Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Drug Resist Updat ; 53: 100730, 2020 12.
Article in English | MEDLINE | ID: mdl-33096284

ABSTRACT

New treatment options of acute myeloid leukemia (AML) are rapidly emerging. Pre-clinical models such as ex vivo cultures are extensively used towards the development of novel drugs and to study synergistic drug combinations, as well as to discover biomarkers for both drug response and anti-cancer drug resistance. Although these approaches empower efficient investigation of multiple drugs in a multitude of primary AML samples, their translational value and reproducibility are hampered by the lack of standardized methodologies and by culture system-specific behavior of AML cells and chemotherapeutic drugs. Moreover, distinct research questions require specific methods which rely on specific technical knowledge and skills. To address these aspects, we herein review commonly used culture techniques in light of diverse research questions. In addition, culture-dependent effects on drug resistance towards commonly used drugs in the treatment of AML are summarized including several pitfalls that may arise because of culture technique artifacts. The primary aim of the current review is to provide practical guidelines for ex vivo primary AML culture experimental design.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Primary Cell Culture/methods , Research Design/standards , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Cryopreservation , Culture Media/chemistry , Culture Media/standards , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Drug Screening Assays, Antitumor/standards , Guidelines as Topic , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Primary Cell Culture/instrumentation , Primary Cell Culture/standards , Reproducibility of Results , Tumor Cells, Cultured
2.
Am J Physiol Heart Circ Physiol ; 319(5): H1112-H1122, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32986966

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enable cardiotoxicity testing and personalized medicine. However, their maturity is of concern, including relatively depolarized resting membrane potential and more spontaneous activity compared with adult cardiomyocytes, implicating low or lacking inward rectifier potassium current (Ik1). Here, protein quantification confirms Kir2.1 expression in hiPSC-CM syncytia, albeit several times lower than in adult heart tissue. We find that hiPSC-CM culture density influences Kir2.1 expression at the mRNA level (potassium inwardly rectifying channel subfamily J member 2) and at the protein level and its associated electrophysiology phenotype. Namely, all-optical cardiac electrophysiology and pharmacological treatments reveal reduction of spontaneous and irregular activity and increase in action potential upstroke in denser cultures. Blocking Ik1-like currents with BaCl2 increased spontaneous frequency and blunted action potential upstrokes during pacing in a dose-dependent manner only in the highest-density cultures, in line with Ik1's role in regulating the resting membrane potential. Our results emphasize the importance of syncytial growth of hiPSC-CMs for more physiologically relevant phenotype and the power of all-optical electrophysiology to study cardiomyocytes in their multicellular setting.NEW & NOTEWORTHY We identify cell culture density and cell-cell contact as an important factor in determining the expression of a key ion channel at the transcriptional and the protein levels, KCNJ2/Kir2.1, and its contribution to the electrophysiology of human induced pluripotent stem cell-derived cardiomyocytes. Our results indicate that studies on isolated cells, out of tissue context, may underestimate the cellular ion channel properties being characterized.


Subject(s)
Giant Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Action Potentials , Animals , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques/methods , Cellular Reprogramming Techniques/standards , Female , Giant Cells/cytology , Giant Cells/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Primary Cell Culture/methods , Primary Cell Culture/standards , Rats
3.
Transfusion ; 60(5): 1004-1014, 2020 05.
Article in English | MEDLINE | ID: mdl-32167176

ABSTRACT

BACKGROUND: Most chimeric antigen receptor T (CAR-T) cells and other adoptive T-cell therapies (ACTs) are currently manufactured by ex vivo expansion of patient lymphocytes in culture media supplemented with human plasma from group AB donors. As lymphocytes do not express A or B antigens, the isoagglutinins of non-AB plasmas are unlikely to cause deleterious effects on lymphocytes in culture. STUDY DESIGN AND METHODS: Seeding cultures with peripheral blood mononuclear cell (PBMNC) concentrates from group A1 donors and using a CAR-T culture protocol, parallel cultures were performed, each with unique donor plasmas as media supplements (including group O plasmas with high-titer anti-A and group AB plasmas as control). An additional variable, a 3% group A1 red blood cell (RBC) spike, was added to simulate a RBC-contaminated PBMNC collection. Cultures were monitored by cell count, viability, flow cytometric phenotype, gene expression analysis, and supernatant chemokine analysis. RESULTS: There was no difference in lymphocyte expansion or phenotype when cultured with AB plasma or O plasma with high-titer anti-A. Compared to controls, the presence of contaminating RBCs in lymphocyte culture led to poor lymphocyte expansion and a less desirable phenotype-irrespective of the isoagglutinin titer of the plasma supplement used. CONCLUSIONS: This study suggests that ABO incompatible plasma may be used as a media supplement when culturing cell types that do not express ABO antigens-such as lymphocytes for CAR-T or other ACT. The presence of contaminating RBCs in culture was disadvantageous independent of isoagglutinin titer.


