The largest database of trusted experimental protocols
> Chemicals & Drugs > Amino Acid > Dispase II

Dispase II

Dispase II is a proteolytic enzyme used in cell and tissue dissociation procedures.
It is a neutral protease derived from Bacillus polymyxa that effectively cleaves collagen and fibronectin, allowing for the isolation of a variety of cell types from complex tissues.
Researchers can leverage PubCompare.ai to optimize Dispase II protocols through AI-driven comparisons of published literature, preprints, and patents, finding the most reproducible and accurate science-backed solutions for their research needs.
This streamlines the workflow and helps achive reliable, high-quality results.

Most cited protocols related to «Dispase II»

The eyes and lids of mice (n=5 per experiment, in four independent sets of experiments, total of 20 per group, in NS, DS5, and DS10 groups and n=5 per experiment, in two independent sets of experiments in corneal scar and control groups) were excised, pooled, and incubated in 10ml of 5mgml−1 Dispase II (Roche Molecular Biochemicals, Indianapolis, IN) in a shaker at 37 ° C for 1 h, followed by neutralization with Hank ’s Buffered Salt Solution (Invitrogen-Gibco, Grand Island, NY) supplemented with 3% fetal bovine serum (Hyclone, Logan, UT). The bulbar and tarsal conjunctivae were scraped with cytology brushes under a dissecting microscope. Respective superficial CLNs were surgically excised, smashed in between two sterile frosted glass slides, and made into a single-cell suspension. Cell populations were individually collected, centrifuged at 2000r.p.m.×5min, filtered, and resuspended. Cells collected by this technique were used either for flow cytometry (desiccating stress experiment) or for ELISPOT (desiccating stress and corneal ulcer experiment).
Publication 2009
Calnexin Cells Conjunctiva Corneal Ulcer Cytological Techniques dispase II Enzyme-Linked Immunospot Assay Eye Fetal Bovine Serum Flow Cytometry Medulla Oblongata Microscopy Mus Operative Surgical Procedures Population Group Sodium Chloride Sterility, Reproductive
Endometrial/decidual/carcinoma tissues were chopped using scalpels into approximately 0.5 mm3 cubes and enzymatically digested in 20-30 mL 1.25U/mL Dispase II (Sigma, D4693)/ 0.4mg/mL collagenase V (Sigma, C-9263) solution in RPMI 1640 medium (ThermoFisher Scientific, 21875-034)/10% FCS (Biosera, FB-1001) with gentle shaking at 37°C for 30-60 min. The supernatant was passed through one or more 100 μm cell sieves (Corning, 431752) and the sieve washed several times with medium. The flow-through was collected for stromal cell culture in Advanced DMEM/F12 (ThermoFisher Scientific, 12634010) +10%FBS+pen/strep (Sigma, P0781) +L-glutamine (Sigma, 25030-024) for several days and subsequent analysis. The sieves were inverted over a petri dish and retained glandular elements were backwashed from the sieve membranes, pelleted by centrifugation and resuspended in ice cold Matrigel (Corning, 536231) at a ratio of 1:20 (vol:vol). 20 μL drops of Matrigel-cell suspension were plated into 48-well plate (Costar, 3548), allowed to set at 37°C and overlaid with 250 μL organoid Expansion Medium (ExM). See Supplementary Table 3 for ExM composition. The medium was changed every 2-3 d. Cultures were passaged by manual pipetting every 7-10 d. For freezing organoids, Matrigel was removed using Cell Recovery Solution (Corning, 354253) and organoids were resuspended in Recovery cell culture freezing medium (ThermoFisher Scientific, 12648-010). A step-by-step protocol of the derivation and maintenance of human endometrial organoid cultures can be found at Nature Protocol Exchange59 (link).
