scispace - formally typeset
Search or ask a question

Hoechst 33342 good for cell viability study? 


Best insight from top research papers

Hoechst 33342 can be a valuable tool for cell viability studies, but its efficacy depends on the concentration used and the specific application. Research indicates that lower concentrations of Hoechst 33342 (7-28 nM) are non-cytotoxic and do not impact cell viability, making it suitable for live cell imaging studies . Additionally, Hoechst 33342 has been shown to be effective in evaluating sperm viability through flow cytometry, especially when combined with propidium iodide (PI) for dual staining . However, caution is advised as higher concentrations of Hoechst 33342 can inhibit proliferation and impair cell viability, emphasizing the importance of optimizing exposure parameters for accurate results . Overall, when used appropriately, Hoechst 33342 can provide valuable insights into cell viability and function in various research settings.

Answers from top 4 papers

More filters
Papers (4)Insight
Hoechst 33342 at concentrations between 7-28 nM is suitable for cell viability studies as it does not affect cell proliferation, viability, or signaling pathways, allowing real-time high-throughput live-cell imaging.
Yes, Hoechst 33342 is suitable for cell viability studies as it does not exhibit toxicity over a 6-hour period and provides consistent results with end-point assays.
Hoechst 33342 is relatively unsuitable for cell viability studies due to rapid redistribution into non-labeled cells, limiting its effectiveness as a cell marker in transplantation research.
Yes, Hoechst 33342 (H342) is effective for assessing rhesus monkey sperm viability, showing strong correlation with expected viability and providing accurate results for flow cytometric analysis.

Related Questions

Does hoechst 33342 stain only live cells?5 answersHoechst 33342 (H33342) is commonly used to stain DNA in living cells. While traditionally believed to be suitable only for endpoint analyses due to cytotoxicity concerns, recent studies challenge this notion. Optimized exposure parameters allow for Hoechst staining in live cells without significant cytotoxic effects at concentrations between 7 and 28 nM. Additionally, Hoechst 33342 can visualize neurites of dorsal root ganglion cells when fixed, providing a practical and cost-effective alternative for staining. Furthermore, Hoechst 33342, in combination with Pyronin Y, enables the distinction between quiescent and proliferating cells based on DNA and RNA content, showcasing its versatility in cell analysis. Therefore, Hoechst 33342 can be used to stain both live and fixed cells, expanding its utility beyond live cell staining.
Why cell viability over 100% in AlamarBlue® assay?5 answersCell viability over 100% in the AlamarBlue® assay can occur due to factors like the presence of certain drugs affecting the assay readings or plate-to-plate variability. In the study by Dinh et al., unexpected right shifts in dose response curves were observed when testing EGFR inhibitors on lung cancer cells, which led to falsely increased readings. However, modifying the assay by removing the drug-containing medium prior to adding AlamarBlue® eliminated this issue. Additionally, Lee highlighted the cost-effectiveness and ease of the AlamarBlue® assay for screening antimicrobial activity in plant extracts against various pathogens. Furthermore, Zachari et al. emphasized the importance of proper cell culture maintenance and initial cell plating numbers to ensure accurate viability measurements in radiation response experiments using the AlamarBlue® assay.
How does DLEU2 influence on cell viability?5 answersDLEU2 plays a crucial role in influencing cell viability through various mechanisms. In the context of idiopathic pulmonary fibrosis (IPF), DLEU2 upregulation was associated with increased cell proliferation and migration. Additionally, in breast cancer, DLEU2 acted as a coactivator for HIF-1α, promoting the transcription of CKAP2, which contributed to the malignancy of cancer cells. Moreover, in the context of metabolic disorders, DLEU2 knockdown led to decreased sirtuin expression, impacting mitochondrial function and oxidative stress, ultimately affecting cell viability. Furthermore, in renal cell cancer, DLEU7-AS1, a related lncRNA, promoted cell growth and metastasis by silencing the miR-26a-5p/coronin-3 axis. These findings collectively demonstrate that DLEU2 family lncRNAs play significant roles in regulating cell viability across different cellular contexts.
How does transfection affect on cell viability?5 answersTransfection can have a significant impact on cell viability. Various factors influence this relationship, such as the type of transfection method used, the duration and intensity of electric pulses, and the composition of the electroporation medium. Studies have shown that longer electric pulses optimize transfection efficiency but may reduce cell viability, while shorter pulses can maintain viability at the cost of slightly lower transfection rates. Additionally, the choice of buffer composition plays a crucial role in cell viability post-transfection, with Mg2+-based buffers enhancing viability possibly through ATPase activation, although they may hinder transfection efficiency compared to K+-based buffers. Overall, balancing transfection efficiency with cell viability requires careful consideration of these factors to design effective and safe transfection protocols.
What are the effects of nicotine on cell viability in different cell types?5 answersNicotine exhibits diverse effects on cell viability across various cell types. Studies have shown that nicotine significantly reduces cell viability in keratinocytes, cardiomyocytes, and hepatocytes, inducing apoptotic cell death and morphological changes. In lung cancer cell lines, nicotine inhibits viability in one type while inducing proliferation in another, with varying impacts on apoptosis. Moreover, nicotine has been found to modulate inflammatory cytokine expression and induce severe cellular damage in cells infected with SARS-CoV-2, resembling pyroptosis and necroptosis. Additionally, nicotine can enhance cell viability, inhibit apoptosis, and activate specific signaling pathways in Raw264.7 and El4 cells, potentially offering protective effects against toxicity. Furthermore, nicotine has been observed to increase the viability of polymorphonuclear leukocytes and macrophages, particularly in their activated states, potentially exacerbating their deleterious effects.
How to prepare MTS solution to measure cell viability?4 answersTo prepare MTS solution for measuring cell viability, follow the protocol described by Bartz. The protocol involves using a MTS cell proliferation assay kit. Unfortunately, the specific details of the protocol are not provided in the abstract.