Subject(s)
ABO Blood-Group System/blood , Culture Media/chemistry , Immunotherapy, Adoptive , Plasma/physiology , Primary Cell Culture/methods , T-Lymphocytes/cytology , Cells, Cultured , Flow Cytometry , HLA-A Antigens/blood , HLA-A Antigens/immunology , Humans , Immunotherapy, Adoptive/methods , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Lymphocyte Activation , Plasma/chemistry , Primary Cell Culture/standards , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Tissue Donors
4.
Transfusion ; 60(5): 986-996, 2020 05.
Article in English | MEDLINE | ID: mdl-32181889

ABSTRACT

BACKGROUND: Platelets have the highest bacterial contamination risk of all blood components, and septic transfusion reactions remain a problem. A good estimate of contamination rates could provide information about residual risk and inform optimal testing strategies. We performed a systematic review and meta-analysis of platelet contamination rates by primary culture. STUDY DESIGN AND METHODS: A literature search in December 2019 identified articles on platelet contamination rates using primary culture. We used meta-analysis to estimate the overall rate of contamination and meta-regression to identify heterogeneity. We studied the following sources of heterogeneity: collection method, sample volume, positivity criteria, and study date. Contamination rate estimates were obtained for apheresis (AP), platelet rich plasma (PRP), and buffy coat (BC) collection methods. RESULTS: The search identified 6102 studies, and 22 were included for meta-analysis. Among these 22 studies, there were 21 AP cohorts (4,072,022 components), 4 PRP cohorts (138,869 components), and 15 BC cohorts (1,474,679 components). The overall mean contamination rate per 1000 components was 0.51 (95% CI: 0.38-0.67) including AP (0.23, 95% CI: 0.18-0.28), PRP, (0.38, 95% CI: 0.15-0.70), and BC (1.12, 95% CI: 0.51-1.96). There was considerable variability within each collection method. Sample volume, positivity criteria, and publication year were significant sources of heterogeneity. CONCLUSION: The bacterial contamination rate of platelets by primary culture is 1 in 1961. AP and PRP components showed a lower contamination rate than BC components. There is clinically significant between-study variability for each method. Larger sample volumes increased sensitivity, and bacterial contamination rates have decreased over time.


Subject(s)
Bacterial Infections/blood , Blood Component Removal/statistics & numerical data , Blood Platelets/microbiology , Drug Contamination/statistics & numerical data , Platelet Transfusion/statistics & numerical data , Primary Cell Culture/statistics & numerical data , Bacterial Infections/epidemiology , Bacterial Infections/transmission , Bacteriological Techniques , Blood Component Removal/adverse effects , Blood Component Transfusion/adverse effects , Blood Component Transfusion/statistics & numerical data , Blood Platelets/cytology , Cells, Cultured , Humans , Platelet Transfusion/adverse effects , Platelet-Rich Plasma/microbiology , Primary Cell Culture/methods , Primary Cell Culture/standards , Transfusion Reaction/epidemiology , Transfusion Reaction/microbiology
5.
Transfusion ; 60(5): 974-985, 2020 05.
Article in English | MEDLINE | ID: mdl-32357261

