Human Cytomegalovirus Influences Host circRNA Transcriptions during Productive Infection

Human Cytomegalovirus Influences Host circRNA Transcriptions during Productive Infection

Objective: To investigate and compare the clinical features of cytomegalovirus (CMV) retinitis after solid organ transplant (SOT) and hematopoietic stem cell transplantation (HSCT) and to define poor prognostic factors.


Methods: Patients consult ophthalmology department for CMV viremia after transplantation between March 2008 and February 2018 and were followed for more than 6 months were analyzed. medical records regarding demographics, serology, and characteristics of the comparison between the SOT and HSCT. Factors associated with poor visual outcome was determined by logistic regression.


Results: CMV retinitis developed in 11.3% of patients with CMV viremia after transplantation. In the group SOT (25 points / 18 patients) and the HSCT group (33 eyes / 21 patients), CMV retinitis occurred in 5.8 months and 3.7 months post-transplantation, respectively. Mortality was significantly higher in the HSCT group (52.4% vs 5.6%, P <0.001). During an average of 11.7 months of follow-up, visual acuity tends to be exacerbated (P = 0.087) despite antiviral treatment, which is especially important in SOT group (P = 0.028). Six eyes (10.3%) due to retinal detachment underwent vitrectomy, which most (5 eyes) should be in the SOT. Multivariate logistic regression analysis showed that the concurrent presence of CMV disease (OR = 14.11, P = 0.009) and foveal involvement (OR = 114.85, P = 0.001) is a poor prognostic factor.


Conclusion: Clinical manifestations of CMV retinitis differ between groups HSCT and SOT. CMV disease simultaneously and foveal involvement is associated with poor visual outcome after transplantation CMV retinitis.


Human cytomegalovirus (HCMV) is a leading infectious agent causing birth defects. HCMV infection risk to the fetus in pregnant women who receive immunosuppressive agents is unknown. We describe two cases of pregnant women with pre-conception evidence of protective immunity HCMV receiving azathioprine for ulcerative colitis or systemic lupus erythematosus.

Both women reactivate HCMV infections and transmitted to the fetus. One newborn showed unilateral hearing deficit and brain disorders while others are asymptomatic. The mother of a newborn baby has symptoms of low levels of CD4 + T cells and HCMV-specific total blood. Women who receive immunosuppressive agents decent information about the risk of congenital HCMV infection and should be monitored for HCMV infection during pregnancy. their infants should be screened for congenital HCMV infection.

 Human Cytomegalovirus Influences Host circRNA Transcriptions during Productive Infection
Human Cytomegalovirus Influences Host circRNA Transcriptions during Productive Infection

The frequency of congenital cytomegalovirus infection in newborn babies in the State of Sao Paulo, 2010-2018

Human cytomegalovirus (HCMV) infection remains a neglected public health problem. The purpose of this study was to evaluate the frequency of congenital HCMV infection in newborns up to 1 month in Sao Paulo State, from 2010 to 2018.

Molecular characterization of HCMV-positive samples were also made. 275 urine samples from prospective patients congenital HCMV infection were tested by real-time Polymerase Chain Reaction (qPCR). HCMV-positive samples amplified by conventional PCR targeting UL89 gene, sorted and look for mutations.

STAT5 Reporter (Luc) - Ba/F3 Cell line

79772 2 vials
EUR 2275
Description: The STAT5 Reporter (Luc)-Ba/F3 cell line is designed for monitoring STAT5 signal transduction pathways. It contains a firefly luciferase gene driven by the STAT5 response element located upstream of the minimal TATA promoter. After activation by cytokines or growth factors, endogenous STAT5 binds to the DNA response elements, inducing transcription of the luciferase reporter gene.

IRF Reporter (Luc) - THP-1 Cell line

79858 2 vials
EUR 1810
Description: The Interferon Regulatory Factor (IRF) reporter (Luc)-THP-1 cell line is designed to study the activation and signaling of Cytosolic DNA Sensors (CDS) in human monocytic cell line THP-1. It contains a firefly luciferase gene driven by multimerized ISRE (Interferon Stimulated Response Element) located upstream of the minimal TATA promoter. _x000D_The cGAS-STING pathway acts to detect cytosolic DNA and induce an immune response. Briefly, upon binding DNA, the protein cGAS (cyclic GMP-AMP Synthase) triggers reaction of GTP and ATP to form cGAMP. cGAMP binds to STING (Stimulator of Interferon Genes) which triggers phosphorylation of IRF3 via TBK1. IRF3 can then bind to interferon-stimulated responsive elements (ISRE) in the nucleus and leads to IFN-α/β production. The IRF reporter (Luc)-THP-1 cell line is highly responsive to STING and CDS ligands.

NF-κB Reporter (Luc) - HCT116 Cell Line

60623 2 vials
EUR 1095
Description: NF-B luciferase reporter construct is stably integrated into the genome of HCT-116 cells. The
firefly luciferase gene is controlled by 4 copies of NF-κB response element located upstream of
the TATA promoter. Following activation by stimulants, endogenous NF-κB transcription factors
bind to the DNA response elements to induce transcription of the luciferase gene._x000D_The NF-κB-luciferase/HCT-116 cell line is suitable for monitoring the activity of NF-κB signaling
in response to stimulants such as the cytokines TNF and IL-1β, pathogen-associated
molecular pattern (PAMP) (i.e. flagellin) or endogenous damage-associated molecular pattern
(DAMP) molecules (i.e. NOD1 ligand) (see application references). It is also suitable for
establishing cell-based screens for inhibitors that target specific NF-κB stimulating molecules.
This cell line can be further modified to allow investigation of downstream NF-κB activities as a
result of targeted genetic mutation(s).

NF-κB reporter (Luc) - HEK293 Cell line

60650 2 vials
EUR 1365
Description: The NF-κB reporter (Luc) HEK293 cell line is designed to monitor nuclear factor Kappa B (NF-κB) activity. It contains a firefly luciferase gene driven by four copies of the NF-κB response element located upstream of the minimal TATA promoter. After activation by pro-inflammatory cytokines or agonists of lymphokine receptors, endogenous NF-κB transcription factors bind to the DNA response elements, inducing transcription of the luciferase reporter gene. The cell line has been functionally validated in response to human TNF-α, IL-1β, and IL-17.

NF-kB reporter (Luc) - HEK293 Cell line

GWB-PS76F8 2X10(6)cells Ask for price

NF- κB Reporter (Luc) - Raw 264.7 Cell line

79978 2 vials
EUR 2045
Description: The NF-κB reporter (Luc)-Raw 264.7 cell line is designed for monitoring nuclear factor Kappa B (NF-κB) signal transduction pathways. It contains a firefly luciferase gene driven by four copies of the NF-κB response element located upstream of the minimal TATA promoter. After activation by pro-inflammatory cytokines or stimulants of lymphokine receptors, endogenous NF-κB transcription factors bind to the DNA response elements, inducing transcription of the luciferase reporter gene.

NF- κB Reporter (Luc) - THP-1 Cell Line

79645 2 vials
EUR 1900
Description: The NF-κB reporter (Luc)-THP-1 cell line is designed for monitoring nuclear factor Kappa B (NF-κB) signal transduction pathways. It contains a firefly luciferase gene driven by four copies of the NF-κB response element located upstream of the minimal TATA promoter. After activation by pro-inflammatory cytokines or stimulants of lymphokine receptors, endogenous NF-κB transcription factors bind to the DNA response elements, inducing transcription of the luciferase reporter gene.