Publication 2017
Cell Culture Techniques Cells Centrifugation Cold Temperature Collagenase Cuboid Bone Culture Media Decidua dispase II Endometrial Carcinoma Endometrium Glutamine Homo sapiens Hyperostosis, Diffuse Idiopathic Skeletal matrigel Organoids Streptococcal Infections Tissue, Membrane Tissues
All cell cultures were maintained at 37 °C, 5% CO2. hES and iPS cells (HUES3 (control), male; H9 (control), female; HS001 (ALS-SOD1 N139K), male; LWM002 (ALS-SOD1 A4V), female; MBN007 (ALS-SOD1 A4V), female; TM008 (ALS-SOD1 A4V), female; DCM009 (ALS-SOD1 V148G), male; 10013.13 (control), male) were maintained on gelatinized tissue-culture plastic on a monolayer of irradiated CF-1 mouse embryonic fibroblasts (MEFs; GlobalStem), in hESC media, consisting of Dulbecco’s Modified Eagle Medium: nutrient mixture F-12 (DMEM/ F:12, Invitrogen) with 20% Knockout Serum Replacer (KSR; Invitrogen), 110 µM β-mercaptoethanol (BME; Sigma), L-Glutamine and non essential amino acids (NEAA; Invitrogen), and 20 ng/ml basic fibroblast growth factor (bFGF; Invitrogen) (Cowan et al., 2004 (link)). Media was changed every 24 hours and lines were passaged with dispase (Gibco, 1 mg/mL in hES media for 15–30min at 37 °C).
To generate motor neurons, undifferentiated hESCs were passaged using dispase (1 mg/mL) and triturated into small, 50- to 100-cell clumps and placed into ultra-low adherent culture dishes (Corning). For the first three days, cells were kept in suspension in hESC medium, supplemented with 10 µM Rho-associated kinase inhibitor Y27632 (Ascent Scientific) to enhance single cell survival (Watanabe et al., 2007 (link)), 20 ng/mL bFGF (Invitrogen) to enhance growth and 10 µM SB435142 (SB, Sigma) and 0.2 µM LDN193189 (LDN, Stemgent) for neuralization. At day 3, eymbroid bodies (EBs) were switched to neural induction medium (DMEM/F:12 with L-glutamine, NEAA, penicillin/streptomycin, heparin (2 µg/ml), N2 supplement (Invitrogen). At day 5, all-trans retinoic acid (RA; 0.1 or 1 µM, Sigma), ascorbic acid (0.4 µg/ml, Sigma), and BDNF (10 ng/mL, R&D) were added. Dual ALK inhibition (SB+LDN) was pursued until day 7. Hedgehog signaling was initiated on day 7 by application of either C25II modified SHH (R&D), at the standard concentration of 200 ng/ml, a human Smo agonist (HAG, 1 µM, gift from Lee Rubin (Boulting et al., 2011 (link); Dimos et al., 2008 (link))), mouse Smo agonist 1.3 (SAG, 1 µM, (Boulting et al., 2011 (link); Frank-Kamenetsky et al., 2002 (link); Wada et al., 2009 (link); Wichterle et al., 2002 (link))), or purmorphamine (PUR, 1 µM, (Li et al., 2008 (link); Sinha and Chen, 2006 (link)), Stemgent). At day 17, basal medium was changed to Neurobasal (Invitrogen), containing all previous factors and with the addition of 10 ng/mL each of IGF-1, GDNF, and CNTF (R&D), plus B27 (Invitrogen). At day 20 or 30, EBs were dissociated with 0.05% trypsin (Invitrogen), and plated onto poly-lysine/laminin-coated 8-well chamber slides (BD Biosciences) at 0.2–0.5.106 cells/well, and/or 15-mm coverslips at 0.5.106. Plated neurons were cultured in the same medium with the addition of 25 µM BME, and 25 µM glutamic acid (Sigma), and fixed 1 day later.
For immunocytochemistry assays, cultures were fixed for 30 minutes with 4% PFA in phosphate buffered saline (PBS) at 4 °C, washed 3 times for 5 min in PBS, quenched and permeabilized in wash buffer (PBS, 0.1% Triton X-100) plus 50 mM glycine for 15 min. For the EB outgrowth RALDH2 staining, samples were fixed for 10 minutes at room temperature with 4% PFA/10% sucrose pre-warmed to 37°C. Samples were blocked with wash buffer plus 10% normal donkey serum for 1 hr and incubated with primary antibody (Table 1) overnight. Cells were then washed, incubated with DyLight coupled donkey primary anti secondary antibodies (Jackson Immunoresearch, 1:1,000). Finally, cells were washed and counterstained with DAPI (Invitrogen).