ABSTRACT

BACKGROUND: The high incidence of septic transfusion reactions (STRs) led to testing being mandated by AABB from 2004. This was implemented by primary culture of single-donor apheresis platelets (APs) from 2004 and prestorage pooled platelets (PSPPs) from 2007. STUDY DESIGN/METHODS: Platelet (PLT) aliquots were cultured at issue and transfusion reactions evaluated at our hospital. Bacterial contamination and STR rates (shown as rates per million transfusions in Results) were evaluated before and after introduction of primary culture by blood centers that used a microbial detection system (BacT/ALERT, bioMerieux) or enhanced bacterial detection system (eBDS, Haemonetics). RESULTS: A total of 28,457 PLTs were cultured during pre-primary culture periods (44.7% APs; 55.3% at-issue pooled PLTs [AIPPs]) and 97,595 during post-primary culture periods (79.3% APs; 20.7% PSPPs). Forty-three contaminated units were identified in preculture and 34 in postculture periods (rates, 1511 vs. 348; p < 0.0001). Contamination rates of APs were significantly lower than AIPPs in the preculture (393 vs. 2415; p < 0.0001) but not postculture period compared to PSPPs (387 vs. 198; p = 0.9). STR rates (79 vs. 90; p = 0.98) were unchanged with APs but decreased considerably with pooled PLTs (826 vs. 50; p = 0.0006). Contamination (299 vs. 324; p = 0.84) and STR rates (25 vs. 116; p = 0.22) were similar for PLTs tested by BacT/ALERT and eBDS primary culture methods. A change in donor skin preparation method in 2012 was associated with decreased contamination and STR rates. CONCLUSION: Primary culture significantly reduced bacterial contamination and STR associated with pooled but not AP PLTs. Measures such as secondary testing near time of use or pathogen reduction are needed to further reduce STRs.


Subject(s)
Bacterial Infections/epidemiology , Drug Contamination/statistics & numerical data , Platelet Transfusion , Primary Cell Culture , Sepsis/epidemiology , Transfusion Reaction/epidemiology , Academic Medical Centers , Adult , Bacterial Infections/blood , Bacterial Infections/transmission , Blood Component Removal/adverse effects , Blood Component Removal/history , Blood Component Removal/standards , Blood Component Removal/statistics & numerical data , Blood Platelets/cytology , Blood Platelets/microbiology , Blood Safety/adverse effects , Blood Safety/history , Blood Safety/statistics & numerical data , Blood Transfusion/history , Blood Transfusion/statistics & numerical data , Cells, Cultured , Child , History, 20th Century , History, 21st Century , Humans , Incidence , Platelet Transfusion/adverse effects , Platelet Transfusion/history , Platelet Transfusion/statistics & numerical data , Primary Cell Culture/history , Primary Cell Culture/standards , Primary Cell Culture/statistics & numerical data , Retrospective Studies , Sepsis/blood , Sepsis/etiology , Transfusion Reaction/microbiology , United States/epidemiology
6.
Transfusion ; 60(5): 997-1002, 2020 05.
Article in English | MEDLINE | ID: mdl-32275069

ABSTRACT

BACKGROUND: Effective and financially viable mitigation approaches are needed to reduce bacterial contamination of platelets in the US. Expected costs of large-volume delayed sampling (LVDS), which would be performed by a blood center prior to shipment to a hospital, were compared to those of pathogen reduction (PR), point-of-release testing (PORt), and secondary bacterial culture (SBC). METHODS: Using a Markov-based decision-tree model, the financial and clinical impact of implementing all variants of LVDS, PR, PORt, and SBC described in FDA guidance were evaluated from a hospital perspective. Hospitals were assumed to acquire leukoreduced apheresis platelets, with LVDS adding $30 per unit. Monte Carlo simulations were run to estimate the direct medical costs for platelet acquisition, testing, transfusion, and possible complications associated with each approach. Input parameters, including test sensitivity and specificity, were drawn from existing literature and costs (2018US$) were based on a hospital perspective. A one-way sensitivity analysis varied the assumed additional cost of LVDS. RESULTS: Under an approach of LVDS (7-day), the total cost per transfused unit is $735.78, which falls between estimates for SBC (7-day) and PORt. Assuming 20,000 transfusions each year, LVDS would cost $14.72 million annually. Per-unit LVDS costs would need to be less than $22.32 to be cheaper per transfusion than all other strategies, less than $32.02 to be cheaper than SBC (7-day), and less than $196.19 to be cheaper than PR (5-day). CONCLUSIONS: LVDS is an effective and cost-competitive approach, assuming additional costs to blood centers and associated charges to hospitals are modest.