PAI-1 Reporter (Luc) - Mv1 Lu Cell Line

60544 2 vials
EUR 3595
Description: PAI-1 Reporter (Luc)-Mv1 Lu cell line is designed for monitoring transforming growth factor β (TGF-β)-induced plasminogen activator inhibitor-1 (PAI-1) expression. Transforming growth factor-β (TGF-β) is a potent regulator of cellular differentiation, proliferation, migration, and protein expression._x000D__x000D_PAI-1 Reporter (Luc) -Mv1 Lu cell line contains a firefly luciferase gene under the control of PAI-1 responsive elements stably integrated into Mv1 Lu (NBL-7) cells, showing TGF-β pathway response. This cell line is validated for the TGF-β response to the induction of PAI-1 gene expression through luciferase activity. _x000D_

Bald Lentiviral Pseudovirion (Luc-eGFP Dual Reporter)

79988 500 µl x 2
EUR 795
Description: The bald lentiviral pseudovirion was produced without envelope glycoproteins such as VSV-G or SARS-CoV-2 spike. It contains a firefly luciferase and eGFP cassette (Luc-P2A-eGFP) as the reporters, driven by a CMV promoter. The bald lentiviral pseudovirion can serve as a negative control when studying virus entry initiated by specific interactions between virus particles and receptors._x000D_

NF-κB reporter (Luc) - NIH/3T3 Cell line

79469 2 vials
EUR 1900
Description: The NF-κB reporter (Luc)-NIH/3T3 cell line is designed for monitoring nuclear factor Kappa B (NF-κB) signal transduction pathways. It contains a firefly luciferase gene driven by four copies of the NF-κB response element located upstream of the minimal TATA promoter. After activation by pro-inflammatory cytokines or stimulants of lymphokine receptors, endogenous NF-κB transcription factors bind to the DNA response elements, inducing transcription of the luciferase reporter gene.

NF-κB Reporter (Luc) - CHO-K1 Cell Line

60622 2 vials
EUR 1095
Description: An NF-κB luciferase reporter construct is stably integrated into the genome of CHO-K1 cells. The firefly luciferase gene is controlled by the NF-κB response element located upstream of the TATA promoter. Following activation by stimulants, endogenous NF-κB transcription factors bind to the DNA response elements to induce transcription of the luciferase gene._x000D_The NF-κB-luciferase / CHO-K1 cell line is suitable for monitoring the activity of NF-κB transcription factor through luminescence readout.). This cell line responds to human cytokine IL-1β, responds moderately to human TNF, and does not respond to human IFN-λ (2 µg/ml). Reducing the amount of serum during incubation period may increase the sensitivity to cytokines. Since CHO-K1 cells do not express endogenous human proteins, this cell line provides an excellent platform to enable exogenous expression of a protein of interest to study its downstream effect on NF-κB signaling.

STAT3 Reporter (Luc) - HEK293 Cell line (Puromycin)

79800-P 2 vials
EUR 3730
Description: The STAT3 Reporter (Luc)-HEK293 cell line is designed for monitoring STAT3 signal transduction pathway. It contains a firefly luciferase gene driven by STAT3 response elements located upstream of the minimal TATA promoter. After activation by cytokines and growth factors, endogenous STAT3 binds to the DNA response elements, inducing transcription of the luciferase reporter gene.

Foxp3 Reporter (Luc) - Jurkat Recombinant Cell Line

60628 2 vials
EUR 7645
Description: Human Foxp3 luciferase reporter construct is stably integrated into the genome of Jurkat T- cells. The firefly luciferase gene is controlled by a human Foxp3 promoter and an enhancer-like conserved noncoding sequence upstream of the Foxp3 promoter.

NF-κB Reporter (Luc) - A549 Stable Cell Line

60625 2 vials
EUR 1915
Description: NF-κB luciferase reporter construct is stably integrated into the genome of A549 cells. The firefly luciferase gene is controlled by 4 copies of NF-κB response element located upstream of the TATA promoter. Following activation by stimulants, endogenous NF-κB transcription factors bind to the DNA response elements to induce transcription of the luciferase gene.

STAT5 Reporter (Luc)- U937 Cell Line (GM-CSF)

79941 2 vials
EUR 1980
Description: The STAT5 Reporter (Luc)-U937 cell line is designed for monitoring STAT5 signal transduction pathway in the U937 cell line. It contains a firefly luciferase gene driven by the STAT5 response element located upstream of the minimal TATA promoter. After activation by GM-CSF, endogenous STAT5 binds to the DNA response elements, inducing transcription of the luciferase reporter gene.

Rev-A3-GFP/Luc HIV Reporter Cells

HRC-3 1 vial of 5x10⁶ cells
EUR 1800

Rev-CEM-GFP/Luc HIV Reporter Cells

HRC-5 1 vial of 5x10⁶ cells
EUR 1500

Spike (B.1.429 Variant) Pseudotyped Lentivirus (Luc Reporter)

78172-1 100 µl
EUR 835
Description: The Spike (B.1.429 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.429 Variant Spike (Genbank Accession #QHD43416.1 with B.1.429 variant mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.429 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.429 variant in a Biosafety Level 2 facility.Spike Mutations in B.1.429 Variant: S13I
W152C
L452R
D614G

Spike (B.1.429 Variant) Pseudotyped Lentivirus (Luc Reporter)

78172-2 500 µl x 2
EUR 4195
Description: The Spike (B.1.429 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.429 Variant Spike (Genbank Accession #QHD43416.1 with B.1.429 variant mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.429 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.429 variant in a Biosafety Level 2 facility.Spike Mutations in B.1.429 Variant: S13I
W152C
L452R
D614G

Spike (B.1.617 Variant) Pseudotyped Lentivirus (Luc Reporter)

78204-1 100 µl
EUR 835
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617 (Kappa, Delta lineage) was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.617 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.617 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617 variant in a Biosafety Level 2 facility. 

Spike (B.1.617 Variant) Pseudotyped Lentivirus (Luc Reporter)

78204-2 500 µl x 2
EUR 4195
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617 (Kappa, Delta lineage) was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.617 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.617 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617 variant in a Biosafety Level 2 facility.

Spike (B.1.617.1 Variant) Pseudotyped Lentivirus (Luc Reporter)

78205-1 100 µl
EUR 835
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.1 (also known as the Kappa Variant) was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.617.1 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.617.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.1 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.1 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.1 variant in a Biosafety Level 2 facility. Spike Mutations in B.1.617.1 Variant:G142DE154KL452RE484QD614GP681RQ1071H

Spike (B.1.617.1 Variant) Pseudotyped Lentivirus (Luc Reporter)

78205-2 500 µl x 2
EUR 4195
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.1 (also known as the Kappa Variant) was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.617.1 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.617.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.1 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.1 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.1 variant in a Biosafety Level 2 facility. Spike Mutations in B.1.617.1 Variant:G142DE154KL452RE484QD614GP681RQ1071H

Spike (B.1.618 Variant) Pseudotyped Lentivirus (Luc Reporter)

78206-1 100 µl
EUR 835
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.618 was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.618 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.618 Variant Spike (Genbank Accession #QHD43416.1 with B.1.618 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.618 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.618 variant in a Biosafety Level 2 facility. Spike Mutations in B.1.618 Variant:Y145delH146delE484KD614G

Spike (B.1.618 Variant) Pseudotyped Lentivirus (Luc Reporter)

78206-2 500 µl x 2
EUR 4195
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.618 was identified in India in the spring of 2021. This variant has a number of mutations that allow the virus to spread more easily and quickly than other variants. The Spike (B.1.618 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.618 Variant Spike (Genbank Accession #QHD43416.1 with B.1.618 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.618 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.618 variant in a Biosafety Level 2 facility. Spike Mutations in B.1.618 Variant:Y145delH146delE484KD614G

Spike (B.1.617.2 Variant) Pseudotyped Lentivirus (Luc Reporter)

78215-1 100 µl
EUR 900
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.2 (also known as the Delta Variant) was identified in India in the spring of 2021. This variant has a number of mutations that increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.617.2 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.617.2 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.2 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.2 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.2 variant in a Biosafety Level 2 facility.

Spike (B.1.617.2 Variant) Pseudotyped Lentivirus (Luc Reporter)

78215-2 500 µl x 2
EUR 4510
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.2 (also known as the Delta Variant) was identified in India in the spring of 2021. This variant has a number of mutations that increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.617.2 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.617.2 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.2 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.2 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.2 variant in a Biosafety Level 2 facility.

