Background: Congenital cytomegalovirus (CMV) is the most common cause of non-genetic sensorineural hearing loss (SHNL) in children. Only about 10% to 15% of children with congenital CMV is a symptom, and most are not diagnosed at birth. Approximately 7% to 15% of clinically asymptomatic patients may develop complications later, including Sensorineural hearing loss, which is the most common sequelae in patients with clinically asymptomatic. In this study, the hearing status were investigated in children with confirmed CMV infection and newborn hearing screening (NHS) is reviewed to explore the history of hearing loss caused by CMV.
Methods: The medical records of 58 children diagnosed with CMV infection was confirmed in terms of clinical symptoms and signs of CMV infection. Status hearing evaluated by audiological test battery according to age.
Results: A total of 58 children (M: F = 32:26 patient, age of study: mean, 5.62 years, range, 1-10 years) serologically diagnosed with CMV infection (14 patients, 21.1%), or diagnosed through PCR of serum (5, 7.9%) and / or PCR from urine (19, 26.8%). Hearing loss was confirmed in 11 children (19.0%), being bilateral in 6 (54.5%), and unilateral in 5 (45.5%). Note that 7 out of 17 ears with hearing loss passed NHS and diagnosed only after a re-evaluation if CMV infection is identified.
Conclusion: Hearing loss is a serious complication of CMV infection in children. Our results highlight the importance of timely audiological evaluation in children with clinical symptoms of CMV infection even if they pass the NHS.
Rare cases of Cytomegalovirus in Gut in immunocompetent hosts With Ischemic Colitis
Cytomegalovirus (CMV) infection is usually seen in individuals with immunosuppressive conditions such as malignancy, HIV / AIDS, and organ transplants, and in patients with chemotherapy or steroids. recurrent disease can occur if the reactivation of the virus because the immune disorder due to factors such as older age or immunosuppressive drugs. CMV is a common, with seroprevalence (CMV IgG) from 40-100% in adults, increases with age. It has been reported that inflammatory bowel disease in remission can be exacerbated by CMV complicate steroid refractory ulcerative colitis or flares. For this reason, steroids should be carefully initiated if a high clinical suspicion for CMV. We report a unique case of CMV colitis is associated with severe ischemic colitis in immunocompetent patients, with a very good response to management with antiviral therapy.
While the cytomegalovirus (CMV) reactivation occurs in critically ill immunocompetent patients and has been associated with worse outcomes, very few cases of CMV reactivation have been reported following the left ventricular assist device (LVAD) implantation. We aimed to evaluate the incidence and risk factors for CMV reactivation following LVAD implantation. A retrospective chart review of patients who have undergone LVAD implantation between July 2004 and December 2018 do. CMV reactivation cases with post-LVAD randomly matched (1: 2) by sex, type of LVAD, and the implant to control utilizing SAS macros.
Bald Lentiviral Pseudovirion (Luc-eGFP Dual Reporter)
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_
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.
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.
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).
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.
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
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
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.
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.
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
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
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
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
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.
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.
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).
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).
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.
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.
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_
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.
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)
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.
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.
Human GFAP Differentiation Reporter (pRedZeo, Virus)
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.
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.
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.
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_
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_
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).
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).
Human Insulin Differentiation Reporter (pGreenZeo, Virus)
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.
Human Osteocalcin Differentiation Reporter (pGreenZeo, Virus)
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)
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.
CD40/NF-κB Reporter (Luc) - HEK293 Stable Cell Line
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_
Human Keratin 14 Differentiation Reporter (pGreenZeo, Virus)
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)
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_
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_
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_
Myc Reporter (Luc) - HCT116 Cell Line (Myc Signaling Pathway)
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
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.
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_
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_
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_
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_
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.
Human E-Cadherin, CDH1 Differentiation Reporter (pGreenZeo, virus)
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_
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 (B.1.621, Mu Variant) (SARS-CoV-2) Pseudotyped Lentivirus (Luc Reporter)
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)
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.
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
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
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).
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 Alpha-Actin 2, ACTA2 Differentiation Reporter (pGreenZeo, virus)
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
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
CRE/CREB Reporter (Luc) - HEK293 Cell Line (cAMP/PKA Signaling Pathway)
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)
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.
Human MLC-2v Differentiation Reporter (pGreenZeo, Virus), EF1-Neo Marker
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.
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.
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.
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.
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.
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.
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_
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
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
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_
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.
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.
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.
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.
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.
NF-kB/293/GFP-Luc Transcriptional Reporter Cell Line
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_
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.
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_
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)
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_
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.
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.
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.
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.
Description: The Xenobiotic response element (XRE) 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 three copies of an XRE located upstream of the minimal TATA promoter (Figure 1), and an antibiotic selection gene (puromycin) for the selection of stable clones. After transduction, the activation of aryl hydrocarbon receptor (AhR) in the target cells can be monitored by measuring the luciferase activity.
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.
Description: The SBE Luciferase Reporter Lentivirus (TGFβ/SMAD signaling pathway) 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 SBE-responsive element located upstream of the minimal TATA promoter. After transduction, activation of the TGFβ/SMAD signaling pathway can be monitored by measuring the luciferase activity._x000D_
Description: The NFAT Luciferase-eGFP 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 eGFP cassette driven by the NFAT response element located upstream of the minimal TATA promoter (Figure 1) and a 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 eGFP expression.
Description: The NFAT Luciferase-eGFP 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 eGFP cassette driven by the NFAT response element located upstream of the minimal TATA promoter (Figure 1) and a 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 eGFP expression.