Subject(s)
Bacterial Infections/prevention & control , Drug Contamination/prevention & control , Infection Control , Platelet Transfusion/economics , Platelet Transfusion/statistics & numerical data , Plateletpheresis , Primary Cell Culture/economics , Bacterial Infections/economics , Bacterial Infections/epidemiology , Bacterial Infections/transmission , Blood Banks/economics , Blood Banks/standards , Blood Banks/statistics & numerical data , Blood Platelets/microbiology , Blood Safety/economics , Blood Safety/methods , Blood Safety/standards , Blood Specimen Collection/adverse effects , Blood Specimen Collection/economics , Blood Specimen Collection/standards , Blood Specimen Collection/statistics & numerical data , Costs and Cost Analysis , Diagnostic Tests, Routine/economics , Diagnostic Tests, Routine/standards , Diagnostic Tests, Routine/statistics & numerical data , Drug Contamination/economics , Drug Contamination/statistics & numerical data , Feasibility Studies , Humans , Implementation Science , Infection Control/economics , Infection Control/methods , Microbiological Techniques , Plateletpheresis/adverse effects , Plateletpheresis/economics , Plateletpheresis/methods , Plateletpheresis/standards , Primary Cell Culture/methods , Primary Cell Culture/standards , Primary Cell Culture/statistics & numerical data , Risk Reduction Behavior , Sample Size , Time Factors , Time-to-Treatment/economics , Time-to-Treatment/statistics & numerical data , Transfusion Reaction/economics , Transfusion Reaction/epidemiology , Transfusion Reaction/microbiology , Transfusion Reaction/prevention & control
7.
Exp Cell Res ; 362(1): 102-110, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29137914

ABSTRACT

Adult bone marrow mesenchymal stromal cells (MSCs) have cross-functional, intrinsic potency that is of therapeutic interest. Their ability to regenerate bone, fat, and cartilage, modulate the immune system, and nurture the growth and function of other bone marrow hematopoietic stem/progenitor cells have all been evaluated by transplant applications of MSCs. These applications require the isolation and expansion scaled cell production. To investigate biophysical properties of MSCs that can be feasibly utilized as predictors of bioactivity during biomanufacturing, we used a low-density seeding model to drive MSCs into proliferative stress and exhibit the hallmark characteristics of in vitro aging. A low-density seeding method was used to generate MSCs from passages 1-7 to simulate serial expansion of these cells to maximize yield from a single donor. MSCs were subjected to three bioactivity assays in parallel to ascertain whether patterns in MSC age, size, and shape were associated with the outcomes of the potency assays. MSC age was found to be a predictor of adipogenesis, while cell and nuclear shape was strongly associated to hematopoietic-supportive potency. Together, these data evaluate morphological changes associated with cell potency and highlight new strategies for purification or alternatives to assessing MSC quality.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Culture Techniques/methods , Cellular Senescence/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Adipogenesis/physiology , Adult , Bone Marrow/pathology , Cell Culture Techniques/standards , Cell Differentiation , Cell Proliferation , Cell Shape , Cells, Cultured , Cryopreservation , Humans , Primary Cell Culture/methods , Primary Cell Culture/standards
8.
Ecotoxicol Environ Saf ; 173: 63-70, 2019 May 30.
Article in English | MEDLINE | ID: mdl-30769204

ABSTRACT

Chemical contaminants are known to accumulate in marine megafauna globally, but little is known about how this impacts animal health. In vitro assays offer an ethical, reproducible and cost-effective alternative to live animal toxicity testing on large, long-lived or threatened species, such as sea turtles. However, using a cell culture from a single animal raise the question of whether the toxicity observed adequately represents the toxicity in that species. This study examined variation in the cytotoxic response of primary skin fibroblasts established from seven green (Chelonia mydas) and five loggerhead (Caretta caretta) sea turtles. Cell viability using resazurin dye was examined in response to exposure to five contaminants. The variation in cytotoxicity was generally low (within a factor of five) for both independent analyses of the same cell culture, and cell cultures from different individuals. This low within and between cell culture variation indicates that primary sea turtle cell cultures can provide a suitable approach to understanding toxicity in sea turtles. In addition, green and loggerhead turtle cells showed similar toxicity to the compounds tested, indicating that only subtle differences in chemical sensitivity may exist between sea turtle species. This study provides a framework for using species-specific cell cultures in future toxicological studies on sea turtles. Although in vivo studies are the gold standard for toxicological studies and species-specific risk assessments, the development of in vitro tools can provide important information when in vivo studies are not possible or practical. For large, endangered species such as sea turtles that are exposed to, and accumulate, a large number of contaminants, using validated cell cultures may facilitate the rapid assessment of chemical risk to these animals.