Quantitative image analysis of differentiated neuronal cultures was performed using the Multi-Wavelength Cell Scoring module in MetaMorph© software (Molecular Devices). Briefly, EBs were dissociated enzymatically and plated in the presence of neurotrophic factors at densities for which cell overlap was minimal. Following immunostaining, images of at least 9 randomly selected fields (>15,000 cells in total) for each condition were captured using a pre-programmed automated microscope stage. Images were analyzed using the “Multi-Wavelength Cell Scoring” module of the MetaMorph© software, using parameters pre-defined to count only unambiguous bright labeling for each antigen. Intensity thresholds were set while blinded to sample identity, to selectively identify positive cells that displayed unambiguous signal intensity above local background. These parameters were used on all samples in a given experiment, and only minimally adjusted for different staining batches as necessary. Script and Parameter files are available upon request (typically, a cell was ~5,000 grey levels above background to be called positive for any nuclear marker, and was ~10,000 for cytoplasmic markers). A minimum of 15,000 cells per sample was analyzed. All samples were imaged using 10× or 20× objectives on a Zeiss AxioObserver with a Coolsnap HQ2 camera (Photometrics). Some images were acquired using a structured illumination technique using an Apotome module (Zeiss) to achieve 1.9 µm optical sections to ensure co-localization of labeling. For the figures, the brightness and contrast of each channel of an image were adjusted in an appropriate manner to improve clarity.
For Ca2+ imaging experiments utilizing the Hb9::GFP reporter, stem cells were differentiated under the motor neuron differentiation protocol described above, dissociated at day 21 or day 31 and FACS-sorted based on GFP intensity with a 5 laser ARIA-IIu ROU Cell Sorter configured with a 100 µm ceramic nozzle and operating at 20 psi, BD BioSciences. The H9 assays were comprised of mixed neuronal cultures, which a parallel coverslip was stained and quantified to have 53% HB9/ISL1+ motor neurons. All cultures were plated onto 15–25 mm diameter coverslips at a density of 100,000–150,000 cells per coverslip in day 17+ neurobasal media with factors described above with the addition of 0.5 µM EdU, and matured 6 days prior to Ca2+ imaging. Cells were loaded with 3 μM Fluo-4 AM (Invitrogen, Carlsbad, CA) dissolved in 0.2% dimethyl sulfoxide/0.04% pluronic acid (Sigma) in HEPES-buffered physiological salt solution (PSS) for 1 hour at room temperature. PSS contained (mM): NaCl 145, KCl 5, HEPES 10, CaCl2 2, MgCl2 2 and glucose 5.5, pH 7.4. Cultures were continuously superfused with PSS at a rate of approximately 0.5 ml/minute. The cultures were imaged using a 10× objective on an inverted epi-fluorescent Zeiss AxioObserver microscope, equipped with a Coolsnap HQ2 camera (Photometrics). For imaging spontaneous Ca2+ transients, single sets of 200–300 images were acquired at a rate of approximately 2 Hz from each coverslip. For the kainate experiments, 36 images were acquired at a rate of 0.033 Hz and the superfusing PSS was replaced with PSS containing kainate (100 μM) for 60 seconds. Image analysis was performed using ImageJ (http://rsb.info.nih.gov/ij/) or AxioVision 4.7 (Zeiss). Ca2+ transients were determined from regions of interest encompassing the soma of individual cells. A minimum of two cultures obtained from a single differentiation of each cell line and each time point were used for the kainate and all Ca2+ imaging experiments.
For whole cell patch clamp recordings, S+P differentiated HUES3 Hb9::GFP cells were plated on polyornithine/laminin-coated 25 mm diameter coverglass at density of 50,000 per coverslip and cultured for 7 days in the presence of 0.5 µM EdU prior to recording (i.e. DIV 21+7). Current clamp recordings were carried out using an Axopatch 2B amplifier. Data were digitized using a Digidata 1322A digital to analogue converter and were recorded at a 10 KHz sample rate using pClamp 10 software (all equipment from Molecular Devices). Patch pipettes were fabricated using a P-97 pipette puller (Sutter Instruments). The external recording solution contained (in mM), 145 NaCl, 5 KCl, 10 HEPES, 10 glucose, 2 CaCl2, 2 MgCl2. The pH was adjusted to 7.3 using NaOH and the osmolality adjusted to 325 mOsm with sucrose. The pipette solution contained (in mM): 130 CH3KO3S, 10 CH3NaO3S, 1 CaCl2, 10 EGTA, 10 HEPES, 5 MgATP, 0.5 Na2GTP, pH 7.3, 305 mOsm. Experiments were carried out at room temperature (21 – 23 °C). During recordings, current was injected to hold the cells at approximately −60 mV. Action potentials were evoked using incrementally increasing current steps 1 s in duration. The maximum amplitude of the current step (20 – 50 pA) and the size of the increment was calculated based on the input resistance of the cell.
