Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.234
Filter
1.
Cancer Immunol Immunother ; 73(12): 248, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39358555

ABSTRACT

BACKGROUND: Tumor-reactive T cells play a crucial role in anti-tumor responses, but T cells induced by DNA vaccination are time-consuming processes and exhibit limited anti-tumor efficacy. Therefore, we evaluated the anti-tumor effectiveness of reactive T cells elicited by electroporation (EP)-mediated DNA vaccine targeting epidermal growth factor receptor variant III (pEGFRvIII plasmid), in conjunction with adoptive cell therapy (ACT), involving the transfer of lymphocytes from a pEGFRvIII EP-vaccinated healthy donor. METHODS: The validation of the established pEGFRvIII plasmid and EGFRvIII-positive cell model was confirmed through immunofluorescence and western blot analysis. Flow cytometry and cytotoxicity assays were performed to evaluate the functionality of antigen-specific reactive T cells induced by EP-mediated pEGFRvIII vaccines, ACT, or their combination. The anti-tumor effectiveness of EP-mediated pEGFRvIII vaccines alone or combined with ACT was evaluated in the B16F10-EGFRvIII tumor model. RESULTS: EP-mediated pEGFRvIII vaccines elicited serum antibodies and a robust cellular immune response in both healthy and tumor-bearing mice. However, this response only marginally inhibited early-stage tumor growth in established tumor models. EP-mediated pEGFRvIII vaccination followed by adoptive transfer of lymphocytes from vaccinated healthy donors led to notable anti-tumor efficacy, attributed to the synergistic action of antigen-specific CD4+ Th1 cells supplemented by ACT and antigen-specific CD8+ T cells elicited by the EP-mediated DNA vaccination. CONCLUSIONS: Our preclinical studies results demonstrate an enhanced anti-tumor efficacy of EP-mediated DNA vaccination boosted with adoptively transferred, vaccinated healthy donor-derived allogeneic lymphocytes.


Subject(s)
Cancer Vaccines , Electroporation , Vaccines, DNA , Animals , Vaccines, DNA/immunology , Electroporation/methods , Mice , Cancer Vaccines/immunology , Mice, Inbred C57BL , Immunotherapy, Adoptive/methods , Female , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Disease Models, Animal , Cell Line, Tumor , Allogeneic Cells/immunology , ErbB Receptors/immunology
2.
Nat Commun ; 15(1): 8099, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284842

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR)-based editing tools have transformed the landscape of genome editing. However, the absence of a robust and safe CRISPR delivery method continues to limit its potential for therapeutic applications. Despite the emergence of various methodologies aimed at addressing this challenge, issues regarding efficiency and editing operations persist. We introduce a microfluidic gene delivery system, called droplet cell pincher (DCP), designed for highly efficient and safe genome editing. This approach combines droplet microfluidics with cell mechanoporation, enabling encapsulation and controlled passage of cells and CRISPR systems through a microscale constriction. Discontinuities created in cell and nuclear membranes upon passage facilitate the rapid CRISPR-system internalization into the nucleus. We demonstrate the successful delivery of various macromolecules, including mRNAs (~98%) and plasmid DNAs (~91%), using this platform, underscoring the versatility of the DCP and leveraging it to achieve successful genome engineering through CRISPR-Cas9 delivery. Our platform outperforms electroporation, the current state-of-the-art method, in three key areas: single knockouts (~6.5-fold), double knockouts (~3.8-fold), and knock-ins (~3.8-fold). These results highlight the potential of our platform as a next-generation tool for CRISPR engineering, with implications for clinical and biological cell-based research.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Editing/methods , Humans , Microfluidics/methods , Gene Transfer Techniques , Electroporation/methods , HEK293 Cells , Plasmids/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics
3.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273092