Subject(s)
Primary Cell Culture , Toxicity Tests/methods , Turtles/physiology , Animals , Cell Survival/drug effects , Fibroblasts/drug effects , Primary Cell Culture/standards , Skin/drug effects , Skin/pathology , Water Pollutants, Chemical/toxicity
9.
Int J Mol Sci ; 19(10)2018 Oct 07.
Article in English | MEDLINE | ID: mdl-30301262

ABSTRACT

Malignant pleural mesothelioma (MPM) is a deadly cancer that is caused by asbestos exposure and that has limited treatment options. The current standard of MPM diagnosis requires the testing of multiple immunohistochemical (IHC) markers on formalin-fixed paraffin-embedded tissue to differentiate MPM from other lung malignancies. To date, no single biomarker exists for definitive diagnosis of MPM due to the lack of specificity and sensitivity; therefore, there is ongoing research and development in order to identify alternative biomarkers for this purpose. In this study, we utilized primary MPM cell lines and tested the expression of clinically used biomarker panels, including CK8/18, Calretinin, CK 5/6, CD141, HBME-1, WT-1, D2-40, EMA, CEA, TAG72, BG8, CD15, TTF-1, BAP1, and Ber-Ep4. The genomic alteration of CDNK2A and BAP1 is common in MPM and has potential diagnostic value. Changes in CDKN2A and BAP1 genomic expression were confirmed in MPM samples in the current study using Fluorescence In situ Hybridization (FISH) analysis or copy number variation (CNV) analysis with digital droplet PCR (ddPCR). To determine whether MPM tissue and cell lines were comparable in terms of molecular alterations, IHC marker expression was analyzed in both sample types. The percentage of MPM biomarker levels showed variation between original tissue and matched cells established in culture. Genomic deletions of BAP1 and CDKN2A, however, showed consistent levels between the two. The data from this study suggest that genomic deletion analysis may provide more accurate biomarker options for MPM diagnosis.


Subject(s)
Biomarkers, Tumor/standards , Cyclin-Dependent Kinase Inhibitor p18/genetics , Lung Neoplasms/genetics , Mesothelioma/genetics , Primary Cell Culture/standards , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Gene Deletion , Humans , Lung Neoplasms/pathology , Male , Mesothelioma/pathology , Mesothelioma, Malignant , Middle Aged , Primary Cell Culture/methods
10.
Cytotherapy ; 18(10): 1312-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27497700

ABSTRACT

Regulatory T cells (Tregs) play a fundamental role in the maintenance of self-tolerance and immune homeostasis. Defects in Treg function and/or frequencies have been reported in multiple disease models. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder affecting upper and lower motor neurons. Compelling evidence supports a neuroprotective role for Tregs in this disease. Indeed, rapid progression in ALS patients is associated with decreased FoxP3 expression and Treg frequencies. Thus, we propose that strategies to restore Treg number and function may slow disease progression in ALS. In this study, we developed a robust, Good Manufacturing Practice (GMP)-compliant procedure to enrich and expand Tregs from ALS patients. Tregs isolated from these patients were phenotypically similar to those from healthy individuals but were impaired in their ability to suppress T-cell effector function. In vitro expansion of Tregs for 4 weeks in the presence of GMP-grade anti-CD3/CD28 beads, interleukin (IL)-2 and rapamcyin resulted in a 25- to 200-fold increase in their number and restored their immunoregulatory activity. Collectively, our data facilitate and support the implementation of clinical trials of adoptive therapy with ex vivo expanded and highly suppressive Tregs in patients with ALS.


Subject(s)
Adoptive Transfer/standards , Amyotrophic Lateral Sclerosis/pathology , Cell Separation , Cell- and Tissue-Based Therapy/standards , Primary Cell Culture , T-Lymphocytes, Regulatory/pathology , Adoptive Transfer/methods , Amyotrophic Lateral Sclerosis/immunology , Case-Control Studies , Cell Separation/methods , Cell Separation/standards , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Guideline Adherence/standards , Humans , Immune Tolerance , Interleukin-2/metabolism , Primary Cell Culture/methods , Primary Cell Culture/standards , T-Lymphocytes, Regulatory/immunology
11.
Neuropathol Appl Neurobiol ; 39(4): 426-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22845849

