scispace - formally typeset
Search or ask a question

Why DMSO toxic to cells? 


Best insight from top research papers

Dimethyl sulfoxide (DMSO) exhibits toxicity to cells due to various reasons outlined in the provided contexts. Studies have shown that high concentrations of DMSO induce cell death mechanisms, including cleavage of caspase-3 and PARP-1 . Furthermore, DMSO has been associated with significant clinical side effects in patients receiving cell-therapy products, affecting cardiovascular, neurological, gastrointestinal, and allergic systems . The toxicity of DMSO is evident in its impact on gene expression, microRNA deregulation, and epigenetic changes in tissues like cardiac and hepatic microtissues . Despite being widely used, DMSO can cause adverse reactions such as nausea, chills, cardiac arrhythmias, and neurological symptoms, leading to concerns about its safety and cytotoxicity . Therefore, the toxic effects of DMSO on cells are multifaceted, ranging from molecular disruptions to clinical manifestations.

Answers from top 4 papers

More filters
Papers (4)Insight
DMSO induces significant changes in cellular processes and the epigenetic landscape, impacting gene expression, microRNAs, and DNA methylation, suggesting it is not inert and potentially toxic to cells.
DMSO is toxic to cells at concentrations above 0.40%, causing inhibition of proliferation and increased cell death in HepG2 and Huh-7 cell lines.
DMSO is toxic to cells due to its broad toxicities causing clinical side effects and altering NK and T-cell markers and function, hindering the development of DMSO-free cryopreservation methods.
DMSO is toxic to cells due to inducing caspase-3 and PARP-1 cleavage, leading to cell death mechanisms even at low concentrations, as shown in RA FLSs studies.

Related Questions

What is the cytotoxic effect of DMSO?4 answersDMSO has been shown to have cytotoxic effects in various studies. It has been found to reduce the toxicity of the anticancer ruthenium complex KP1019, leading to a decrease in its bioactivity. Additionally, DMSO has been shown to induce cytotoxicity in preimplantation embryos, leading to developmental defects and decreased implantation rates. Furthermore, DMSO has been found to have cytotoxic effects on human leukemic T cells, causing a decrease in viability and proliferative response. These studies suggest that DMSO can have cytotoxic effects on different cell types and can impact the effectiveness of certain drugs.
What are the applications of dimethyl sulfoxide (DMSO)?5 answersDimethyl sulfoxide (DMSO) has various applications. It is commonly used as a solvent due to its low toxicity, affordability, and accessibility. DMSO is also utilized as a reagent for the synthesis of functionalized molecules, as it can serve as a carbon source, sulfur source, and oxygen source. Additionally, DMSO has been used for cryopreservation of cells and tissues, as it can penetrate cell membranes and prevent freeze-thaw injuries. Recent studies have shown that DMSO possesses immunomodulatory effects, including immune enhancement and anti-inflammatory effects, making it a potential therapeutic option for cancer, autoimmune diseases, and chronic inflammatory diseases. Furthermore, DMSO has been investigated for its preventive effects against radiation-induced nervous system diseases in individuals receiving radiotherapy for head and neck cancers.
What are the effects of DMSO2 on the human body?5 answersDMSO2, also known as dimethyl sulfoxide, is not mentioned in any of the provided abstracts. Therefore, there is no information available on the effects of DMSO2 on the human body.
What are the mechanisms of action of DMSO?5 answersDimethyl sulfoxide (DMSO) has multiple mechanisms of action. It can induce cell fusion, cell differentiation, and enhance the permeability of lipid membranes. DMSO also acts as a cryoprotectant, protecting against ischemic injury and enhancing resistance to infection. In terms of heme synthesis, DMSO stimulates the synthesis of hemoglobin in erythroid cells. Additionally, DMSO can modulate membrane structure and function by inducing water pores in lipid bilayers, which enhances the penetration of active molecules through the membrane. The presence of DMSO in lipid membranes also makes them more flexible, facilitating membrane fusion and enabling the membrane to withstand osmotic and mechanical stresses during cryopreservation. These findings provide insights into the various ways in which DMSO can affect biological systems and suggest its potential applications in fields such as cell therapy and drug delivery.
What role of DMSO in MTT assay?3 answersDMSO is used as a solvent in the MTT assay to dissolve the insoluble formazan crystals. The MTT assay is based on the conversion of MTT into formazan crystals by living cells, which determines the mitochondrial activity of these cells. The formazan crystals are insoluble in water, so DMSO is used to solubilize them and create a colored solution that can be quantified. The use of DMSO as a solvent allows for accurate measurement of the optical densities directly on the MultiScreen plate, improving the reproducibility and accuracy of the assay. DMSO has the same optical refraction index as the filters used in the assay, making it possible to measure the absorbance without loss of cells or formazan crystals.
What is DMSO solvent?5 answersDMSO (dimethyl sulfoxide) is a polar aprotic solvent that is commonly used in various applications. It has the ability to solubilize both polar and nonpolar molecules that are otherwise poorly soluble in other solvents. DMSO is known for its extensive miscibility with water and most common organic solvents, making it a versatile solvent in chemical reactions and organic synthesis. It has a high dipole moment and dielectric constant, which contribute to its solubilizing properties. DMSO is also used as a co-solvent in systems containing guanine-rich DNA, where it can affect the stability of G-quadruplex structures. However, it is important to note that DMSO can have toxic effects at higher concentrations, causing cellular toxicity and apoptosis-inducing factor translocation in neuronal cells. Researchers should consider the concentration of DMSO and its potential toxicity when using it as a solvent in biological assays or in vivo administration.