Human 5-HT1A (Luc) HEK293 Reporter Cell

CHEK-ATF131 2Vials
EUR 14209.6
Description: This gene encodes a G protein-coupled receptor for 5-hydroxytryptamine (serotonin), and belongs to the 5-hydroxytryptamine receptor subfamily. Serotonin has been implicated in a number of physiologic processes and pathologic conditions. Inactivation of this gene in mice results in behavior consistent with an increased anxiety and stress response. Mutation in the promoter of this gene has been associated with menstrual cycle-dependent periodic fevers.

Rev-A3R5-GFP/Luc HIV Reporter Cells

HRC-2 1 vial of 5x10⁶ cells
EUR 1900

Spike (SARS-CoV-1) Pseudotyped Lentivirus (Luc Reporter)

78614-1 100 µl
EUR 860
Description: Severe acute respiratory syndrome (SARS) was the first new infectious disease identified in the twenty-first century. It is a viral respiratory disease caused by severe acute respiratory syndrome coronavirus (SARS-CoV-1). The first known cases occurred in November 2002, and the syndrome caused the 2002-2004 SARS outbreak. Since 2004, no cases of SARS-CoV-1 have been reported worldwide. A virus very similar to SARS-CoV-1 was discovered in late 2019. This virus, named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the causative pathogen of COVID-19, the spread of which started the COVID-19 pandemic.SARS-CoV-1 attaches to the host cell surface before entering the cell. The Spike protein on the virus recognizes and binds to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of human airway epithelia as well as lung parenchyma. Drugs targeting the interaction between the Spike protein of SARS-CoV-1 and ACE2 may offer protection against the viral infection.The Spike (SARS-CoV-1) Pseudotyped Lentiviruses were produced with SARS-CoV-1 Spike (Genbank Accession #YP_009825051.1) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-1) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-1 in a cellular context, using a Biosafety Level 2 facility.The Spike (SARS-CoV-1) Pseudotyped Lentiviruses has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).

Spike (SARS-CoV-1) Pseudotyped Lentivirus (Luc Reporter)

78614-2 500 µl x 2
EUR 4320
Description: Severe acute respiratory syndrome (SARS) was the first new infectious disease identified in the twenty-first century. It is a viral respiratory disease caused by severe acute respiratory syndrome coronavirus (SARS-CoV-1). The first known cases occurred in November 2002, and the syndrome caused the 2002-2004 SARS outbreak. Since 2004, no cases of SARS-CoV-1 have been reported worldwide. A virus very similar to SARS-CoV-1 was discovered in late 2019. This virus, named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the causative pathogen of COVID-19, the spread of which started the COVID-19 pandemic.SARS-CoV-1 attaches to the host cell surface before entering the cell. The Spike protein on the virus recognizes and binds to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of human airway epithelia as well as lung parenchyma. Drugs targeting the interaction between the Spike protein of SARS-CoV-1 and ACE2 may offer protection against the viral infection.The Spike (SARS-CoV-1) Pseudotyped Lentiviruses were produced with SARS-CoV-1 Spike (Genbank Accession #YP_009825051.1) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-1) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-1 in a cellular context, using a Biosafety Level 2 facility.The Spike (SARS-CoV-1) Pseudotyped Lentiviruses has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).

Oct4 CR4-pGreenFire Response Reporter (virus)

SR20070-VA-1 >2 x 10^6 IFUs
EUR 670

A549-Dual NFkb-SEAP-IRF-Luc Reporter Cells

S0016001 One Frozen vial
EUR 1400

NF-κB-Luciferase Reporter (Luc) - Jurkat Cell Line

60651 2 vials
EUR 2340
Description: NF-κB luciferase reporter construct is stably integrated into the genome of Jurkat T- cells. The firefly luciferase gene is controlled by 4 copies of NF-kB response element located upstream of the TATA promoter. Following activation by stimulants, endogenous NF-κB transcription factors bind to the DNA response elements to induce transcription of the luciferase gene.

Rev-CEM-Luc HIV Reporter Cells

HRC-6 1 vial of 5x10⁶ cells
EUR 1500

Rat NSE Differentiation Reporter (pGreenZeo, Virus)

SR10024VA-1 >2 x 10^6 IFUs
EUR 691

Human GFAP Differentiation Reporter (pRedZeo, Virus)

SR10051VA-1 >2 x 10^6 IFUs
EUR 691

CD40/NF-κB Reporter (Luc) - HEK293 Stable Cell Line

60626 2 vials
EUR 6825
Description: Recombinant HEK293 cell line expressing full length human CD40 (Tumor necrosis factor receptor superfamily member 5; TNFRSF5). Expression is confirmed by real-time qPCR and Western Blot. This NF-κB luciferase reporter construct is stably integrated into the genome. The firefly luciferase gene is controlled by 4 copies of NF-κB response element located upstream of the TATA promoter. Following activation by human CD40 ligand, NF-κB transcription factor binds to the DNA response elements to induce transcription of the luciferase gene. _x000D_

Mouse MBP Differentiation Reporter (pGreenZeo, Virus)

SR10026VA-1 >2 x 10^6 IFUs
EUR 691

Human LCK Differentiation Reporter (pGreenZeo, Virus)

SR10032VA-1 >2 x 10^6 IFUs
EUR 691

Human B29 Differentiation Reporter (pGreenZeo, Virus)

SR1004VA-1 >2 x 10^6 IFUs
EUR 691

Mouse B29 Differentiation Reporter (pGreenZeo, Virus)

SR1005VA-1 >2 x 10^6 IFUs
EUR 691

Mouse CD8 Differentiation Reporter (pGreenZeo, Virus)

SR1006VA-1 >2 x 10^6 IFUs
EUR 691

Human CD2 Differentiation Reporter (pGreenZeo, Virus)

SR1009VA-1 >2 x 10^6 IFUs
EUR 691

Mouse Actc Differentiation Reporter (pGreenZeo, Virus)

SR10010VA-1 >2 x 10^6 IFUs
EUR 691

Human GFAP Differentiation Reporter (pGreenZeo, Virus)

SR10015VA-1 >2 x 10^6 IFUs
EUR 691

Mouse GFAP Differentiation Reporter (pGreenZeo, Virus)

SR10016VA-1 >2 x 10^6 IFUs
EUR 691

Mouse EMR1 Differentiation Reporter (pGreenZeo, Virus)

SR10018VA-1 >2 x 10^6 IFUs
EUR 691

Mouse CD44 Differentiation Reporter (pGreenZeo, Virus)

SR10020VA-1 >2 x 10^6 IFUs
EUR 691

Human BM88 Differentiation Reporter (pGreenZeo, Virus)

SR10021VA-1 >2 x 10^6 IFUs
EUR 691

Rat Nestin Differentiation Reporter (pGreenZeo, Virus)

SR10034VA-1 >2 x 10^6 IFUs
EUR 691

Mouse ALBP Differentiation Reporter (pGreenZeo, Virus)

SR10036VA-1 >2 x 10^6 IFUs
EUR 691

Human NGN3 Differentiation Reporter (pGreenZeo, Virus)

SR10037VA-1 >2 x 10^6 IFUs
EUR 691

Human PDX1 Differentiation Reporter (pGreenZeo, Virus)

SR10039VA-1 >2 x 10^6 IFUs
EUR 691

Mouse PDX1 Differentiation Reporter (pGreenZeo, Virus)

SR10040VA-1 >2 x 10^6 IFUs
EUR 691

Human MAP2 Differentiation Reporter (pGreenZeo, Virus)

SR10047VA-1 >2 x 10^6 IFUs
EUR 691

Human ACTC Differentiation Reporter (pGreenZeo, Virus)

SR10049VA-1 >2 x 10^6 IFUs
EUR 691

Human NKX2.5 Differentiation Reporter (pGreenZeo, virus)

SR10067VA-1 >2 x 10^6 IFUs
EUR 691

Mouse CD68 Differentiation Reporter (pGreenZeo, Virus)

SR1008VA-1 >2 x 10^6 IFUs
EUR 691

Spike (SARS-CoV-2, D614G) Pseudotyped Lentivirus (Luc Reporter)