ABSTRACT

The utilization of electroporation for delivering CRISPR/Cas9 system components has enabled efficient gene editing in mammalian zygotes, facilitating the development of genome-edited animals. In this study, our research focused on targeting the ACTG1 and MSTN genes in sheep, revealing a threshold phenomenon in electroporation with a voltage tolerance in sheep in vitro fertilization (IVF) zygotes. Various poring voltages near 40 V and pulse durations were examined for electroporating sheep zygotes. The study concluded that stronger electric fields required shorter pulse durations to achieve the optimal conditions for high gene mutation rates and reasonable blastocyst development. This investigation also assessed the quality of Cas9/sgRNA ribonucleoprotein complexes (Cas9 RNPs) and their influence on genome editing efficiency in sheep early embryos. It was highlighted that pre-complexation of Cas9 proteins with single-guide RNA (sgRNA) before electroporation was essential for achieving a high mutation rate. The use of suitable electroporation parameters for sheep IVF zygotes led to significantly high mutation rates and heterozygote ratios. By delivering Cas9 RNPs and single-stranded oligodeoxynucleotides (ssODNs) to zygotes through electroporation, targeting the MSTN (Myostatin) gene, a knock-in efficiency of 26% was achieved. The successful generation of MSTN-modified lambs was demonstrated by delivering Cas9 RNPs into IVF zygotes via electroporation.


Subject(s)
CRISPR-Cas Systems , Electroporation , Fertilization in Vitro , Gene Editing , RNA, Guide, CRISPR-Cas Systems , Ribonucleoproteins , Zygote , Animals , Gene Editing/methods , Electroporation/methods , Zygote/metabolism , Fertilization in Vitro/methods , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , Sheep , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Myostatin/genetics , Female , Animals, Genetically Modified
4.
Curr Protoc ; 4(9): e70000, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39228292

ABSTRACT

The sexually transmitted pathogen, Neisseria gonorrhoeae, undergoes natural transformation at high frequency. This property has led to the rapid dissemination of antibiotic resistance markers and the panmictic structure of the gonococcal population. However, high-frequency transformation also makes N. gonorrhoeae one of the easiest bacterial species to manipulate genetically in the laboratory. Techniques have been developed that result in transformation frequencies >50%, allowing the identification of mutants by screening and without selection. Constructs have been created to take advantage of this high-frequency transformation, facilitating genetic mutation, complementation, and heterologous gene expression. Similar methods have been developed for N. meningitidis and nonpathogenic Neisseria including N. mucosa and N. musculi. Techniques are described for genetic manipulation of N. gonorrhoeae and commensal Neisseria species, as well as for growth of these fastidious organisms. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Spot transformation of Neisseria gonorrhoeae on agar plates Basic Protocol 2: Spot transformation of commensal Neisseria on agar plates Basic Protocol 3: Transformation of Neisseria gonorrhoeae in liquid culture Basic Protocol 4: Electroporation of Neisseria gonorrhoeae Basic Protocol 5: Creation of unmarked mutations using a positive and negative selection cassette Basic Protocol 6: In vitro mutagenesis of Neisseria gonorrhoeae chromosomal DNA using EZ-Tn5 Basic Protocol 7: Chemical mutagenesis Basic Protocol 8: Complementation on the Neisseria gonorrhoeae chromosome Alternate Protocol 1: Complementation with replicating plasmids Alternate Protocol 2: Complementation on the Neisseria musculi or Neisseria mucosa chromosome Basic Protocol 9: Preparation of chromosomal DNA from Neisseria gonorrhoeae grown on solid medium Alternate Protocol 3: Preparation of chromosomal DNA from Neisseria gonorrhoeae grown in broth Support Protocol: Preparing PCR templates from Neisseria gonorrhoeae colonies.