ABSTRACT

AIMS: Diffuse intrinsic pontine glioma (DIPG) is a fatal paediatric malignancy. Tumour resection is not possible without serious morbidity and biopsies are rarely performed. The resulting lack of primary DIPG material has made preclinical research practically impossible and has hindered the development of new therapies for this disease. The aim of the current study was to address the lack of primary DIPG material and preclinical models by developing a multi-institutional autopsy protocol. METHODS: An autopsy protocol was implemented in the Netherlands to obtain tumour material within a brief post mortem interval. A team of neuropathologists and researchers was available at any time to perform the autopsy and process the material harvested. Whole brain autopsy was performed and primary DIPG material and healthy tissue were collected from all affected brain areas. Finally, the study included systematic evaluation by parents. RESULTS: Five autopsies were performed. The mean time interval between death and time of autopsy was 3 h (range 2-4). All tumours were graded as glioblastoma. None of the parents regretted their choice to participate, and they all derived comfort in donating tissue of their child in the hope to help future DIPG patients. In addition, we developed and characterized one of the first DIPG cell cultures from post mortem material. CONCLUSION: Here we show that obtaining post mortem DIPG tumour tissue for research purposes is feasible with short delay, and that the autopsy procedure is satisfying for participating parents and can be suitable for the development of preclinical DIPG models.


Subject(s)
Autopsy/standards , Brain Stem Neoplasms/pathology , Glioma/pathology , Primary Cell Culture/standards , Animals , Child , Child, Preschool , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Female , Humans , Infant , Male , Mice , Mice, Nude , Parents , Pons/pathology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
12.
Methods Mol Biol ; 2286: 227-235, 2021.
Article in English | MEDLINE | ID: mdl-32504295

ABSTRACT

Schwann cells as glial cells in the peripheral nervous system can participate in neurons protection and forming myelin. Additionally, they are important for nerve pulse conduction supporting along axons. On the other hand, it was demonstrated that they are promising cells for the treatment of demyelinating disorders and also central nervous system damages. Herein, for therapeutic application, Schwann cells should be manufactured based on good manufacturing practice standards to achieve safe and effective clinical products. In this respect, the current chapter tries to introduce a standard protocol for manufacturing of human GMP-compliant Schwann cells for clinical application.


Subject(s)
Cell Transplantation/standards , Practice Guidelines as Topic , Primary Cell Culture/methods , Schwann Cells/cytology , Tissue and Organ Harvesting/methods , Cell Transplantation/methods , Cells, Cultured , Humans , Primary Cell Culture/standards , Tissue and Organ Harvesting/standards
13.
Methods Mol Biol ; 2286: 121-129, 2021.
Article in English | MEDLINE | ID: mdl-33381853

ABSTRACT

Mesenchymal stem cells (MSCs) have been used in therapies owing to their regenerative potential, paracrine regulatory effects, and immunomodulatory activity. To foster commercialization and implementation of stem cells treatments, researchers have recently derived MSCs from human induced pluripotent stem cells (iMSCs). For therapeutic applications, human iMSCs must be produced in xeno-free culture conditions and following procedures that are compatible with the principles of Good Manufacturing Practice.


Subject(s)
Biomedical Technology/standards , Induced Pluripotent Stem Cells/cytology , Primary Cell Culture/methods , Biomedical Technology/methods , Humans , Practice Guidelines as Topic , Primary Cell Culture/standards
14.
Methods Mol Biol ; 2286: 131-166, 2021.
Article in English | MEDLINE | ID: mdl-33381854