78028-1 100 µl
EUR 900
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein and ACE2 may offer protection against the viral infection. A SARS-CoV-2 variant carrying the spike protein amino acid change D614G has become the most prevalent form in the global pandemic.
The SARS-CoV-2 Spike D614G Pseudotyped Lentivirus were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1; with D614G mutation) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The SARS-CoV-2 Spike D614G pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 in a Biosafety Level 2 facility._x000D_

Spike (SARS-CoV-2, D614G) Pseudotyped Lentivirus (Luc Reporter)

78028-2 500 µl x 2
EUR 4510
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein and ACE2 may offer protection against the viral infection. A SARS-CoV-2 variant carrying the spike protein amino acid change D614G has become the most prevalent form in the global pandemic.
The SARS-CoV-2 Spike D614G Pseudotyped Lentivirus were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1; with D614G mutation) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The SARS-CoV-2 Spike D614G pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 in a Biosafety Level 2 facility._x000D_

Human Tnnt2 Differentiation Reporter (pGreenZeo, Virus)

SR10012VA-1 >2 x 10^6 IFUs
EUR 691

Mouse Tnnt2 Differentiation Reporter (pGreenZeo, Virus)

SR10013VA-1 >2 x 10^6 IFUs
EUR 691

Mouse SM22a Differentiation Reporter (pGreenZeo, Virus)

SR10014VA-1 >2 x 10^6 IFUs
EUR 691

Human CD11b Differentiation Reporter (pGreenZeo, Virus)

SR10017VA-1 >2 x 10^6 IFUs
EUR 691

Mouse GAD67 Differentiation Reporter (pGreenZeo, Virus)

SR10023VA-1 >2 x 10^6 IFUs
EUR 691

Human Opsin Differentiation Reporter (pGreenZeo, Virus)

SR10027VA-1 >2 x 10^6 IFUs
EUR 691

Human FABP7 Differentiation Reporter (pGreenZeo, Virus)

SR10048VA-1 >2 x 10^6 IFUs
EUR 691

Spike (B.1.1.529, Omicron Variant) Pseudotyped Lentivirus (Luc Reporter)

78348-1 100 µl
EUR 900
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.1.529 (also known as the Omicron Variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants.The Spike (B.1.1.529 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.1.529 Variant Spike (Genbank Accession #QHD43416.1 with B.1.1.529 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.1.529 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.1.529 variant in a Biosafety Level 2 facility.The Spike Omicron pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience, #79951).

Spike (B.1.1.529, Omicron Variant) Pseudotyped Lentivirus (Luc Reporter)

78348-2 500 µl x 2
EUR 4510
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.1.529 (also known as the Omicron Variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants.The Spike (B.1.1.529 Variant) (SARS-CoV-2) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.1.529 Variant Spike (Genbank Accession #QHD43416.1 with B.1.1.529 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.1.529 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.1.529 variant in a Biosafety Level 2 facility.The Spike Omicron pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience, #79951).

AADACL4 3'UTR Luciferase Stable Cell Line

TU000018 1.0 ml
EUR 1672.8

Mouse Col2a1 Differentiation Reporter (pGreenZeo, Virus)

SR1001VA-1 >2 x 10^6 IFUs
EUR 691

Mouse Camk2a Differentiation Reporter (pGreenZeo, Virus)

SR10022VA-1 >2 x 10^6 IFUs
EUR 691

Human Nestin Differentiation Reporter (pGreenZeo, Virus)

SR10035VA-1 >2 x 10^6 IFUs
EUR 691

Myc Reporter (Luc) - HCT116 Cell Line (Myc Signaling Pathway)

60520 2 vials
EUR 2175
Description: The Myc Reporter - HCT116 cell line contains the firefly luciferase gene under the control of Myc responsive elements stably integrated into HCT116 cells, a human colon cancer cell line. HCT116 contains a mutated beta-catenin which leads to the accumulation of β-catenin and constitutive activation of downstream Myc that induces the expression of Myc luciferase reporter. The cell line is validated for the inhibition of the expression of Myc luciferase reporter.

GITR / NF-κB-Luciferase Reporter (Luc) - Jurkat Cell Line

60546 2 vials
EUR 10175
Description: This cell line expresses a surface human GITR (glucocorticoid-induced TNFR family related gene; TNFRSF18; CD357) and an NF-κB luciferase reporter construct that are stably integrated into the genome of Jurkat T-cells. The firefly luciferase gene is controlled by 4 copies of NF-κB response element located upstream of the TATA promoter. Following activation by stimulants, endogenous NF-κB transcription factors bind to the DNA response elements to induce transcription of the luciferase gene. The cells have been validated using purified human GITRL and anti-GITR neutralizing antibody.

Human Insulin Differentiation Reporter (pGreenZeo, Virus)

SR10028VA-1 >2 x 10^6 IFUs
EUR 691

Mouse IBA-1 Differentiation Reporter (pGreenZeo, Virus)

SR10019VA-1 >2 x 10^6 IFUs
EUR 691

Human SPP-1 Differentiation Reporter (pGreenZeo, Virus)

SR1002VA-1 >2 x 10^6 IFUs
EUR 691

Mouse Myogenin Differentiation Reporter (pGreenZeo, Virus)

SR10050VA-1 >2 x 10^6 IFUs
EUR 691

Human MLC-2v Differentiation Reporter (pGreenZeo, Virus)

SR10011VA-1 >2 x 10^6 IFUs
EUR 691

GAS Reporter (Luc) - HeLa Cell Line (IFNγ/JAK/STAT1 Pathway)

79041 2 vials
EUR 1810
Description: The GAS reporter (Luc)-HeLa cell line is designed to monitor the activity of interferon gamma-induced signal transduction pathways in cultured cells by measuring activated STAT1 homodimers. It contains a firefly luciferase gene driven by three copies of the interferon gamma-activated sites (GAS) located upstream of the minimal TATA promoter. IFNγ first binds to a heterodimeric receptor consisting of two chains, IFNGR1 and IFNGR2, causing its dimerization and the activation of specific Janus family kinases (JAK1 and JAK2). Two STAT1 molecules associate with this ligand-activated receptor complex and are activated by phosphorylation to form active homodimer. The active STAT1 homodimers translocate to the nucleus where they bind interferon gamma-activated sites (GAS) in the promoter of IFNγ inducible genes, including luciferase reporter gene.

Lentiviral Dual Reporter: CMV-GFP-T2A-Luciferase pre-packaged virus

BLIV101VA-1 >2 x10^6 IFUs
EUR 722

Lentiviral Dual Reporter: UBC-RFP-T2A-Luciferase pre-packaged virus

BLIV200VA-1 >2 x10^6 IFUs
EUR 722

Human GFAP Differentiation Reporter (pGreenZeo, Virus) Puro

SR10015VA-P >2 x 10^6 IFUs
EUR 691

Human HLA-DRa Differentiation Reporter (pGreenZeo, Virus)

SR1007VA-1 >2 x 10^6 IFUs
EUR 691

Human Osteocalcin Differentiation Reporter (pGreenZeo, Virus)

SR1003VA-1 >2 x 10^6 IFUs
EUR 691

Spike (B.1.617.2.1; Delta Plus Variant) Pseudotyped Lentivirus (Luc Reporter)

78218-1 100 µl
EUR 835
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.2.1 (also known as the Delta Plus Variant) was identified in India in the spring of 2021. This variant has a number of mutations that increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.617.2.1 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.617.2.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.2.1 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G.  Compared to the Delta variant (B.1.617.2), variant Delta Plus has an additional mutation, K417N. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.2.1 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.2.1 variant in a Biosafety Level 2 facility.

Spike (B.1.617.2.1; Delta Plus Variant) Pseudotyped Lentivirus (Luc Reporter)

78218-2 500 µl x 2
EUR 4195
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.617.2.1 (also known as the Delta Plus Variant) was identified in India in the spring of 2021. This variant has a number of mutations that increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.617.2.1 Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.617.2.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.617.2.1 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G.  Compared to the Delta variant (B.1.617.2), variant Delta Plus has an additional mutation, K417N. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.617.2.1 Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.617.2.1 variant in a Biosafety Level 2 facility.