Subject(s)
Neisseria gonorrhoeae , Neisseria , Transformation, Bacterial , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/drug effects , Neisseria/genetics , Neisseria/drug effects , Electroporation , Gonorrhea/microbiology , Gonorrhea/drug therapy , Humans
5.
Curr Protoc ; 4(9): e70014, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240247

ABSTRACT

Candida glabrata (Nakaseomyces glabratus) is an opportunistic fungal pathogen that has become a significant concern in clinical settings due to its increasing resistance to antifungal treatments. Understanding the genetic basis of its pathogenicity and resistance mechanisms is crucial for developing new therapeutic strategies. One powerful method of studying gene function is through targeted gene deletion. This paper outlines a comprehensive protocol for the deletion of genes in C. glabrata, encompassing primer design, preparation of electrocompetent cells, transformation, and finally confirmation of the gene deletion. The protocol begins with the identification and design of primers necessary for generating deletion constructs, involving the precise targeting of up- and downstream regions flanking the gene of interest to ensure high specificity and efficiency of homologous recombination. Followed is the preparation of electrocompetent cells, a critical step for successful transformation. Transformation of the competent cells is achieved through electroporation, facilitating the introduction of exogenous DNA into the cells. This is followed by the selection and confirmation of successfully transformed colonies. Confirmation involves the use of colony PCR to verify the correct integration of the NAT resistance cassette and deletion of the target gene. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Primer design for gene deletion in C. glabrata Basic Protocol 2: Preparing competent C. glabrata cells Basic Protocol 3: Transforming C. glabrata using electroporation Basic Protocol 4: Confirming deletion strains with colony PCR.


Subject(s)
Candida glabrata , Gene Deletion , Candida glabrata/genetics , Candida glabrata/pathogenicity , Electroporation , Transformation, Genetic , Homologous Recombination , DNA Primers/genetics
6.
Radiol Oncol ; 58(3): 406-415, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39287162

ABSTRACT

BACKGROUND: Administering gadolinium-based contrast agent before electroporation allows the contrast agent to enter the cells and enables MRI assessment of reversibly electroporated regions. The aim of this study was evaluation of contrast agent entrapment in Chinese hamster ovary (CHO) cells and comparison of these results with those determined by standard in vitro methods for assessing cell membrane permeability, cell membrane integrity and cell survival following electroporation. MATERIALS AND METHODS: Cell membrane permeabilization and cell membrane integrity experiments were performed using YO-PRO-1 dye and propidium iodide, respectively. Cell survival experiments were performed by assessing metabolic activity of cells using MTS assay. The entrapment of gadolinium-based contrast agent gadobutrol inside the cells was evaluated using T1 relaxometry of cell suspensions 25 min and 24 h after electroporation and confirmed by inductively coupled plasma mass spectrometry. RESULTS: Contrast agent was detected 25 min and 24 h after the delivery of electric pulses in cells that were reversibly electroporated. In addition, contrast agent was present in irreversibly electroporated cells 25 min after the delivery of electric pulses but was no longer detected in irreversibly electroporated cells after 24 h. Inductively coupled plasma mass spectrometry showed a proportional decrease in gadolinium content per cell with shortening of T1 relaxation time (R 2 = 0.88 and p = 0.0191). CONCLUSIONS: Our results demonstrate that the contrast agent is entrapped in cells exposed to reversible electroporation but exits from cells exposed to irreversible electroporation within 24 h, thus confirming the hypothesis on which detection experiments in vivo were based.


Subject(s)
Cell Survival , Contrast Media , Cricetulus , Electroporation , Magnetic Resonance Imaging , Organometallic Compounds , Animals , Electroporation/methods , CHO Cells , Magnetic Resonance Imaging/methods , Organometallic Compounds/pharmacokinetics , Cricetinae , Cell Membrane Permeability
7.
J Vis Exp ; (211)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39311601

ABSTRACT

Excitable cells such as neuronal and muscle cells can be primary targets in rapidly emerging electroporation-based treatments. However, they can be affected by electric pulses even in therapies where they are not the primary targets, and this can cause adverse side effects. Therefore, to optimize the electroporation-based treatments of excitable and non-excitable tissues, there is a need to study the effects of electric pulses on excitable cells, their ion channels, and excitability in vitro. For this purpose, a protocol was developed for optical monitoring of changes in action potential generation due to electroporation on a simple excitable cell model of genetically engineered tet-on spiking HEK cells. With the use of a fluorescent potentiometric dye, the changes in transmembrane voltage were monitored under a fluorescence microscope, and relevant parameters of cell responses were extracted automatically with a MATLAB application. This way, the excitable cell responses to different electric pulses and the interplay between excitation and electroporation could be efficiently evaluated.