ABSTRACT

Cardiac explant-derived cells (cEDC), also referred as cardiac progenitors cells (CPC) (Barile et al., Cardiovasc Res 103(4):530-541, 2014; Barile et al., Cardiovasc Res 114(7):992-1005, 2018), represent promising candidates for the development of cell-based therapies, a novel and interesting treatment for cardioprotective strategy in heart failure (Kreke et al., Expert Rev Cardiovasc Ther 10(9):1185-1194, 2012). CPC have been tested in a preclinical setting for direct cell transplantation and tissue engineering or as a source for production of extracellular vesicles (EV) (Oh et al., J Cardiol 68(5):361-367, 2016; Barile et al., Eur Heart J 38(18):1372-1379, 2017; Rosen et al., J Am Coll Cardiol 64(9):922-937, 2014). CPC cultured as cardiospheres derived cells went through favorable Phase 1 and 2 studies demonstrating safety and possible efficacy (Makkar et al., Lancet 379(9819):895-904, 2012; Ishigami et al., Circ Res 120(7):1162-1173, 2017; Ishigami et al., Circ Res 116 (4):653-664, 2015; Tarui et al., J Thorac Cardiovasc Surg 150(5):1198-1207, 1208 e1191-1192, 2015). In this context and in view of clinical applications, cells have to be prepared and released according to Good Manufacturing Practices (GMP) (EudraLex-volume 4-good manufacturing practice (GMP) guidelines-Part I-basic requirements for medicinal products. http://ec.europa.eu/health/documents/eudralex/vol-4 ; EudraLex-volume 4-good manufacturing practice (GMP) guidelines-Part IV-guidelines on good manufacturing practices specific to advanced therapy medicinal products. http://ec.europa.eu/health/documents/eudralex/vol-4 ). This chapter describes GMP-grade methods for production and testing of a CPC Master Cell Bank (MCB), consisting of frozen aliquots of cells that may be used either as a therapeutic product or as source for the manufacturing of Exo for clinical trials.The MCB production method has been designed to isolate and expand CPC from human cardiac tissue in xeno-free conditions (Andriolo et al., Front Physiol 9:1169, 2018). The quality control (QC) methods have been implemented to assess the safety (sterility, endotoxin, mycoplasma, cell senescence, tumorigenicity) and identity/potency/purity (cell count and viability, RT-PCR, immunophenotype) of the cells (Andriolo et al., Front Physiol 9:1169, 2018).


Subject(s)
Biomedical Technology/standards , Myoblasts/cytology , Myocytes, Cardiac/cytology , Primary Cell Culture/methods , Biological Specimen Banks/standards , Biomedical Technology/methods , Cells, Cultured , Humans , Practice Guidelines as Topic , Primary Cell Culture/standards , Tissue Preservation/standards
15.
Methods Mol Biol ; 2286: 167-178, 2021.
Article in English | MEDLINE | ID: mdl-33381855

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) have the potential to be used in a variety of biomedical applications, including drug discovery and Regenerative Medicine. The success of these approaches is, however, limited by the difficulty of generating the large quantities of cells required in a reproducible and controlled system. Bioreactors, widely used for industrial manufacture of biological products, constitute a viable strategy for large-scale production of stem cell derivatives. In this chapter, we describe the expansion of hiPSCs using the Vertical-Wheel™ bioreactor, a novel bioreactor configuration specifically designed for the culture of shear-sensitive cells. We provide protocols for the expansion of hiPSCs in suspension, both as floating aggregates and using microcarriers for cell adhesion. These methods may be important for the establishment of a scalable culture of hiPSCs, allowing the manufacturing of industrial- or clinical-scale cell numbers.


Subject(s)
Biomedical Technology/methods , Bioreactors/standards , Induced Pluripotent Stem Cells/cytology , Primary Cell Culture/methods , Biomedical Technology/instrumentation , Biomedical Technology/standards , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology , Practice Guidelines as Topic , Primary Cell Culture/instrumentation , Primary Cell Culture/standards
16.
Methods Mol Biol ; 2286: 67-71, 2021.
Article in English | MEDLINE | ID: mdl-33349901

ABSTRACT

Over the past few years, a large number of clinical studies for advanced therapy medicinal products have been registered and/or conducted for treating various diseases around the world and many have generated very exciting outcomes. Media fill, the validation of the aseptic manufacturing process, is the simulation of medicinal product manufacturing using nutrient media. The purpose of this study is to explain the media fill procedure stepwise in the context of cellular therapy medicinal products. The aseptic preparation of patient individual cellular product is simulated by using tryptic soy broth as the growth medium, and sterile vials as primary packaging materials.


Subject(s)
Biomedical Technology/standards , Culture Media/standards , Primary Cell Culture/methods , Sterilization/standards , Tissue Culture Techniques/methods , Biomedical Technology/instrumentation , Cells, Cultured , Human Embryonic Stem Cells/cytology , Humans , Practice Guidelines as Topic , Primary Cell Culture/standards , Sterilization/methods , Tissue Culture Techniques/standards
17.
Methods Mol Biol ; 2286: 73-84, 2021.
Article in English | MEDLINE | ID: mdl-33349902

ABSTRACT

Mesenchymal stem cells have gained popularity in cell-based therapies due to their regenerative capabilities, immunomodulation properties, and paracrine activity through trophic factors. It is of utmost importance to establish clinical-grade procedures for the preparation of the mesenchymal stem cells for clinical applications. Here, we describe detailed procedures for isolation, culture, cryopreservation, and preparation of mesenchymal stem cells derived from umbilical cord as a final product under good manufacturing practices-compliant conditions.