Human Keratin 14 Differentiation Reporter (pGreenZeo, Virus)

SR10038VA-1 >2 x 10^6 IFUs
EUR 691

Spike (SARS-CoV-2, UK Variant) Pseudotyped Lentivirus (Luc Reporter)

78112-1 100 µl
EUR 875
Description: The Spike (SARS-CoV-2, UK variant) Pseudotyped Lentivirus were produced with SARS-CoV-2 UK Variant Spike (Genbank Accession #QHD43416.1 with UK variant mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2, UK variant) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 UK variant in a Biosafety Level 2 facility._x000D_

Spike (SARS-CoV-2, UK Variant) Pseudotyped Lentivirus (Luc Reporter)

78112-2 500 µl x 2
EUR 4405
Description: The Spike (SARS-CoV-2, UK variant) Pseudotyped Lentivirus were produced with SARS-CoV-2 UK Variant Spike (Genbank Accession #QHD43416.1 with UK variant mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2, UK variant) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 UK variant in a Biosafety Level 2 facility._x000D_

Lentiviral Triple Reporter: CMV-Luciferase-RFP-TK pre-packaged virus

BLIV102VA-1 >2 x10^6 IFUs
EUR 722

Lentiviral Triple Reporter: UBC-Luciferase-RFP-TK pre-packaged virus

BLIV202VA-1 >2 x10^6 IFUs
EUR 722

Lentiviral Triple Reporter: MSCV-Luciferase-RFP-TK pre-packaged virus

BLIV302VA-1 >2 x10^6 IFUs
EUR 722

Spike(SARS-CoV-2) Pseudotyped Lentivirus (Luc-eGFP Dual Reporter)

79982-1 100 µl
EUR 1075
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection._x000D_
The SARS-CoV-2 Spike Pseudotyped Lentivirus (Luc-eGFP dual reporter) were produced by replacing the VSV-G fusion glycoprotein with SARS-CoV-2 Spike protein (Genbank Accession #QHD43416.1) as a surrogate viral envelope protein. These pseudovirions also contain a firefly luciferase and eGFP cassette (Luc-P2A-eGFP) driven by a CMV promoter. The luciferase and eGFP are coexpressed under the CMV promoter in the transduced cells. Therefore, the Spike-mediated entry into the target cell can be conveniently measured via luciferase reporter activity or eGFP expression. The SARS-CoV-2 Spike pseudotyped lentivirus can be used in a cellular assay to measure the activity of neutralizing antibody against SARS-CoV-2._x000D_

Spike(SARS-CoV-2) Pseudotyped Lentivirus (Luc-eGFP Dual Reporter)

79982-2 500 µl x 2
EUR 8110
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection._x000D_
The SARS-CoV-2 Spike Pseudotyped Lentivirus (Luc-eGFP dual reporter) were produced by replacing the VSV-G fusion glycoprotein with SARS-CoV-2 Spike protein (Genbank Accession #QHD43416.1) as a surrogate viral envelope protein. These pseudovirions also contain a firefly luciferase and eGFP cassette (Luc-P2A-eGFP) driven by a CMV promoter. The luciferase and eGFP are coexpressed under the CMV promoter in the transduced cells. Therefore, the Spike-mediated entry into the target cell can be conveniently measured via luciferase reporter activity or eGFP expression. The SARS-CoV-2 Spike pseudotyped lentivirus can be used in a cellular assay to measure the activity of neutralizing antibody against SARS-CoV-2._x000D_

AADACL4 3'UTR GFP Stable Cell Line

TU050018 1.0 ml
EUR 1672.8

Sox2 SRR2-pGreenFire Response Reporter, pre-packaged virus

SR20071-VA-1 >2 x 10^6 IFUs
EUR 670

GLP-1R/CRE (Luc) Reporter - HEK293 Recombinant Cell Line

78176 2 vials
EUR 10105
Description: Recombinant HEK293 cells expressing firefly luciferase gene under the control of cAMP response element (CRE) with constitutive expression of human GLP-1R (Glucagon-like peptide 1 receptor; accession number BC113493)._x000D_GLP-1R, a member of the class B family of G protein-coupled receptors (GPCRs) primarily found in pancreatic β cells, is activated by a peptide hormone, glucagon-like peptide 1 (GLP-1) that is secreted from intestinal L-cells after nutrient ingestion. GLP-1R plays an important role in controlling blood sugar level by enhancing glucose-stimulated insulin secretion, so various research efforts have focused on the regulation of the GLP-1R mediated signaling pathway as a therapeutic approach to diabetes.

Mouse Alpha-Tubulin Differentiation Reporter (pGreenZeo, Virus)

SR10025VA-1 >2 x 10^6 IFUs
EUR 691

Human Doublecortin (DCX) Differentiation Reporter (pGreenZeo, Virus)

SR10041VA-1 >2 x 10^6 IFUs
EUR 691

Spike (B.1.351 Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78142-1 100 µl
EUR 860
Description: The Spike (SARS-CoV-2) (B.1.351) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.351 Variant Spike (Genbank Accession #QHD43416.1 with B.1.351 mutations (L18F, D80A, D215G, R246I, K417N, E484K, N501Y, D614G, A701V) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2) (B.1.351) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.351 variant in a Biosafety Level 2 facility._x000D_

Spike (B.1.351 Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78142-2 500 µl x 2
EUR 4320
Description: The Spike (SARS-CoV-2) (B.1.351) Pseudotyped Lentivirus were produced with SARS-CoV-2 B.1.351 Variant Spike (Genbank Accession #QHD43416.1 with B.1.351 mutations (L18F, D80A, D215G, R246I, K417N, E484K, N501Y, D614G, A701V) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2) (B.1.351) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 B.1.351 variant in a Biosafety Level 2 facility._x000D_

Spike (P.1 Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78144-1 100 µl
EUR 835
Description: In Brazil, a variant called P.1 was first identified in the summer of 2020. This variant has many mutations that may lead to higher transmissibility and infectivity. The Spike (P.1) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Variant Spike (Genbank #QHD43416.1 with P.1 mutations (L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, D614G, H655Y, T1027I) as the envelope glycoproteins instead of the commonly used VSVG. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (P.1) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 (P.1) variant using a Biosafety Level 2 facility._x000D_

Spike (P.1 Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78144-2 500 µl x 2
EUR 4195
Description: In Brazil, a variant called P.1 was first identified in the summer of 2020. This variant has many mutations that may lead to higher transmissibility and infectivity. The Spike (P.1) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Variant Spike (Genbank #QHD43416.1 with P.1 mutations (L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, D614G, H655Y, T1027I) as the envelope glycoproteins instead of the commonly used VSVG. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (P.1) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 (P.1) variant using a Biosafety Level 2 facility._x000D_

Human E-Cadherin, CDH1 Differentiation Reporter (pGreenZeo, virus)

SR10070VA-1 >2 x 10^6 IFUs
EUR 691

Spike (K417T, E484K, N501Y) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78143-1 100 µl
EUR 835
Description: The Spike (K417T, E484K, N501Y) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Variant Spike (Genbank Accession #QHD43416.1 with mutations K417T, E484K, and N501Y) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2, K417T, E484K, N501Y) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 K417T, E484K, N501Y variant in intact cells using a Biosafety Level 2 facility._x000D_

Spike (K417T, E484K, N501Y) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78143-2 500 µl x 2
EUR 4195
Description: The Spike (K417T, E484K, N501Y) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Variant Spike (Genbank Accession #QHD43416.1 with mutations K417T, E484K, and N501Y) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (SARS-CoV-2, K417T, E484K, N501Y) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 K417T, E484K, N501Y variant in intact cells using a Biosafety Level 2 facility._x000D_

CRE/CREB Reporter (Luc) - HEK293 Cell Line (cAMP/PKA Signaling Pathway)

60515 2 vials
EUR 2070
Description: The cAMP/PKA Signaling Pathway CRE/CREB Reporter (Luc) - HEK293 Cell Line is designed for monitoring the activity of the cAMP/ PKA signaling pathway. The cAMP/PKA Signaling Pathway CRE/CREB Reporter (Luc) - HEK293 Cell Line contains a firefly luciferase gene under the control of multimerized cAMP response element (CRE) stably integrated into HEK293 cells. Elevation of the intracellular cAMP level activates cAMP response element binding protein (CREB) to bind CRE and induces the expression of luciferase.