Subject(s)
Action Potentials , Electroporation , Electroporation/methods , Humans , HEK293 Cells , Action Potentials/physiology , Genetic Engineering/methods , Microscopy, Fluorescence/methods , Fluorescent Dyes/chemistry
8.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273287

ABSTRACT

The phenomenon known as bipolar cancellation is observed when biphasic nanosecond electric field pulses are used, which results in reduced electroporation efficiency when compared to unipolar pulses of the same parameters. Basically, the negative phase of the bipolar pulse diminishes the effect of the positive phase. Our study aimed to investigate how bipolar cancellation affects Ca2+ electrochemotherapy and cellular response under varying electric field intensities and pulse durations (3-7 kV/cm, 100, 300, and 500 ns bipolar 1 MHz repetition frequency pulse bursts, n = 100). As a reference, standard microsecond range parametric protocols were used (100 µs × 8 pulses). We have shown that the cancellation effect is extremely strong when the pulses are closely spaced (1 MHz frequency), which results in a lack of cell membrane permeabilization and consequent failure of electrochemotherapy in vitro. To validate the observations, we have performed a pilot in vivo study where we compared the efficacy of monophasic (5 kV/cm × ↑500 ns × 100) and biphasic sequences (5 kV/cm × ↑500 ns + ↓500 ns × 100) delivered at 1 MHz frequency in the context of Ca2+ electrochemotherapy (B16-F10 cell line, C57BL/6 mice, n = 24). Mice treated with bipolar pulses did not exhibit prolonged survival when compared to the untreated control (tumor-bearing mice); therefore, the bipolar cancellation phenomenon was also occurrent in vivo, significantly impairing electrochemotherapy. At the same time, the efficacy of monophasic nanosecond pulses was comparable to 1.4 kV/cm × 100 µs × 8 pulses sequence, resulting in tumor reduction following the treatment and prolonged survival of the animals.


Subject(s)
Electrochemotherapy , Melanoma, Experimental , Animals , Electrochemotherapy/methods , Mice , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Pilot Projects , Electroporation/methods , Mice, Inbred C57BL , Calcium/metabolism
9.
J Nanobiotechnology ; 22(1): 552, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256765

ABSTRACT

Natural Killer (NK) cells are exciting candidates for cancer immunotherapy with potent innate cytotoxicity and distinct advantages over T cells for Chimeric Antigen Receptor (CAR) therapy. Concerns regarding the safety, cost, and scalability of viral vectors has ignited research into non-viral alternatives for gene delivery. This review comprehensively analyses recent advancements and challenges with non-viral genetic modification of NK cells for allogeneic CAR-NK therapies. Non-viral alternatives including electroporation and multifunctional nanoparticles are interrogated with respect to CAR expression and translational responses. Crucially, the link between NK cell biology and design of drug delivery technologies are made, which is essential for development of future non-viral approaches. This review provides valuable insights into the current state of non-viral CAR-NK cell engineering, aimed at realising the full potential of NK cell-based immunotherapies.


Subject(s)
Cell Engineering , Gene Transfer Techniques , Immunotherapy, Adoptive , Killer Cells, Natural , Receptors, Chimeric Antigen , Killer Cells, Natural/immunology , Humans , Receptors, Chimeric Antigen/genetics , Animals , Immunotherapy, Adoptive/methods , Cell Engineering/methods , Nanoparticles/chemistry , Neoplasms/therapy , Neoplasms/immunology , Electroporation/methods , Immunotherapy/methods
10.
J Cancer Res Ther ; 20(4): 1357-1361, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39206999