Subject(s)
Biomedical Technology/standards , Cryopreservation/standards , Mesenchymal Stem Cells/cytology , Primary Cell Culture/standards , Tissue and Organ Harvesting/standards , Umbilical Cord/cytology , Biomedical Technology/methods , Cells, Cultured , Humans , Practice Guidelines as Topic , Tissue and Organ Harvesting/methods
18.
Methods Mol Biol ; 2286: 85-94, 2021.
Article in English | MEDLINE | ID: mdl-33349903

ABSTRACT

Cell-based therapies have become a popular approach in the field of regenerative medicine. Human fibroblast cells, one of the cell types widely used in clinical applications, have been used for skin regeneration and wound healing procedures. Furthermore, they are utilized for aesthetic purposes since fibroblasts lose their abilities such as collagen synthesis with age. Here, we describe detailed procedures for isolation, culture, cryopreservation, and preparation of fibroblasts derived from adult human skin as a final product under good manufacturing practice-compliant conditions.


Subject(s)
Biomedical Technology/standards , Cryopreservation/methods , Fibroblasts/cytology , Primary Cell Culture/methods , Skin/cytology , Biomedical Technology/methods , Cells, Cultured , Cryopreservation/standards , Humans , Practice Guidelines as Topic , Primary Cell Culture/standards , Tissue and Organ Harvesting/methods , Tissue and Organ Harvesting/standards
19.
Methods Mol Biol ; 2286: 95-105, 2021.
Article in English | MEDLINE | ID: mdl-33534112

ABSTRACT

Bone marrow stromal cells (BMSCs, also known as bone marrow mesenchymal stem cells) are a plastic-adherent heterogeneous cell population that contain inherent skeletal progenitors and a subset of multipotential skeletal stem cells (SSCs). Application of BMSCs in therapeutic protocols implies its isolation and expansion under good manufacturing practices (GMP). Here we describe the procedures we have found to successfully generate practical BMSCs numbers, with preserved biological potency.


Subject(s)
Biomedical Technology/standards , Bone Marrow Cells/cytology , Bone and Bones/cytology , Primary Cell Culture/methods , Antigens, CD34/genetics , Antigens, CD34/metabolism , Biomedical Technology/methods , Cells, Cultured , Coculture Techniques/economics , Coculture Techniques/methods , Coculture Techniques/standards , Costs and Cost Analysis , Culture Media, Serum-Free/chemistry , Humans , Practice Guidelines as Topic , Primary Cell Culture/economics , Primary Cell Culture/standards , Stromal Cells/cytology , Stromal Cells/metabolism
20.
Cells ; 10(5)2021 05 05.
Article in English | MEDLINE | ID: mdl-34063138

ABSTRACT

In plastic surgery, lipofilling is a frequent procedure. Unsatisfactory vascularization and impaired cell vitality can lead to unpredictable take rates in the fat graft. The proliferation and neovascularization inducing properties of adipose tissue-derived stem cells may contribute to solve this problem. Therefore, the enrichment of fat grafts with stem cells is studied intensively. However, it is difficult to compare these studies because many factors-often not precisely described-are influencing the results. Our study summarizes some factors which influence the cell yield like harvesting, isolation procedure and quantification. Stem cells were isolated after liposuction. Quantification was done using a cell chamber, colony counting, or flow cytometry with changes to one parameter, only, for each comparison. Quantification of cells isolated after liposuction at the same harvesting site from the same patient can vary greatly depending on the details of the isolation protocol and the method of quantification. Cell yield can be influenced strongly by many factors. Therefore, a comparison of different studies should be handled with care.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cells/cytology , Primary Cell Culture/methods , Tissue and Organ Harvesting/methods , Cells, Cultured , Flow Cytometry/methods , Flow Cytometry/standards , Humans , Lipectomy/methods , Lipectomy/standards , Primary Cell Culture/standards , Tissue and Organ Harvesting/standards
SELECTION OF CITATIONS
SEARCH DETAIL