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.
Volume of DACC that should be added to wound dressings?
5 answers
The optimal volume of Dialkylcarbamoyl chloride (DACC) to be added to wound dressings is not explicitly mentioned in the provided contexts. However, studies highlight the effectiveness of DACC-coated dressings in managing wound infections by irreversibly binding and removing bacteria. DACC technology offers a passive control of bacterial load without releasing antimicrobial agents, reducing the risk of resistance and allergies. Research confirms the high antibacterial activity of DACC-coated dressings against various pathogens, even at increased inoculum densities, and in the presence of higher protein loads. The dressing's ability to bind bacteria and limit biofilm formation while maintaining bacterial cell integrity suggests its potential as a beneficial wound care option. Further research may provide insights into the ideal DACC volume for optimal wound healing outcomes.
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.
What are the drawbacks of using cryoprotectants for sperm cryopreservation?
5 answers
The drawbacks of using cryoprotectants for sperm cryopreservation include potential cell damage, reduced motility, and negative effects on embryo quality. Cryoprotectants can lead to cell injury, affecting embryo quality. Additionally, high concentrations of non-penetrative cryoprotectants may cause excessive cell shrinkage and solute effects, potentially damaging cells. In donkey semen cryopreservation, the absence of permeable cryoprotectants resulted in reduced motility, vitality, and membrane integrity of sperm. Furthermore, common contraindications related to cryopreservation in bovine spermatozoa include the presence of ice crystals, oxidative stress, and possible toxicity of cryoprotectants, all impacting sperm quality and fertility potential. These findings highlight the importance of optimizing cryoprotectant formulations to minimize adverse effects during sperm cryopreservation.
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.
What activates myfibroblasten?
5 answers
Myofibroblasts are activated by various stimuli, leading to their secretory and contractile phenotype. Activation traditionally involved the de novo expression of alpha-smooth muscle actin stress fibers, previously viewed as a binary process. Recent studies have shifted towards considering myofibroblast activation as a continuous spectrum, necessitating a method to quantify this activation level. Mechanisms of myofibroblast activation include responses to changes in the cytokine milieu and extracellular matrix composition, leading to dynamic phenotypic alterations. Additionally, compounds like cyclolanostane and rophenol have been identified as fibroblast activators, promoting the production of collagen and other extracellular matrix components. These findings collectively highlight the multifaceted nature of myofibroblast activation and the diverse pathways involved in their stimulation.
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.
How does macromolecular crowding affect ECM deposition in cells?
5 answers
Macromolecular crowding (MMC) impacts extracellular matrix (ECM) deposition in cells by mimicking in vivo macromolecule concentrations. Studies using MMC with Ficoll show enhanced deposition of ECM proteins like collagen, fibronectin, tenascin C, and basement membrane molecules. For instance, MMC promotes fibrillin-1 deposition by lung fibroblasts, crucial for elastic fiber formation and ECM regulation. In neural cultures, MMC induces native ECM deposition by astrocytes, leading to improved neuronal growth, synaptic contacts, and pharmacological responses, making the cultures more physiologically relevant. Overall, MMC creates a high macromolecule environment that influences ECM composition, potentially offering novel insights for regenerative therapies and tissue engineering.
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.