ABSTRACT

PURPOSE: This study evaluates the clinical efficacy and safety of irreversible electroporation (IRE) therapy combined with chemotherapy in patients with stage IV pancreatic cancer. METHODS: Between September 2021 and November 2023, we enrolled 38 patients with stage IV pancreatic cancer, with 20 receiving IRE plus chemotherapy and 18 receiving only chemotherapy. We recorded the general information of the patients and regularly followed up postoperative IRE-related adverse reactions. Progression-free survival (PFS) and overall survival (OS) were evaluated during follow-up. RESULTS: Median OS was longer in the IRE group than in the chemotherapy group. Median PFS was slightly extended with IRE compared to chemotherapy alone. The mean hospital stay for the IRE group was 5.90 ± 0.75 days. Four serious adverse events occurred after IRE. Postoperative pain scores were significantly lower than preoperative scores. CONCLUSION: IRE combined with chemotherapy showed clinical effectiveness in stage IV pancreatic cancer treatment, offering potential pain reduction benefits with fewer adverse effects and shorter hospital stays.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Electroporation , Neoplasm Staging , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Male , Female , Middle Aged , Electroporation/methods , Aged , Combined Modality Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Treatment Outcome , Survival Rate , Follow-Up Studies , Length of Stay/statistics & numerical data , Adult , Progression-Free Survival
11.
Front Immunol ; 15: 1415328, 2024.
Article in English | MEDLINE | ID: mdl-39192973

ABSTRACT

Background: The non-viral production of CAR-T cells through electroporation of transposon DNA plasmids is an alternative approach to lentiviral/retroviral methods. This method is particularly suitable for early-phase clinical trials involving novel types of CAR-T cells. The primary disadvantage of non-viral methods is the lower production efficiency compared to viral-based methods, which becomes a limiting factor for CAR-T production, especially in chemotherapy-pretreated lymphopenic patients. Methods: We describe a good manufacturing practice (GMP)-compliant protocol for producing CD19 and CD123-specific CAR-T cells based on the electroporation of transposon vectors. The lymphocytes were purified from the blood of patients undergoing chemotherapy for B-NHL or AML and were electroporated with piggyBac transposon encoding CAR19 or CAR123, respectively. Electroporated cells were then polyclonally activated by anti-CD3/CD28 antibodies and a combination of cytokines (IL-4, IL-7, IL-21). The expansion was carried out in the presence of irradiated allogeneic blood-derived mononuclear cells (i.e., the feeder) for up to 21 days. Results: Expansion in the presence of the feeder enhanced CAR-T production yield (4.5-fold in CAR19 and 9.3-fold in CAR123). Detailed flow-cytometric analysis revealed the persistence of early-memory CAR-T cells and a low vector-copy number after production in the presence of the feeder, with no negative impact on the cytotoxicity of feeder-produced CAR19 and CAR123 T cells. Furthermore, large-scale manufacturing of CAR19 carried out under GMP conditions using PBMCs obtained from B-NHL patients (starting number=200x10e6 cells) enabled the production of >50x10e6 CAR19 in 7 out of 8 cases in the presence of the feeder while only in 2 out of 8 cases without the feeder. Conclusions: The described approach enables GMP-compatible production of sufficient numbers of CAR19 and CAR123 T cells for clinical application and provides the basis for non-viral manufacturing of novel experimental CAR-T cells that can be tested in early-phase clinical trials. This manufacturing approach can complement and advance novel experimental immunotherapeutic strategies against human hematologic malignancies.


Subject(s)
Antigens, CD19 , DNA Transposable Elements , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Antigens, CD19/immunology , Antigens, CD19/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/genetics , Feeder Cells , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Electroporation , Allogeneic Cells/immunology
12.
Biotechnol Adv ; 76: 108421, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39127411

ABSTRACT

Advances in synthetic biology allow the design and manipulation of DNA from the scale of genes to genomes, enabling the engineering of complex genetic information for application in biomanufacturing, biomedicine and other areas. The transfer and subsequent maintenance of large DNA are two core steps in large scale genome rewriting. Compared to small DNA, the high molecular weight and fragility of large DNA make its transfer and maintenance a challenging process. This review outlines the methods currently available for transferring and maintaining large DNA in bacteria, fungi, and mammalian cells. It highlights their mechanisms, capabilities and applications. The transfer methods are categorized into general methods (e.g., electroporation, conjugative transfer, induced cell fusion-mediated transfer, and chemical transformation) and specialized methods (e.g., natural transformation, mating-based transfer, virus-mediated transfection) based on their applicability to recipient cells. The maintenance methods are classified into genomic integration (e.g., CRISPR/Cas-assisted insertion) and episomal maintenance (e.g., artificial chromosomes). Additionally, this review identifies the major technological advantages and disadvantages of each method and discusses the development for large DNA transfer and maintenance technologies.