To perform xenotransplantations day 21 EBs from HUES3 Hb9::GFP under the ventralization with SAG+PUR were collected and placed into L-15 media (Invitrogen) containing penicillin/streptomycin (GIBCO). Transplantation was performed as previously described (Wichterle et al., 2002 (link)). Briefly, after a small suction lesion at the prospective intraspinal site was created in a chick embryo at stage 15–18 at somites 15–20, lightly triturated EBs were loaded into a handheld micro-injector. The EBs was placed into the lesion. After 48 hours, the chicks were sacrificed, fixed with 4% PFA for 2 hours at 4°C, and neurite outgrowth and cell body placement was accessed by cutting 200 µm vibratome sections (n = 2), and by cutting 30 µm sections along the spinal cord (n = 5).
Human fetal spinal cords were collected in accordance with the national guidelines of the United States (NIH, FDA) and the State of New York and under Columbia University institutionally approved ethical guidelines relating to anonymous tissue. The fetal material was obtained after elective abortions, and was classified on the basis of external morphology according to the Carnegie stages. Gestational age was determined by last menstrual period of the patient or by ultrasound, if the ultrasound estimate differed by more than one week as indicated by the obstetrician. The spinal cord was removed as intact as possible prior to fixation with fresh, cold 4% PFA for 1.5 hours on ice. Post fixation, the cord was measured and cut into 3 anatomical sections to accommodate embedding in OCT Compound (Tissue-Tek, Redding, CA) and stored at −80 °C prior to cutting on a microtome. 12µm sections were cut along the full length of the cord, taking care to have all 3 sections on each slide in 7 independent sections. This allowed for full analysis and internal staining controls since each slide had cervical, brachial, thoracic and lumbar sections that clearly showed staining within the various motor columns present at different rostal-caudal levels of the spinal cord.
cDNA was obtained from 50,000 FACS purified MN’s from either day 21 S+P (methods described above), or from RA/SHH MN’s at day 31. cDNA preparation was carried out using commercially available kits following the manufacturer’s instructions: RNA isolation (Trizol LS; Invitrogen), cDNA by Brilliant II SYBR green (Stratagene) without amplification. All samples were processed in parallel on the same qPCR plate.
primers:ForwardReverse
RALDH2TTTTGCTGATGCTGACTTGGGCAGCACTGACCTTGATTGA
FOXP1TGACCTTTTGAGGTGACTATAACTGTGGCTGAACCGTTACTTTTTG
LHX3GTTCAGGAGGGGCAGGACCCCAAGCTCCCGTAGAGG
CHT1AAGCCATCATAGTTGGTGGCCGAGCCAAGCTAGGCCATAACCTGGTAC
HOXA5CAGCACCCACATCACGGAGAGGCAAAGA
HOXC6CCAGGACCAGAAAGCCAGTAGTTAGGTAGCGATTGAAGTGAAA
HOXC8CTTCGCTGTTTGATTTCTATTCTGTACGCTGGAGGTTTCTTTCTTT
HOXD9TCGCTGAAGGAGGAGGAGACAAACACCCACAAAGGAAAAC
STD qPCR amplification: 95°- 30”, 55°-60”, 72°-45”
For paired-end RNA-Seq experiments, 400 ng of total RNA was prepared after FACS purification of 500,000 GFP+ or GFP cells. The RNA samples were then amplified using a NuGEN RNA kit for genomic sample amplification, and sequenced to a depth of 21 (S+P) and 35 (SHH) million paired-end reads on an Illumina HiSeq instrument at the HudsonAlpha Institute of Biotechnology. The reads were aligned to the reference transcriptome as well as a library of exon junctions using Bowtie (Version 1) (Langmead et al., 2009 (link)). Data was analyzed using Expression Plot (Friedman and Maniatis, 2011 (link)) using a P value of 0.001 and a 2 fold change threshold. Gene ontology was performed using DAVID (Huang et al., 2008 , 2009 (link)) with enrichment sets from Expression Plot. The RNA-seq data is available in the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE41795.