See what other people are reading

What is hoechst 33342 concentration for live cell staining?
5 answers
The optimal Hoechst 33342 concentration for live cell staining varies depending on the specific application. Research indicates that concentrations as low as 7-28 nM are suitable for staining nuclei without inducing cytotoxicity or affecting cell viability, proliferation, or signaling pathways. However, caution is advised as concentrations below 1 µM of SiR-Hoechst have been shown to induce DNA damage responses and G2 arrest in human cells. Additionally, Hoechst 33342 can be used in combination with Pyronin Y for measuring DNA and RNA content in live cells, allowing the distinction between quiescent and proliferating cells. It is crucial to consider the specific experimental conditions and desired outcomes when determining the appropriate concentration of Hoechst 33342 for live cell staining.
What does moisture content in propolis affect?
5 answers
The moisture content in propolis affects various aspects such as the physicochemical properties, antioxidant capacity, and stability of bioactive compounds. High moisture content can impact the flow properties of propolis powders and alter the crystalline structure when encapsulated. Propolis with moisture content ranging from 1.96% to 8.26% showed variability in physicochemical parameters and antioxidant activity, indicating the importance of moisture levels in determining propolis quality. Additionally, moisture content in raw propolis samples collected from different regions ranged from 0.98% to 2.97%, influencing the total phenolics and flavonoids content, which are crucial for the antioxidant potential of propolis. Therefore, controlling and maintaining optimal moisture content in propolis is essential for preserving its bioactive compounds and ensuring its beneficial properties.
What kind of cells are best to be used for cell viability test for novel materials for OECT?
5 answers
Human fibroblasts, keratinocytes, and periodontal ligament (PDL) cells are suitable for cell viability tests on novel materials for Organic Electrochemical Transistors (OECTs). These cells have been extensively used in biocompatibility studies to assess the response of human tissues to various materials, including stainless steel, polyether-ether-ketone (PEEK), and different sealers. Evaluating cell viability is crucial for determining the cytotoxic effects and biocompatibility of materials, ensuring their safety and effectiveness for medical applications. Studies have shown that the viability and morphology of these cells can provide valuable insights into the biological performance of materials, guiding the development of implant platforms and prosthetic components. Therefore, utilizing human fibroblasts, keratinocytes, and PDL cells in cell viability tests can aid in assessing the suitability of novel materials for OECT applications.
Which countries have implemented regulations that require somatic cell count testing for milk production?
5 answers
Several countries have implemented regulations mandating somatic cell count (SCC) testing for milk production. The European Union (EU) established a maximum threshold of 400,000 cells/mL for raw milk in 2014. Additionally, various countries have specific acceptable SCC limits, with the necessity of SCC detection being emphasized for monitoring udder health and ensuring milk quality and safety. The importance of SCC testing is further highlighted by the fact that high SCC levels can lead to physical and chemical changes in milk composition, affecting its nutritional value and shelf life. Methods for SCC detection include conventional microscopic techniques and state-of-the-art biosensor-based methods, with biosensor-based approaches being favored for their speed, sensitivity, and cost-effectiveness.
What types of nanoparticles have shown promise in treating colorectal cancer?
10 answers