Subject(s)
Bacteria , DNA , Fungi , Animals , Fungi/genetics , Bacteria/genetics , DNA/genetics , Humans , Electroporation , Gene Transfer Techniques , Mammals/genetics , Synthetic Biology/methods , CRISPR-Cas Systems/genetics
13.
Int J Mol Sci ; 25(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39201461

ABSTRACT

Electroporation-based procedures employing nanosecond bipolar pulses are commonly linked to an undesirable phenomenon known as the cancelation effect. The cancellation effect arises when the second pulse partially or completely neutralizes the effects of the first pulse, simultaneously diminishing cells' plasma membrane permeabilization and the overall efficiency of the procedure. Introducing a temporal gap between the positive and negative phases of the bipolar pulses during electroporation procedures may help to overcome the cancellation phenomenon; however, the exact thresholds are not yet known. Therefore, in this work, we have tested the influence of different interphase delay values (from 0 ms to 95 ms) using symmetric bipolar nanoseconds (300 and 500 ns) on cell permeabilization using 10 Hz, 100 Hz, and 1 kHz protocols. As a model mouse hepatoma, the MH-22a cell line was employed. Additionally, we conducted in vitro electrochemotherapy with cisplatin, employing reduced interphase delay values (0 ms and 0.1 ms) at 10 Hz. Cell plasma membrane permeabilization and viability dependence on a variety of bipolar pulsed electric field protocols were characterized. It was shown that it is possible to minimize bipolar cancellation, enabling treatment efficiency comparable to monophasic pulses with identical parameters. At the same time, it was highlighted that bipolar cancellation has a significant influence on permeabilization, while the effects on the outcome of electrochemotherapy are minimal.


Subject(s)
Cell Membrane Permeability , Electrochemotherapy , Electrochemotherapy/methods , Animals , Mice , Cell Membrane Permeability/drug effects , Cell Line, Tumor , Electroporation/methods , Cisplatin/pharmacology , Cell Membrane/metabolism , Cell Membrane/drug effects , Cell Survival/drug effects , Carcinoma, Hepatocellular/drug therapy , Antineoplastic Agents/pharmacology
14.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39201653

ABSTRACT

Exposing cells to intense and brief electric field pulses can modulate cell permeability, a phenomenon termed electroporation. When applied in medical treatments of diseases like cancer and cardiac arrhythmias, depending on level of cellular destruction, it is also referred to as irreversible electroporation (IRE) or Pulsed Field Ablation (PFA). For ablation device testing, several pulse parameters need to be characterized in a comprehensive manner to assess lesion boundary and efficacy. Overly aggressive voltages and application numbers increase animal burden. The potato tuber is a widely used initial model for the early testing of electroporation. The aim of this study is to characterize and refine bench testing for the ablation outcomes of PFA in this simplistic vegetal model. For in vitro assays, several pulse parameters like voltage, duration, and frequency were modulated to study effects not only on 2D ablation area but also 3D depth and volume. As PFA is a relatively new technology with minimal thermal effects, we also measured temperature changes before, during, and after ablation. Data from experiments were supplemented with in silico modeling to examine E-field distribution. We have estimated the irreversible electroporation threshold in Solanum Tuberosum to be at 240 V/cm. This bench testing platform can screen several pulse recipes at early stages of PFA device development in a rapid and high-throughput manner before proceeding to laborious trials for IRE medical devices.