All quantitative data was analyzed using Sigma Plot 11 or Microsoft Excel. Sample groups were subject to Student’s t-test or where appropriate a One-Way ANOVA with Holm-Sidak post hoc pair-wise comparisons was performed. All experimental data passed an equal variance and normality test (Shapiro-Wilk).
Publication 2013
Mouse hearts were isolated for single cell preparation as previously described9 (link) with atria and valves removed. Isolated mouse hearts were digested using one of three protocols (designated Protocol 1, 2 and 3). For Protocol 1, each mouse heart was divided in approximately 20 pieces, placed in 10 ml of Protocol 1 digestion buffer [2 mg/ml collagenase type II in 1× HBSS (Worthington Biochemical Corporation)] in gentleMacs C-tubes (Miltenyi Biotec), dissociated using Heart 1 program of a gentleMacs Dissociator, and incubated at 37° C for 40 min with gentle agitation. Following incubation, tissue was further dissociated using Heart 2 program before placing on ice. For Protocol 2, isolated hearts were finely minced using forceps to ~2 mm pieces and placed in 3 ml of Protocol 2 digestion buffer [2 mg/ml collagenase type IV (Worthington Biochemical Corporation) and 1.2 U/ml dispase II (Sigma-Aldrich or Thermofisher Scientific) in Dulbecco's phosphate-buffered saline (DPBS) supplemented with 0.9 mM CaCl2]. Tissue was incubated at 37° C for 15 min with gentle rocking. Following incubation, tissue digestion buffer with tissue clusters was triturated by pipetting 12 times using a 10 ml serological pipette. Dishes were again incubated at 37° C and triturated twice more (45 min of total digestion time). The final trituration was conducted by pipetting 30 times with a p1000 pipette. For Protocol 3, isolated mouse hearts were finely minced using forceps, placed in 10 ml Protocol 3 digestion buffer [125 U/ml collagenase type XI (Sigma-Aldrich), 60 U/ml hyaluronidase type I-s (Sigma-Aldrich), and 60 U/ml DNase 1 (Sigma-Aldrich) in DPBS supplemented with 0.9 mM CaCl2 and 20 mM HEPES] incubated at 37° C for 1 hour with gentle agitation, triturated 20 times using a 10 ml serological pipette, and placed on ice. All cell suspensions were filtered using a 40 µm cell strainer. Filtered suspensions were placed into 50 ml tubes with 40 ml of DPBS and centrifuged at 200 g for 20 min with centrifuge brakes deactivated to remove small tissue debris. Cell pellets were resuspended in 250 µl 2% FBS/HBSS solution before staining with various antibodies and reagents for flow cytometry or FACS.
Publication 2015
Antibodies Cells Collagenase Deoxyribonucleases Digestion dispase II Flow Cytometry Forceps Heart Heart Atrium HEPES Hyaluronidase Hyperostosis, Diffuse Idiopathic Skeletal Mucolipidosis Type IV Mus Pellets, Drug Pepsin 3 Phosphates Saline Solution Tissues Type II Mucolipidosis
Skeletal muscle from both hindlimbs was carefully dissected and then gently torn with tissue forceps until homogeneous. Collagenase type 2 (Sigma; 2ml of 2.5 U ml−1), in 10 mM CaCl2, was added to every two hindlimbs, and the preparation was placed at 37 C for 30 min. After washing, a second enzymatic digestion was performed with Collagenase D (Roche Biochemicals; 1.5 U ml−1) and Dispase II (Roche Biochemicals; 2.4 U ml−1), in a total volume of 1 ml per mouse, at 37 C for 60 min. Preparations were passed through a 40-μm cell strainer (Becton Dickenson), and washed. Resulting single cells were collected by centrifugation at 400g for 5 min.