Nanoparticles have emerged as a significant advancement in the treatment of colorectal cancer (CRC), offering innovative approaches to overcome the limitations of conventional therapies. Various types of nanoparticles have been identified and explored for their potential in CRC treatment, each with unique properties and mechanisms of action. Liposomes, niosomes, solid lipid nanoparticles, micelles, and polymeric nanoparticles are among the miniaturized nanocarriers that have been employed for both passive and active targeting in CRC, showing promise in enhancing diagnosis, treatment, and theranostic applications. Magneto-fluorescent NANO3 nanoparticles, specifically designed for combinatory anti-tumoral nanotherapy, have demonstrated significant cytotoxicity against CRC cells, especially when modified with targeting agents such as an anti-CA IX acetazolamide derivative and Cetuximab. The exploration of nanomaterials like carbon nanotubes, dendrimers, silica nanoparticles, gold nanoparticles, and metal-organic frameworks has also been highlighted for their potential in targeted anticancer drug delivery and diagnostic purposes. Photodynamic therapy using porphysome nanoparticles represents another innovative approach, offering outpatient neoadjuvant treatment possibilities for CRC. The application of nanotechnology has been further emphasized for its role in overcoming therapeutic resistance and preventing metastasis, with nanomaterials enhancing drug mobility, treatment efficacy, and reducing systemic toxicity. Nanoparticles have also been utilized to reverse the immunosuppressive tumor microenvironment (TME), thereby potentiating the effect of immunotherapeutic agents. Diatomite nanoparticles (DNPs) have been introduced as biocompatible nanocarriers capable of delivering chemotherapeutic agents against specific targets while reducing off-target effects. The review of nanocarrier-based strategies for CRC treatment has underscored the necessity for effective and targeted options, with several nanoparticles recommended for clinical purposes. Plasmonic (gold) nanoparticles have been identified for their dual therapeutic functionalities as photothermal therapy agents and radiosensitizers, offering a synergistic combination of radio- and phototherapies. Lastly, the use of polymeric nanoparticles, gold nanoparticles, liposomes, and solid lipid nanoparticles has been discussed for their efficacy in drug delivery systems specifically targeting CRC. These findings collectively underscore the diverse and promising role of nanoparticles in advancing the treatment of colorectal cancer, highlighting the potential for more effective, targeted, and less toxic therapeutic options.
How does the RAS gene contribute to the development of preeclampsia?
5 answers
The renin-angiotensin system (RAS) plays a crucial role in the pathogenesis of preeclampsia (PE). Dysregulation of the RAS gene components, such as angiotensin-converting enzyme (ACE) and angiotensin II (Ang II), contributes to the development of PE. Genetic variations in the ACE gene, like the rs4343 polymorphism, have been associated with an increased risk of PE. Additionally, the overactivation of the Ang II/ACE/AT1R axis, along with decreased levels of protective components like ACE 2/Ang 1–7/MasR, can lead to hypertension characteristic of PE. Furthermore, studies suggest that the compensatory alterations in the RAAS during normal pregnancy are disrupted in PE, leading to a disturbance in the delicate equilibrium necessary for maternal and fetal well-being. These findings highlight the intricate involvement of the RAS gene in the pathogenesis of preeclampsia.
Dose of SN-38 for colorectal cancer treatment
5 answers
The dose of SN-38 for colorectal cancer treatment varies depending on the formulation and delivery method. Studies have explored different approaches to enhance the efficacy of SN-38. For instance, one study developed SN-38-loaded gold nanoshells nanoparticles for combined chemo-photothermal therapy, showing significant tumor growth suppression. Another study focused on a lipophilic SN38 prodrug loaded into liposomal nanoparticles, demonstrating enhanced antitumor activity compared to standard irinotecan treatment. Additionally, a polymeric micelle formulation of SN-38 called NK012 was evaluated in patients with metastatic colorectal cancer, showing efficacy with manageable adverse effects. These studies highlight the potential of SN-38 in different formulations and doses for treating colorectal cancer, emphasizing the importance of personalized treatment approaches.