Subject(s)
Electroporation , Solanum tuberosum , Electroporation/methods , Ablation Techniques/methods , Ablation Techniques/instrumentation , Temperature , Computer Simulation , Irreversible Electroporation Therapy
15.
Theriogenology ; 229: 158-168, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39178617

ABSTRACT

Genome editing is recognized as a powerful tool in agriculture and research, enhancing our understanding of genetic function, diseases, and productivity. However, its progress in buffaloes has lagged behind other mammals due to several challenges, including long gestational periods, single pregnancies, and high raising costs. In this study, we aimed to generate MSTN-edited buffaloes, known for their distinctive double-muscling phenotype, as a proof of concept. To meet our goal, we used somatic cell nuclear transfer (SCNT) and zygotic electroporation (CRISPR-EP) technique. For this, we firstly identified the best transfection method for introduction of RNP complex into fibroblast which was further used for SCNT. For this, we compared the transfection, cleavage efficiency and cell viability of nucleofection and lipofection in adult fibroblasts. The cleavage, transfection efficiency and cell viability of nucleofection group was found to be significantly (P ≤ 0.05) higher than lipofection group. Four MSTN edited colony were generated using nucleofection, out of which three colonies was found to be biallelic and one was monoallelic. Further, we compared the efficacy, embryonic developmental potential and subsequent pregnancy outcome of SCNT and zygotic electroporation. The blastocyst rate of electroporated group was found to be significantly (P ≤ 0.05) higher than SCNT group. However, the zygotic electroporation group resulted into two pregnancies which were confirmed to be MSTN edited. Since, the zygotic electroporation does not require complex micromanipulation techniques associated with SCNT, it has potential for facilitating the genetic modification in large livestock such as buffaloes. The present study lays the basis for inducing genetic alternation with practical or biological significance.


Subject(s)
Buffaloes , CRISPR-Cas Systems , Electroporation , Gene Editing , Nuclear Transfer Techniques , Transfection , Animals , Buffaloes/genetics , Electroporation/veterinary , Electroporation/methods , Female , Pregnancy , Gene Editing/methods , Gene Editing/veterinary , Transfection/veterinary , Transfection/methods , Nuclear Transfer Techniques/veterinary , Myostatin/genetics , Zygote/metabolism
16.
Bioelectrochemistry ; 160: 108789, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39128409

ABSTRACT

Electroporation (EP) of the normal cell and cancer cell both in single-cell and multicellular models was investigated by the meshed transport network method (MTNM) in this paper. The simulation results suggest that the cancer cell undergoes faster and more significant local EP than that of the corresponding normal cell induced by nanosecond pulsed electric fields (nsPEFs) both in single-cell and multicellular models. Furthermore, the results of the multicellular model indicate that there is a unidirectional neighboring effect in the multicellular model, meaning that cells at the center are affected and their pore formation is significantly reduced, but this effect is very weak for cells at the edges of the system. This means that the electric field selectively kills cells in different distribution locations. This work can provide guidance for the selection of parameters for the cancer cell EP process.


Subject(s)
Electroporation , Models, Biological , Neoplasms , Electroporation/methods , Humans , Neoplasms/pathology , Neoplasms/therapy , Computer Simulation
17.
STAR Protoc ; 5(3): 103261, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39141518

ABSTRACT

Electroporation temporarily enhances cell membrane permeability and promotes the absorption of external molecules. We have developed a device termed the rolling microneedle electrode array (RoMEA) that combines a densely arranged microneedle array of electrodes with rolling structures. Use RoMEA to create uniform skin micropores for efficient, low-damage transfection of nucleic acids over extended areas of the body. We describe in detail the design, fabrication, and assembly of the device and the application of in vivo electroporation of nucleic acids. For complete details on the use and execution of this protocol, please refer to Tongren Yang et al. 1.