Publication 2010
Cells Centrifugation Collagenase Digestion dispase II Enzymes Forceps Hindlimb Laceration Mus Neutrophil Collagenase Skeletal Muscles Tissues

Most recents protocols related to «Dispase II»

Freshly excised skin tissues were immersed in 0.25% dispase II (Sigma Aldrich, USA) overnight at 4 °C. Cells were washed with Hank's balanced salt solution (HBSS) and immersed in 0.25% dispase II for 5 min at 37 °C. Dispase solution was then replaced with HBSS. The epidermal tissues were carefully separated. The keratinocytes were carefully scraped together. Mechanical stress was applied by an electric pipette (Eppendorf, Germany) of 1 mL pipetting up and down for 5 times. The fragments of epidermis tissues and keratinocyte monolayers were counted.
Publication 2024
For the dataset GSE181276, the pancreatic tissues were digested with the collagenase IV-dispase II (2 mg/ml) at 37 °C for 45 min and further treated with 0.25% trypsin solution at 37 °C for 5 min to obtain single-cell suspension [5 (link)]. For the dataset GSE188819, the pancreatic tissues were incubated with the mixture of digestive enzymes (1 mg/ml collagenase P, 2 U/ml dispase II, 0.1 mg/ml soybean trypsin inhibitor and 0.1 mg/ml DNase I in HBSS with Ca2+/Mg2+) and digested by a gentle MACS™ Octo Dissociator at 37 °C for 40 min, followed by further treatment with 0.05% Trypsin–EDTA at 37 °C for 5 min and removal of red blood cells [6 (link)].
Full text: Click here
Publication 2024
Small pieces of JE tissue were digested in 3 steps. First, they were incubated with dispase II (4 mg/ml) at 4°C overnight. Next, they were incubated with trypsin/EDTA (0.25%, 0.1%) for 2 × 15 min at 37°C, and then with collagenase I/dispase II (3 mg/ml, 4 mg/ml) for 2 × 15 min at 37°C. The digestion was stopped by adding defined K-SFM containing 10% FBS, followed by filtration using a 100-μm mesh cell strainer, and centrifugation. The resulting pellet was mixed in complete defined K-SFM to form a single cell suspension. To culture the generated primary JECs, cells were seeded at a concentration of 105 cells/ml in FBS-precoated 6-well plates.
Publication 2024
RPE cells were isolated according to the method reported by Shang et al. [24 (link)], with some modifications. The enucleated eyes were digested for 35 min at 37 °C in dispase II solution (2 % dispase II [Thermo Fisher Scientific], 10 mM HEPES [pH 7.4], 30 mM NaCl in DMEM [4.5 g/L glucose]). Digested eyeballs were washed twice with growth medium (DMEM [4.5 g/L glucose] containing 10 % FBS, 100 units/mL penicillin, 100 μg/mL streptomycin, and 2.5 mM l-glutamine). An incision was made near the ora serrata of the eyes and the anterior segment of the eye was removed. The remaining posterior eye was transferred to a new growth medium, and the neurosensory retina (NSR) was removed. The RPE–choroid complex was transferred to a new growth medium and RPE cell sheets were peeled from the choroid. Isolated RPE cells were washed twice with growth medium and centrifuged (800×g, 4 °C, 5 min).
Full text: Click here
Publication 2024
The separation process was performed as described previously [45 (link)]. Briefly, dorsal skin samples were collected and immersed into an isolation buffer, 2 mg/ml dispase II (Sigma-Aldrich, St. Louis, MO, USA), overnight at 4 °C. We separated the epidermis from the dermis gently the next day.
Full text: Click here
Publication 2024

Top products related to «Dispase II»

Sourced in United States, Germany, Switzerland, Japan, United Kingdom, China, Austria, Israel, Canada, France
Dispase II is a highly purified protease enzyme that can be used for the dissociation of a variety of cell types, including epithelial, endothelial, and neural cells. It is an effective tool for the isolation and culture of primary cells from tissues.
Sourced in United States, Germany, United Kingdom, France, Switzerland, Israel, Canada
Dispase II is a proteolytic enzyme that can be used for the dissociation and isolation of cells from a variety of tissues. It is a neutral metalloprotease that effectively disperses cells while maintaining their viability and functionality.
Sourced in United States, Switzerland, France, United Kingdom, Japan, China
Dispase II is a proteolytic enzyme derived from Bacillus polymyxa. It is commonly used for the dissociation of a variety of cell and tissue types, including epithelial, endothelial, and fibroblast cells, to facilitate cell isolation and tissue disaggregation.