CRE/CREB Reporter (Luc) - Jurkat Cell Line (cAMP/PKA Signaling Pathway)

79636 2 vials
EUR 1810
Description: The CRE/CREB Reporter (Luc) - Jurkat Cell Line contains a firefly luciferase gene under the control of multimerized cAMP response element (CRE) stably integrated into Jurkat cells. Elevation of the intracellular cAMP level activates cAMP response element binding protein (CREB) to bind CRE and induces the expression of luciferase. This cell line is validated for response to stimulation by Forskolin.

Spike (B.1.621, Mu Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78618-1 100 µl
EUR 795
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.621 (also known as the Mu Variant) was first identified in Columbia in early 2021. This variant has a number of mutations that may increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.621, Mu Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.621 Variant Spike (Genbank Accession #QHD43416.1 with B.1.621 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G.  These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.621, Mu Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against the B.1.621 variant in a Biosafety Level 2 facility.

Spike (B.1.621, Mu Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78618-2 500 µl x 2
EUR 3995
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.621 (also known as the Mu Variant) was first identified in Columbia in early 2021. This variant has a number of mutations that may increase morbidity and mortality and allow the virus to spread more easily and quickly than other variants.The Spike (B.1.621, Mu Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.621 Variant Spike (Genbank Accession #QHD43416.1 with B.1.621 mutations; see below for details) as the envelope glycoproteins instead of the commonly used VSV-G.  These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.621, Mu Variant) (SARS-CoV-2) pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against the B.1.621 variant in a Biosafety Level 2 facility.

GR-GAL4 Reporter (Luc)-HEK293 Cell Line (Glucocorticoid Receptor Pathway)

60655 2 vials
EUR 2275
Description: The Glucocorticoid Receptor Pathway GAL4 Reporter (Luc) - HEK293 Cell Line contains a_x000D_firefly luciferase gene under the control of glucocorticoid receptor ligand binding domain that is_x000D_fused to the DNA binding domain (DBD) of GAL4 (GAL4 DBD-GR) stably integrated into_x000D_HEK293 cells. This fusion construct activates firefly luciferase expression under the control of a_x000D_multimerized GAL4 upstream activation sequence (UAS). This allows for specific detection of_x000D_glucocorticoid-induced activation of the glucocorticoid receptor without the need for individual_x000D_transcriptional targets and with low cross-reactivity for other nuclear receptor pathways. This cell_x000D_line is validated for response to stimulation of dexamethasone and to the treatment with_x000D_mifepristone, an inhibitor of the glucocorticoid signaling pathway.

Human Alpha-Actin 2, ACTA2 Differentiation Reporter (pGreenZeo, virus)

SR10068VA-1 >2 x 10^6 IFUs
EUR 691

BLIV 2.0 Reporter: CMV-Luciferase-EF1a-copGFP Pre-packaged Virus

BLIV511VA-1 >2 x10^6 IFUs
EUR 722

Spike (BA.2, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78625-1 100 µl
EUR 900
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. The Omicron Variant (B.1.1.529 variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of February 2022, Omicron variants have been divided into four distinct sub-lineages: BA.1, BA.1.1, BA.2, and BA.3.The Spike (BA.2, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.2 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.2, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.2 variant in a Biosafety Level 2 facility.The Spike Omicron BA.2 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in BA.2, Omicron Variant: T19I, LPPA24-27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, S477N, T478K, E484A, Q493R, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Spike (BA.2, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78625-2 500 µl x 2
EUR 4510
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. The Omicron Variant (B.1.1.529 variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of February 2022, Omicron variants have been divided into four distinct sub-lineages: BA.1, BA.1.1, BA.2, and BA.3.The Spike (BA.2, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.2 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.2, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.2 variant in a Biosafety Level 2 facility.The Spike Omicron BA.2 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in BA.2, Omicron Variant: T19I, LPPA24-27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, S477N, T478K, E484A, Q493R, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78645-1 100 µl
EUR 835
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. Omicron Variant was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of May 2022, Omicron variants have been divided into seven distinct sub-lineages: BA.1, BA.1.1, BA.2, BA.3, BA.2.12.1, BA.4, and BA.5.The Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.2.12.1 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.2.12.1 variant in a Biosafety Level 2 facility.The Spike Omicron BA.2.12.1 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).

Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78645-2 500 µl x 2
EUR 4195
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. Omicron Variant was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of May 2022, Omicron variants have been divided into seven distinct sub-lineages: BA.1, BA.1.1, BA.2, BA.3, BA.2.12.1, BA.4, and BA.5.The Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.2.12.1 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.2.12.1, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.2.12.1 variant in a Biosafety Level 2 facility.The Spike Omicron BA.2.12.1 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).

Human MLC-2v Differentiation Reporter (pGreenZeo, Virus), EF1-Neo Marker

SR10011VA-N >2 x 10^6 IFUs
EUR 691

SRE Luciferase Reporter Lentivirus

78627 500 µl x 2
EUR 835
Description: The SRE (Serum Response Element) Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the Serum Response Element located upstream of the minimal TATA promoter . After transduction, activation of the MAPK/ERK signaling pathway in the target cells can be monitored by measuring the luciferase activity.

Myc Luciferase Reporter Lentivirus

78628 500 µl x 2
EUR 835
Description: The Myc Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the Myc response element located upstream of the minimal TATA promoter and an antibiotic selection gene (puromycin) for the selection of stable clones. After transduction, the Myc signaling pathway in the target cells can be monitored by measuring the luciferase activity.

UAS Luciferase Reporter Lentivirus

78631 500 µl x 2
EUR 835
Description: The UAS (Upstream Activation Sequence) Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by a multimerized GAL4 upstream activation sequence (UAS) located upstream of the minimal TATA promoter and an antibiotic selection gene (puromycin) for the selection of stable clones. After transduction, the UAS-controlled signaling pathway in the target cells can be monitored by measuring the luciferase activity.

p53 Luciferase Reporter Lentivirus

78666 500 µl x 2
EUR 835
Description: The p53 Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce most types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by p53 response elements located upstream of the minimal TATA promoter (Figure 1) and an antibiotic selection gene (puromycin) for the selection of stable clones. After transduction, p53-regulated gene expression in the target cells can be monitored by measuring the luciferase activity.

HRE Luciferase Reporter Lentivirus

78668 500 µl x 2
EUR 835
Description: The Hypoxia Response Element (HRE) Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce most types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by four copies of a hypoxia response elements (HRE) located upstream of the minimal TATA promoter (Figure 1) and an antibiotic selection gene (puromycin) for the selection of stable clones. After transduction, the induction of hypoxia in the target cells can be monitored by measuring the luciferase activity.

ARE Luciferase Reporter Lentivirus

79869 500 µl x 2
EUR 875
Description: The Nrf2 antioxidant response pathway plays an important role in the cellular antioxidant defense. Nrf2, a basic leucine zipper transcription factor, induces the expression of antioxidant and phase II enzymes by binding to the ARE (antioxidant response element) region of the gene promoter. Under basal conditions, Nrf2 is retained in the cytosol by binding to the cytoskeletal protein Keap1. Upon exposure to oxidative stress or other ARE activators, Nrf2 is released from Keap1 and translocates to the nucleus, where it can bind to the ARE, leading to the expression of antioxidant and phase II enzymes that protect the cell from oxidative damage.
The ARE Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by ARE located upstream of the minimal TATA promoter. After transduction, activation of the Nrf2 antioxidant response pathway in the target cells can be monitored by measuring the luciferase activity.

Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78651-1 100 µl
EUR 875
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. Omicron Variant was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of May 2022, Omicron variants have been divided into seven distinct sub-lineages: BA.1, BA.1.1, BA.2, BA.3, BA.2.12.1, BA.4, and BA.5. Among them, BA.4 and BA.5 have identical mutations on their spike protein. The spike protein of BA.4 and BA.5 are referred as BA.4/5 in this datasheet.The Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.4/5 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.4/5 variant in a Biosafety Level 2 facility.As shown in Figures 2 and 3, the Spike Omicron BA.4/5 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in BA.4/5, Omicron Variant:Del69-70, T19I, LPPA24-27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, L452R, S477N, T478K, E484A, F486V, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78651-2 500 µl x 2
EUR 4405
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. Omicron Variant was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. As of May 2022, Omicron variants have been divided into seven distinct sub-lineages: BA.1, BA.1.1, BA.2, BA.3, BA.2.12.1, BA.4, and BA.5. Among them, BA.4 and BA.5 have identical mutations on their spike protein. The spike protein of BA.4 and BA.5 are referred as BA.4/5 in this datasheet.The Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1 containing all the Omicron BA.4/5 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (BA.4/5, Omicron Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 Omicron BA.4/5 variant in a Biosafety Level 2 facility.As shown in Figures 2 and 3, the Spike Omicron BA.4/5 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in BA.4/5, Omicron Variant:Del69-70, T19I, LPPA24-27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, L452R, S477N, T478K, E484A, F486V, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

pGL3 3'UTR reporter WT 1.3 kb CD274 Hs 3'UTR Final Plasmid

PVT17094 2 ug
EUR 390

TEAD Luciferase Reporter Lentivirus

79833 500 µl x 2
EUR 875
Description: The Hippo pathway regulates cell proliferation and cell death. It is activated by high cell density and cell stress to stop cell proliferation and induce apoptosis. The mammalian Hippo pathway comprises MST kinases and LATS kinases. When the Hippo pathway is activated, MST kinases phosphorylate LATS kinases, which phosphorylate transcriptional co-activators YAP and TAZ. Unphosphorylated YAP and TAZ remain in nucleus and interact with TEAD/TEF transcriptional factors to turn on cell cycle-promoting gene transcription. However, when phosphorylated, YAP and TAZ are recruited from the nucleus to the cytosol, so that the YAP and TAZ-dependent gene transcription is turned off. Dysfunction of the Hippo pathway is frequently detected in human cancer and its down-regulation correlates with the aggressive properties of cancer cells and poor prognosis.
The TEAD Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the TEAD response elements located upstream of the minimal TATA promoter. After transduction, activation of the Hippo pathway in the target cells can be monitored by measuring the luciferase activity._x000D_

NF-kB/293/GFP-Luc Transcriptional Reporter Cell Line

TR860A-1 >2 x 10^6 cells
EUR 3142

STAT3 Luciferase Reporter Lentivirus

79744 500 µl x 2
EUR 860
Description: The STAT3 Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene under the control of STAT3-responsive element located upstream of the minimal TATA promoter. After transduction, activation of the STAT3 signaling pathway in the target cells can be monitored by measuring the luciferase activity._x000D_

STAT5 Luciferase Reporter Lentivirus

79745 500 µl x 2
EUR 835
Description: The STAT5 Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene under the control of STAT5-responsive element located upstream of the minimal TATA promoter. After transduction, activation of the STAT5 signaling pathway in the target cells can be monitored by measuring the luciferase activity.

Spike (BA.1.1, Omicron Variant R346K) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78623-1 100 µl
EUR 900
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.1.529 BA.1 (also known as the Omicron Variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. A sub-lineage of BA.1 with an R346K substitution in the spike protein is classified as B.1.1.529 BA.1.1.The Spike (B.1.1.529 BA.1.1, Omicron Variant R346K) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.1.529 BA.1.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.1.529 BA.1.1 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.1.529 BA.1.1, Omicron Variant R346K Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 B.1.1.529 BA.1.1 variant in a Biosafety Level 2 facility.The Spike B.1.1.529 BA.1.1 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in B.1.1.529 BA.1.1 Omicron Variant R346K:A67V, Δ69-70, T95I, G142D, Δ143-145, Δ211, L212I, ins214EPE, G339D, R346K, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, L981F

Spike (BA.1.1, Omicron Variant R346K) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)

78623-2 500 µl x 2
EUR 4510
Description: The pandemic coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). As the first step of the viral replication, the virus attaches to the host cell surface before entering the cell. The viral Spike protein recognizes and attaches to the Angiotensin-Converting Enzyme 2 (ACE2) receptor found on the surface of type I and II pneumocytes, endothelial cells, and ciliated bronchial epithelial cells. Drugs targeting the interaction between the Spike protein of SARS-CoV-2 and ACE2 may offer protection against the viral infection. A variant called B.1.1.529 BA.1 (also known as the Omicron Variant) was identified in South Africa in November of 2021. This variant has a large number of mutations that allow the virus to spread more easily and quickly than other variants. A sub-lineage of BA.1 with an R346K substitution in the spike protein is classified as B.1.1.529 BA.1.1.The Spike (B.1.1.529 BA.1.1, Omicron Variant R346K) (SARS-CoV-2) Pseudotyped Lentiviruses were produced with SARS-CoV-2 B.1.1.529 BA.1.1 Variant Spike (Genbank Accession #QHD43416.1 with B.1.1.529 BA.1.1 mutations; see below for details) as the envelope glycoprotein instead of the commonly used VSV-G. These pseudovirions contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be measured via luciferase activity. The Spike (B.1.1.529 BA.1.1, Omicron Variant R346K Variant) (SARS-CoV-2) pseudovirus can be used to measure the activity of a neutralizing antibody against SARS-CoV-2 B.1.1.529 BA.1.1 variant in a Biosafety Level 2 facility.The Spike B.1.1.529 BA.1.1 pseudovirus has been validated for use with target cells ACE2-HEK293 (which overexpress ACE2; BPS Bioscience #79951).Spike Mutations in B.1.1.529 BA.1.1 Omicron Variant R346K:A67V, Δ69-70, T95I, G142D, Δ143-145, Δ211, L212I, ins214EPE, G339D, R346K, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, L981F

Sox2 SRR2-pGreenFire Response Reporter (pre-packaged virus, EF1-Puro marker)

SR20071-VA-P >2 x 10^6 IFUs
EUR 670

pGreenFire 2.0 AP-1 reporter virus (pGF2-AP1-rFluc-T2A-GFP-mPGK-Puro)

TR452VA-P >2 x 10^6 IFUs
EUR 702

pGreenFire 2.0 NFkB reporter virus (pGF2-NFκB-rFluc-T2A-GFP-mPGK-Puro)

TR412VA-P >2 x 10^6 IFUs
EUR 702

pGreenFire 2.0 NFAT reporter virus (pGF2-NFAT-rFluc-T2A-GFP-mPGK-Puro)

TR451VA-P >2 x 10^6 IFUs
EUR 702

pGreenFire 2.0 HIF-1 reporter virus (pGF2-HIF1-rFluc-T2A-GFP-mPGK-Puro)

TR426VA-P >2 x 10^6 IFUs
EUR 702

NF-κB Luciferase Reporter Lentivirus

79564 500 µl x 2
EUR 875
Description: The NF-κB Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by four copies of the NF-κB response element located upstream of the minimal TATA promoter. After transduction, activation of the NF-κB signaling pathway in the target cells can be monitored by measuring the luciferase activity.