Subject(s)
Electrodes , Electroporation , Needles , Electroporation/methods , Electroporation/instrumentation , Animals , Nucleic Acids , Mice , Transfection/methods , Transfection/instrumentation , Gene Transfer Techniques/instrumentation , Equipment Design
18.
Bull Cancer ; 111(10): 967-979, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39198085

ABSTRACT

The advent of robotic systems in interventional radiology marks a significant evolution in minimally invasive medical procedures, offering enhanced precision, safety, and efficiency. This review comprehensively analyzes the current state and applications of robotic system usage in interventional radiology, which can be particularly helpful for complex procedures and in challenging anatomical regions. Robotic systems can improve the accuracy of interventions like microwave ablation, radiofrequency ablation, and irreversible electroporation. Indeed, studies have shown a notable decrease of an average 30% in the mean deviation of probes, and a 40% lesser need for adjustments during interventions carried out with robotic assistance. Moreover, this review highlights a 35% reduction in radiation dose and a stable-to-30% reduction in operating time associated with robot-assisted procedures compared to manual methods. Additionally, the potential of robotic systems to standardize procedures and minimize complications is discussed, along with the challenges they pose, such as setup duration, organ movement, and a lack of tactile feedback. Despite these advancements, the field still grapples with a dearth of randomized controlled trials, which underscores the need for more robust evidence to validate the efficacy and safety of robotic system usage in interventional radiology.


Subject(s)
Radiology, Interventional , Robotic Surgical Procedures , Humans , Robotic Surgical Procedures/methods , Robotic Surgical Procedures/instrumentation , Robotic Surgical Procedures/trends , Radiology, Interventional/methods , Radiology, Interventional/instrumentation , Neoplasms/surgery , Radiation Dosage , Operative Time , Radiofrequency Ablation/methods , Radiofrequency Ablation/instrumentation , Robotics/instrumentation , Robotics/methods , Electroporation/methods , Electroporation/instrumentation , Microwaves/therapeutic use
19.
J Cardiovasc Electrophysiol ; 35(10): 2071-2075, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39138830

ABSTRACT

INTRODUCTION: Pulsed-field ablation (PFA) is a novel nonthermal energy that shows unique features that can be of use beyond pulmonary vein ablation, like tissue selectivity or proximity rather than contact dependency. METHODS AND RESULTS: We report three cases of right focal atrial tachycardias arising from the superior cavoatrial junction and the crista terminalis, in close relationship with the phrenic nerve, effectively ablated using a commercially available PFA catheter designed for pulmonary vein isolation without collateral damage. CONCLUSION: PFA can be useful for treating right atrial tachycardias involving sites near the phrenic nerve, avoiding the need for complex nerve-sparing strategies.


Subject(s)
Electroporation , Phrenic Nerve , Humans , Phrenic Nerve/physiopathology , Male , Middle Aged , Treatment Outcome , Female , Tachycardia, Supraventricular/surgery , Tachycardia, Supraventricular/physiopathology , Tachycardia, Supraventricular/diagnosis , Heart Rate , Aged , Catheter Ablation , Electrophysiologic Techniques, Cardiac , Action Potentials , Cardiac Catheters
20.
Poult Sci ; 103(10): 104144, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39173570

ABSTRACT

The most current in vitro genetic methods, including gene preservation, gene editing and developmental modelling, require a significant number of healthy cells. In poultry species, primordial germ cells (PGCs) are great candidates for all the above-mentioned purposes, given their easy culturing and well-established freezing method for chicken. However, the constant monitoring of cultures can be financially challenging and consumes large amounts of solutions and accessories. This study aimed to introduce the Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI) complex into the chicken PGCs. FUCCI is a powerful transgenic tool based on the periodic protein expression changes during the cell cycle. It includes chromatin licensing and DNA replication factor 1 attached monomeric Kusabira-Orange and Geminin-attached monomeric Azami-Green fluorescent proteins, that cause the cells to express a red signal in the G1 phase and a green signal in S and G2 phases. Modification of the chicken PGCs was done via electroporation and deemed to be successful according to confocal microscopy, DNA sequencing and timelapse video analysis. Stable clone cell lines were established, cryopreserved, and injected into recipient embryos to prove the integrational competency. The cell health monitoring was tested with medium change experiments, that proved the intended reactions of the FUCCI transgene. These results established the future for FUCCI experiments in chicken, including heat treatment and toxin treatment.


Subject(s)
Cell Cycle , Chickens , Germ Cells , Animals , Ubiquitination , Chick Embryo , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Electroporation/veterinary , Electroporation/methods
SELECTION OF CITATIONS
SEARCH DETAIL