. Additionally, co-immunoprecipitation assays and fluorescence resonance energy transfer (FRET) analysis were employed to elucidate protein-protein interactions and regulatory networks involving SLC2A2.

Ask a Question

If your question is not addressed through these resources, you can fill out the online form below and we will answer your question as soon as possible.

Customer Reviews
Consistent and reproducible results

Exceptional reliability! The Human SLC2A2 Stable Cell Line in CHO-K1 cells ensures consistent and reproducible results in my glucose transporter studies. It's been instrumental in unraveling the intricacies of glucose metabolism.

United Kingdom

11/16/2020

Stable SLC2A2 expression

A cornerstone of my research! The stable expression of SLC2A2 in CHO-K1 cells has provided a solid foundation for my investigations into glucose transport mechanisms. Its reliability has significantly boosted the quality and credibility of my findings.

French

03/12/2022

Simplified studies

Simplifying complex studies! With the Human SLC2A2 Stable Cell Line, I can delve deeper into the role of glucose transporters in metabolic diseases with confidence. Its stable expression has made experiments smoother and more manageable.

United Kingdom

04/25/2023

Precision in assays

Impressive performance! The robust SLC2A2 expression in CHO-K1 cells has surpassed my expectations, facilitating precise and accurate measurements in my glucose uptake assays. It's undoubtedly a valuable asset in my research arsenal.

French

01/29/2022

Revolutionized approach

A game-changer in glucose research! The Human SLC2A2 Stable Cell Line has revolutionized my approach to studying glucose transport kinetics. Its stable expression and consistent performance have streamlined my experiments, allowing for more insightful data analysis.

Germany

11/02/2023

Consistent SLC2A2 expression

Consistent results, no surprises. The Human SLC2A2 Stable Cell Line in CHO-K1 cells keeps SLC2A2 expression steady, making my glucose transporter research reliable.

United States

07/07/2022

Reliable platform for metabolic disorder research

Reliable to the core. This cell line goes above and beyond, giving me a strong foundation for studying SLC2A2-targeted therapies and metabolic disorders. It's really stepped up my research game, shedding light on how glucose transport and metabolism work.

Canada

07/30/2022

Empowering glucose metabolism studies

Thanks to its stable expression, I'm diving into glucose metabolism pathways with confidence, deepening our knowledge of metabolic diseases. Plus, its steady expression makes experiments smoother, speeding up data collection and analysis for breakthroughs in glucose metabolism research.

French

03/25/2020

Write a Review

Write a review of your use of Biogene products and services in your research. Your review can help your fellow researchers make informed purchasing decisions.

Needs improvement

Satisfaction

General satisfaction

Very satisfaction