Description: The stress-activated protein kinase/c-jun N-terminal kinase (SAPK/JNK) family of proteins includes mitogen-activated protein kinases (MAPKs) that are activated by stress, inflammatory cytokines, mitogens, oncogenes, and inducers of cell differentiation and morphogenesis. Upon activation of the SAPK/JNK pathway, MAP Kinase Kinases phosphorylate and activate JNKs. The activated JNKs translocate to the nucleus where they phosphorylate and activate transcription factors such as c-Jun. c-Jun then binds to the activator protein-1 (AP1) response element and induces AP1 transcription. The AP1 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 AP1 response element located upstream of the minimal TATA promoter. After transduction, activation of the JNK signaling pathway and AP1 mediated activity in the target cells can be monitored by measuring the luciferase activity.
Description: The bald lentiviral pseudovirion was produced without envelope glycoproteins such as VSV-G or SARS-CoV-2 spike. It contains the eGFP 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_
Description: The GAS 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 three copies of the interferon gamma (IFN-γ) activated sites (GAS) located upstream of the minimal TATA promoter and a puromycin selection gene for the selection of stable clones. After transduction, the GAS-regulated gene expression in the target cells can be monitored by measuring the luciferase activity.
Description: The NFAT reporter (Luciferase)-THP-1 cell line is designed for monitoring the NFAT (nuclear factor of activated T-cells) signaling pathway in THP-1 cells by measuring luciferase activity. It contains a firefly luciferase gene driven by the NFAT response element located upstream of the minimal TATA promoter. Upon activation by NFAT activators such as Ionomycin, endogenous NFAT transcription factors bind to the DNA response elements, inducing transcription of the luciferase reporter gene.
Description: The JAK/STAT pathway is activated by various cytokines and growth factors and plays a critical role in cell growth, hematopoiesis, and immune response. In mammals, there are four JAKs (JAK1, JAK2, JAK3 and TYK2) and seven STAT proteins. IFNα is a Type I interferon. Binding of IFNα to its receptor leads to the activation of JAK1 and TYK2, which in turn phosphorylate and activate STAT1 and STAT2. The phosphorylated STAT1 and 2 form a heterodimer and bind to IRF9/p48, forming a protein complex ISGF3. This complex translocates to the nucleus and binds to the ISRE (Interferon Stimulated Response Element) in the promoter region, thereby promoting transcription of interferon-inducible genes. The ISRE 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 ISRE response element located upstream of the minimal TATA promoter. After transduction, the activity of Type I interferon-induced JAK/STAT signaling pathway in the target cells can be monitored by measuring the luciferase activity.
Description: The SARS-CoV-2 Spike Pseudotyped Lentivirus were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions also contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be conveniently measured via luciferase reporter activity. The SARS-CoV-2 Spike pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 in a Biosafety Level 2 facility._x000D_ _x000D_
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._x000D_The SARS-CoV-2 Spike Pseudotyped Lentivirus were produced with SARS-CoV-2 Spike (Genbank Accession #QHD43416.1) as the envelope glycoproteins instead of the commonly used VSV-G. These pseudovirions also contain the firefly luciferase gene driven by a CMV promoter, therefore, the spike-mediated cell entry can be conveniently measured via luciferase reporter activity. The SARS-CoV-2 Spike pseudotyped lentivirus can be used to measure the activity of neutralizing antibody against SARS-CoV-2 in a Biosafety Level 2 facility._x000D_ _x000D_
Description: The bald VSV Delta G (Luciferase Reporter) was produced without envelope glycoproteins. It contains the firefly luciferase gene as the reporter. The bald VSV Delta G (Luciferase Reporter) can serve as a negative control when studying virus entry initiated by specific interactions between virus particles and receptors.
Description: The bald VSV Delta G (Luciferase Reporter) was produced without envelope glycoproteins. It contains the firefly luciferase gene as the reporter. The bald VSV Delta G (Luciferase Reporter) can serve as a negative control when studying virus entry initiated by specific interactions between virus particles and receptors.
1 Vial, Each vial contains 2 ~ 3 x 10^6 cells in 1 ml of 90% FBS + 10% DMSO.
EUR 1864.8
Description: The HRE Luciferase Reporter cell line is a stably transfected HeLa cell line which expresses Renilla luciferase reporter gene under the transcriptional control of the hypoxia response element (HRE). In response to hypoxia (low oxygen), HREs of target genes are recognized and regulated by the hypoxia-inducible factors (HIFs) which belong to the family of basic helix-loop-helix transcription factors and form heterodimeric complex comprising the alpha subunit (HIF-1 alpha, HIF-2 alpha and HIF-3 alpha) and beta subunit (Arnt1, Arnt2 and Arnt3), among which HIF-1 alpha and HIF-2 alpha are predominant isoforms. Activation of HIFs can also be mediated by chemical hydroxylase inhibitors as hypoxia mimetics including the iron chelator desferrioxamine and cobalt chloride.The HRE induction by cobalt chloride is shown in Figure 1.
×
Fisher’s exact and paired sample t-tests were conducted to evaluate the difference between continuous and categorical variables, respectively. Valid for reactivation implantation post-LVAD counted in cases, and implantation of the appropriate time post-LVAD determined in control patients for comparisonsDays variable reactivation of implantation post-LVAD counted in cases, and implantation of the appropriate time post-LVAD determined in patients controls for comparison variables.