Sourced in United States, Germany, Switzerland, United Kingdom, Italy, Japan, Macao, Canada, Sao Tome and Principe, China, France, Australia, Spain, Belgium, Netherlands, Israel, Sweden, India
DNase I is a laboratory enzyme that functions to degrade DNA molecules. It catalyzes the hydrolytic cleavage of phosphodiester linkages in the DNA backbone, effectively breaking down DNA strands.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in Switzerland, United States, Germany, United Kingdom, France, Japan, Canada, Australia, Ireland
Collagenase D is an enzyme solution used for the dissociation and isolation of cells from various tissues. It is a mixture of proteolytic enzymes that cleave the collagen present in the extracellular matrix, allowing for the release of individual cells.
Sourced in United States, United Kingdom, Germany, Japan, Switzerland, France, Canada, Italy, China, Belgium
Dispase is an enzymatic cell dissociation reagent used for the isolation and dissociation of cells from various tissue types, including epithelial, endothelial, and connective tissues. It functions by breaking down extracellular matrix proteins, allowing for the efficient release of cells from their surrounding matrix.
Sourced in United States, Switzerland, Germany, Japan, United Kingdom, France, Canada, Italy, Macao, China, Australia, Belgium, Israel, Sweden, Spain, Austria
DNase I is a lab equipment product that serves as an enzyme used for cleaving DNA molecules. It functions by catalyzing the hydrolytic cleavage of phosphodiester bonds in the DNA backbone, effectively breaking down DNA strands.
Sourced in United States, Germany, United Kingdom, China, Canada, France, Japan, Australia, Switzerland, Israel, Italy, Belgium, Austria, Spain, Gabon, Ireland, New Zealand, Sweden, Netherlands, Denmark, Brazil, Macao, India, Singapore, Poland, Argentina, Cameroon, Uruguay, Morocco, Panama, Colombia, Holy See (Vatican City State), Hungary, Norway, Portugal, Mexico, Thailand, Palestine, State of, Finland, Moldova, Republic of, Jamaica, Czechia
Penicillin/streptomycin is a commonly used antibiotic solution for cell culture applications. It contains a combination of penicillin and streptomycin, which are broad-spectrum antibiotics that inhibit the growth of both Gram-positive and Gram-negative bacteria.
Sourced in United States, China, United Kingdom, Germany, France, Australia, Canada, Japan, Italy, Switzerland, Belgium, Austria, Spain, Israel, New Zealand, Ireland, Denmark, India, Poland, Sweden, Argentina, Netherlands, Brazil, Macao, Singapore, Sao Tome and Principe, Cameroon, Hong Kong, Portugal, Morocco, Hungary, Finland, Puerto Rico, Holy See (Vatican City State), Gabon, Bulgaria, Norway, Jamaica
DMEM (Dulbecco's Modified Eagle's Medium) is a cell culture medium formulated to support the growth and maintenance of a variety of cell types, including mammalian cells. It provides essential nutrients, amino acids, vitamins, and other components necessary for cell proliferation and survival in an in vitro environment.

More about "Dispase II"

Dispase II is a powerful proteolytic enzyme used extensively in cell and tissue dissociation procedures.
Derived from the bacterium Bacillus polymyxa, this neutral protease effectively cleaves collagen and fibronectin, allowing researchers to isolate a wide variety of cell types from complex tissues.
Harnessing the power of AI-driven comparisons, PubCompare.ai helps optimize Dispase II protocols by analyzing published literature, preprints, and patents, enabling researchers to identify the most reproducible and accurate science-backed solutions for their needs.
This streamlines the workflow and helps achieve reliable, high-quality results.
In addition to Dispase II, other enzymes like DNase I and Collagenase D are often utilized in cell isolation and dissociation processes.
Supplemented with fetal bovine serum (FBS) and antibiotics like penicillin/streptomycin, these reagents are typically combined with Dulbecco's Modified Eagle Medium (DMEM) to create a complete cell culture environment.
By leveraging the insights and comparisons provided by PubCompare.ai, researchers can fine-tune their Dispase II protocols, optimizing the use of these complementary reagents for maximum efficiency and reproducibility.
This streamlined approach helps drive scientific progress and unlock new discoveries in the field of cell biology and tissue engineering.