CRE/CREB Luciferase Reporter Lentivirus

79580 500 µl x 2
EUR 835
Description: The main role of the cAMP response element, or CRE, is mediating the effects of Protein Kinase A (PKA) by way of transcription. Upon phosphorylation, CREB forms a functionally active dimer that binds the CRE element within the promoters of target genes and activates transcription. CRE is at the focus of many extracellular and intracellular signaling pathways, including cAMP, calcium, GPCR (G-protein coupled receptors) and neurotrophins. The cAMP/PKA signaling pathway is critical to numerous life processes in living organisms.The CRE/CREB Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by multimerized cAMP response element (CRE) located upstream of the minimal TATA promoter. After transduction, activation of the cAMP/PKA signaling pathway in the target cells can be monitored by measuring the luciferase activity.

pGreenFire 2.0 TCF/LEF reporter virus (pGF2-TCF/LEF-rFluc-T2A-GFP-mPGK-Puro)

TR413VA-P >2 x 10^6 IFUs
EUR 702

BLIV 2.0 Reporter: MSCV-Luciferase-EF1a-copGFP-T2A-Puro Pre-packaged Virus

BLIV713VA-1 >2 x10^6 IFUs
EUR 722

NFAT Luciferase-RFP Reporter Lentivirus

78617-H 500 µl x 2
EUR 835
Description: The NFAT Luciferase-RFP Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase and RFP (Red Fluorescent Protein) cassette driven by the NFAT response element located upstream of the minimal TATA promoter and a hygromycin or puromycin selection gene to generate stable clones. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by measuring the luciferase activity or RFP expression. RFP fluoresces red-orange when excited; it has an excitation wavelength of 553 nm, and an emission wavelength of 574 nm.

NFAT Luciferase-RFP Reporter Lentivirus

78617-P 500 µl x 2
EUR 835
Description: The NFAT Luciferase-RFP Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase and RFP (Red Fluorescent Protein) cassette driven by the NFAT response element located upstream of the minimal TATA promoter and a hygromycin or puromycin selection gene to generate stable clones. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by measuring the luciferase activity or RFP expression. RFP fluoresces red-orange when excited; it has an excitation wavelength of 553 nm, and an emission wavelength of 574 nm.

NFAT eGFP Reporter Lentivirus

79922 500 µl x 2
EUR 875
Description: The NFAT eGFP Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain an enhanced GFP gene driven by the NFAT response element located upstream of the minimal TATA promoter. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by examining eGFP expression._x000D_

AADACL4 siRNA

20-abx906242
  • Ask for price
  • Ask for price
  • 15 nmol
  • 30 nmol

AADACL4 siRNA

20-abx906243
  • Ask for price
  • Ask for price
  • 15 nmol
  • 30 nmol

STAT3 eGFP Reporter Lentivirus

78197 500 µl x 2
EUR 795
Description: The STAT3 eGFP Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain an eGFP gene under the control of a STAT3-responsive element located upstream of the minimal TATA promoter . After transduction, activation of the STAT3 signaling pathway in the target cells can be monitored by examining eGFP expression.

AADACL4 Antibody

31298 100ul
EUR 319

AADACL4 Antibody

31298-100ul 100ul
EUR 302.4

AADACL4 Antibody

31298-50ul 50ul
EUR 224.4

AADACL4 Antibody

1-CSB-PA782584
  • Ask for price
  • Ask for price
  • 100ul
  • 50ul
Description: A polyclonal antibody against AADACL4. Recognizes AADACL4 from Human. This antibody is Unconjugated. Tested in the following application: ELISA, WB;ELISA:1:1000-1:2000, WB:1:200-1:1000

AADACL4 Antibody

E031298 100μg/100μl
EUR 255
Description: Available in various conjugation types.

AADACL4 Antibody

E301539 100ug/200ul
EUR 275
Description: Available in various conjugation types.

AADACL4 antibody

70R-6766 50 ug
EUR 467
Description: Rabbit polyclonal AADACL4 antibody raised against the middle region of AADACL4

AADACL4 antibody

70R-6671 50 ug
EUR 467
Description: Rabbit polyclonal AADACL4 antibody raised against the N terminal of AADACL4

AADACL4 Antibody

GWB-MP906F 50ug Ask for price

AADACL4 Antibody

GWB-MP907G 50ug Ask for price

Lenti ORF particles, AADACL4 (mGFP-tagged) - Human arylacetamide deacetylase-like 4 (AADACL4), 200ul, >10^7 TU/mL

RC222634L4V 200 µl Ask for price

NF-κB eGFP Reporter Lentivirus

79926 500 µl x 2
EUR 820
Description: The NF-κB eGFP Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to infect almost all types of mammalian cells, including primary and non-dividing cells. The particles contain an enhanced GFP gene driven by the NF-κB response element located upstream of the minimal TATA promoter. After transduction, activation of the NF-κB signaling pathway in the target cells can be monitored by examining eGFP expression.

Lenti ORF particles, AADACL4 (Myc-DDK tagged) - Human arylacetamide deacetylase-like 4 (AADACL4), 200ul, >10^7 TU/mL

RC222634L3V 200 µl Ask for price

CRE/CREB eGFP Reporter Lentivirus

78153 500 µl x 2
EUR 795
Description: The CRE/CREB eGFP Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain an eGFP gene driven by a multimerized cAMP response element (CRE) located upstream of the minimal TATA promoter . After transduction, activation of the cAMP/PKA signaling pathway in the target cells can be monitored by examining eGFP expression.

Luciferase Reporter Assay Kit

55R-1540 200 assays
EUR 180
Description: Assay Kit for detection of Luciferase Reporter in the research laboratory

Luciferase Reporter Assay Kit

GWB-AXR371 200 assays Ask for price

Luciferase Reporter Assay Kit

K2181-200 200 assays
EUR 130

Luciferase Reporter Assay Kit

K801-200 each
EUR 235.2

IL-2 Promoter Luciferase Reporter Lentivirus

79825 500 µl x 2
EUR 795
Description: The IL-2 Promoter Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the human IL-2 promoter. After transduction, activation of the IL-2 signaling pathway in the target cells can be monitored by measuring the luciferase activity._x000D_

IL-8 Promoter Luciferase Reporter Lentivirus

79827 500 µl x 2
EUR 795
Description: The IL-8 Promoter Luciferase Reporter Lentivirus are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to be transduced into almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the human IL-8 promoter. After transduction, activation of the IL-8 signaling pathway in the target cells can be monitored by measuring the luciferase activity._x000D_

Bald Lentiviral Pseudovirion (Luciferase Reporter)

79943 500 µl x 2
EUR 875
Description: The bald lentiviral pseudovirion was produced without envelope glycoproteins such as VSV-G or SARS-CoV-2 spike. It contains the firefly luciferase gene driven by a CMV promoter as the reporter. The bald lentiviral pseudovirion can serve as a negative control when studying virus entry initiated by specific interactions between virus particles and receptors._x000D_

AADACL4 (untagged)-Human arylacetamide deacetylase-like 4 (AADACL4)

SC301805 10 µg Ask for price

NFAT Luciferase Reporter Lentivirus-79579-G

79579-G 500 µl x 2
EUR 835
Description: The NFAT Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the NFAT response element located upstream of the minimal TATA promoter (Figure 1) and an antibiotic selection gene (hygromycin, puromycin, or G418) for the selection of stable clones. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by measuring the luciferase activity.

NFAT Luciferase Reporter Lentivirus-79579-H

79579-H 500 µl x 2
EUR 860
Description: The NFAT Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the NFAT response element located upstream of the minimal TATA promoter and an antibiotic selection gene (hygromycin or puromycin) for the selection of stable clones. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by measuring the luciferase activity.

NFAT Luciferase Reporter Lentivirus-79579-P

79579-P 500 µl x 2
EUR 860
Description: The NFAT Luciferase Reporter Lentiviruses are replication incompetent, HIV-based, VSV-G pseudotyped lentiviral particles that are ready to transduce almost all types of mammalian cells, including primary and non-dividing cells. The particles contain a firefly luciferase gene driven by the NFAT response element located upstream of the minimal TATA promoter and an antibiotic selection gene (hygromycin or puromycin) for the selection of stable clones. After transduction, activation of the NFAT signaling pathway in the target cells can be monitored by measuring the luciferase activity.

A total of 32 (11.6%) HCMV-positive cases detected (mean Ct 30.59); the mean and median age of 10.3 and 6 days old, respectively. Children aged between 0-3 weeks HCMV has a higher detection rate (84.4%; 27/32). UL89 gene successfully sorted into two samples, both classified as human betaherpesvirus 5. Not Specified associated resistance mutations were identified. A routine screening in newborns coupled with the genetic characterization of viral genes important key to lowering the sequel associated with congenital HCMV infection.

Leave A Comment