Recommended
product-img
  • Products
  • Discover MP Targets
  • Discover Research Areas
  • mProX™ Human OPRM1 Stable Cell Line

    [CAT#: S01YF-0923-PY153]
    Product Category:
    Membrane Protein Stable Cell Lines
    Subcategory:
    GPCR Cell Lines

    Datasheet MSDS Request COA

    Certificate of Analysis Lookup
    To download a Certificate of Analysis, please enter a lot number in the search box below. Note: Certificate of Analysis not available for kit components.
    Lot Number

    Made to Order Inquiry

    Inquiry
    Host Cell Type:
    Membrane Protein Engineering:
    Fluorescent Marker:
    Resistance:
    Deliverable:

    Product Information

    Target Protein
    OPRM1
    Target Family
    Opioid Family
    Target Protein Species
    Human
    Host Cell Type
    FaDu;MDA686Tu;CHO-K1;HEK293
    Target Classification
    GPCR Cell Lines
    Target Research Area
    Pain and Addiction Research
    Related Diseases
    Substance Dependence;Drug Dependence
    Gene ID
    Human: 4988
    UniProt ID
    Human: P35372

    Product Properties

    Biosafety Level
    Level 1
    Activity
    Yes
    Quantity
    10⁶ cells per vial
    Applications
    The OPRM1 gene encodes the mu opioid receptor, which is the primary target of most opioid analgesics. This receptor plays a crucial role in pain perception, reward mechanisms, and addiction. Research has shown that polymorphisms in the OPRM1 gene can influence an individual's response to opioid medications, making it a significant focus in the field of pharmacogenetics. Additionally, OPRM1 has been studied for its role in various psychiatric disorders, including depression and anxiety.

    Protocols

    Please visit our protocols page.

    Customer Reviews

    There are currently no Customer reviews or questions for mProX™ Human OPRM1 Stable Cell Line (S01YF-0923-PY153). Click the button above to contact us or submit your feedback about this product.

    FAQ

    chat Ashley (Verified Customer)

    Are there any known associations between OPRM1 polymorphisms and alcohol use disorder? Aug 08 2020

    chat Patrick Liam (Creative Biolabs Scientific Support)

    The OPRM1 gene has been studied in relation to its response to naltrexone treatment for alcohol dependence. However, some studies did not find an association of the OPRM1 Asn40Asp polymorphism with naltrexone treatment response for alcohol dependence. Aug 08 2020

    chat Gary (Verified Customer)

    How does OPRM1 gene variation influence the effects of certain treatments on heavy drinkers? May 30 2022

    chat Patrick Liam (Creative Biolabs Scientific Support)

    Studies have explored the therapeutic effects of targeted treatments like nalmefene on heavy drinkers in relation to polymorphic variation in opioid receptor genes, including OPRM1. However, the findings suggest that the therapeutic effects were not moderated by such polymorphic variations5. response for alcohol dependence. May 30 2022

    Published Data

    Fig.1 Downregulation of OPRM1 had a pronounced impact, resulting in a substantial reduction in colony formation for both FaDu and MDA686Tu cells.

    The inhibition of OPRM1 expression led to a substantial reduction in colony formation rates in both FaDu (mean difference [MD], 95% confidence interval [CI]: 98.50%, 69.50 to 127.5, p < 0.0001) and MDA686Tu (MD, 95% CI: 98.50%, 69.50 to 127.5, p < 0.0001) cell lines when compared to untreated control cells.

    Ref: Gorur, Aysegul, et al. "Mu-opioid receptor activation promotes in vitro and in vivo tumor growth in head and neck squamous cell carcinoma." Life Sciences 278 (2021): 119541.

    Pubmed: 33930368

    DOI: 10.1016/j.lfs.2021.119541

    Research Highlights

    van Dokkum NH, et al. "Neonatal stress exposure and DNA methylation of stress-related and ." Early human development, 2023.
    In the study conducted, it was found that extremely preterm infants may experience long-lasting effects on neurodevelopmental outcomes as a result of stress exposure during their stay in the Neonatal Intensive Care Unit (NICU). This may be attributed to altered DNA methylation of stress-related and neurodevelopmentally relevant genes. The study aimed to explore the association between neonatal stress exposure and DNA methylation in these genes at two different time points: 7-14 days after birth and at discharge from the NICU. The study included 45 infants, with a gestational age range of 24-30 weeks, and utilized the Neonatal Infant Stressor Scale (NISS) to measure daily neonatal stress exposure. Fecal samples were collected at days 7-14 and discharge, and DNA methylation analysis was performed on a subset of samples due to low DNA concentrations. Results showed that methylation rates of most genes did not significantly differ between the two time points, except for one gene (OPRM1). However, a moderate and mostly negative correlation was found between cumulative NISS scores and average DNA methylation status upon discharge for several genes, including NR3C1, SLC6A4, SLC1A2, IGF2, BDNF, and OPRM1. The study highlights the potential for differential gene regulation in response to higher neonatal stress exposure and suggests the need for further research to confirm and study the effects on gene expression.
    Pubmed: 37797474   DOI: 10.1016/j.earlhumdev.2023.105868

    Zeng H, et al. "Unbiased multitissue transcriptomic analysis reveals complex neuroendocrine ." Journal of neuroinflammation, 2023.
    Spinal cord injury (SCI) results in the loss of sensory and motor function below the level of injury and is a devastating disease of the central nervous system. It leads to severe secondary immunosuppression, known as SCI-induced immunodeficiency syndrome (SCI-IDS), which increases the susceptibility to infections and worsens neurological dysfunction. Multiple studies have indicated that SCI-IDS is an independent risk factor for poor neurological prognosis. SCI-IDS is primarily observed in injuries above the T5 level and is believed to cause systemic immune failure through the sympathetic-adrenal medullary axis and the hypothalamic-pituitary-adrenal (HPA) axis, although the exact mechanism is unclear. The concentrations of adrenocorticotropic hormone and cortisol in plasma, as well as changes in sympathetic activity, were evaluated in rats with high-level (T3) spinal cord injury (T3-SCI) and low-level (T10) spinal cord injury (T10-SCI). Furthermore, the underlying gene network regulation between the sympathetic-adrenal medullary axis and the HPA axis was investigated through histology and multitissue transcriptomics to better understand the neuroendocrine-immune network associated with SCI-IDS. The results showed significant atrophy in secondary immune organs, such as the spleen and thymus gland, in rats with T3-SCI, and a marked increase in cortisol levels mediated by the adrenal glands but a decrease in norepinephrine levels. Transcriptome analysis also revealed distinct gene expression patterns in the hypothalamus and adrenal glands of rats with T3-SCI, with notable changes in genes involved in inflammatory pathways and immune response pathways. Additionally, disruptions in the hypothalamic-adrenal gland connection led to autonomous adrenal gland regulation, disturbance in circadian rhythm, and ultimately elevated cortisol levels, causing overall immunosuppression in the periphery. This study sheds light on the possible mechanism of SCI-IDS and its impact on neurological prognosis through persistent neuroinflammation and systemic immunosuppression.
    Pubmed: 37775760   DOI: 10.1186/s12974-023-02906-7

    Please note: All products are "FOR RESEARCH USE ONLY. NOT FOR USE IN DIAGNOSTIC OR CLINICAL PROCEDURES" For licensing inquiries, please contact
    Send Inquiry Send Inquiry
    Inquiry Basket
    compare

    Go to compare