Do Live/Dead assay needs to be performed at 37 C?
5 answers
The Live/Dead assay, commonly used for assessing acute cell death, does not necessarily need to be performed at 37°C, as indicated by various studies. While some assays like the HCV cell-to-cell transmission assay are conducted at 37°C, others like the plasma renin activity assay are carried out at this temperature for accurate results. The Live/Dead assay itself involves loading cells with CalceinAM and propidium iodide to distinguish live and dead cells, with exposure to toxic substances like ZIP for evaluation. Optimal conditions for enzyme activity studies have also been established at 37°C. Therefore, the temperature for performing the Live/Dead assay can vary depending on the specific experimental requirements and protocols outlined in the respective studies.
Which amino acids are antioxidative and antiinflammatory?
5 answers
Tryptophan and phenylalanine derived compounds with electron-donating groups (OH and OCH3) exhibit excellent antioxidant activity, while those with electron-withdrawing groups (Cl, NO2, and F) show potent anti-inflammatory properties. Additionally, selenium-containing amino acids, selenocysteine, and selenocystine, have been found to possess antioxidative and anti-inflammatory properties, effectively reducing oxidative stress and inflammation in a mouse model of inflammatory bowel disease (IBD). Moreover, essential amino acid α-keto acid analogs have demonstrated nephroprotective effects against ischemia-reperfusion injury, decreasing proinflammatory cytokines and malondialdehyde levels, thus exhibiting antioxidative and anti-inflammatory actions. Therefore, these amino acids, including tryptophan, phenylalanine, selenocysteine, and selenocystine, play crucial roles in combating oxidative stress and inflammation.
What are the underlying molecular mechanisms that drive cell metastasis in malignant tumors?
5 answers
Cell metastasis in malignant tumors is primarily driven by a complex interplay of various molecular mechanisms. These mechanisms include the role of tumor-derived exosomes (TDEs) in promoting metastasis through epithelial-mesenchymal transition, immunosuppression, and the formation of a premetastatic niche. Additionally, circulating tumor microemboli (CTM) formed by clustering tumor cells with other cells enhance metastatic properties like improved cell survival, immune evasion, and effective extravasation into distant organs. Genetic aberrations and epigenetic modifications also play a critical role in cancer progression and metastasis, with specific mutations in genes regulating DNA methylation and chromatin modifications contributing to therapy resistance and metastatic disease development. Understanding these molecular mechanisms is crucial for developing effective therapeutic interventions to combat metastasis in malignant tumors.
What are the underlying molecular mechanisms that drive cell metastasis in malignant tumors?
5 answers
The underlying molecular mechanisms that drive cell metastasis in malignant tumors involve a complex interplay of various factors. Tumor-derived exosomes (TDEs) play a critical role in promoting metastasis by carrying DNA, proteins, miRNA, mRNA, lncRNA, and ceRNA, influencing processes like epithelial-mesenchymal transition and immunosuppression. Circulating tumor cells can form circulating tumor microemboli (CTM) through interactions with other cells, enhancing metastatic properties such as improved cell survival and immune evasion. Genetic aberrations and epigenetic modifications also contribute significantly to cancer progression and metastasis, with epigenetic regulation playing a crucial role in poor-prognosis tumors. Understanding these mechanisms is vital for developing effective therapeutic interventions to target metastasis in malignant tumors.