Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Hum Cell ; 34(6): 1601-1616, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34378170

ABSTRACT

Induced pluripotent stem cells (iPSCs) are produced from adult somatic cells through reprogramming, which behave like embryonic stem cells (ESCs) but avoiding the controversial ethical issues from destruction of embryos. Since the first discovery in 2006 of four factors that are essential for maintaining the basic characteristics of ESC, global researches have rapidly improved the techniques for generating iPSCs. In this paper, we review new insights into patient-specific iPSC and summarize selected "disease-in-a-dish" examples that model the genetic and epigenetic variations of human diseases. Although more researches need to be done, studies have increasingly focused on the potential utility of iPSCs. The usability of iPSC technology is changing the fields of disease modeling and precision treatment. Aside from its potential use in regenerative cellular therapy for degenerative diseases, iPSC offers a range of new opportunities for the study of genetic human disorders, particularly, rare diseases. We believe that this rapidly moving field promises many more developments that will benefit modern medicine.


Subject(s)
Cell Engineering/methods , Cell Engineering/trends , Induced Pluripotent Stem Cells , Animals , Cell- and Tissue-Based Therapy , Cellular Reprogramming , Embryonic Stem Cells , Humans , Mice , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/therapy , Rare Diseases/genetics , Rare Diseases/therapy , Regenerative Medicine
2.
Sci Rep ; 11(1): 2056, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479431

ABSTRACT

Tremendous advancements in cell and protein engineering methodologies and bioinformatics have led to a vast increase in bacterial production clones and recombinant protein variants to be screened and evaluated. Consequently, an urgent need exists for efficient high-throughput (HTP) screening approaches to improve the efficiency in early process development as a basis to speed-up all subsequent steps in the course of process design and engineering. In this study, we selected the BioLector micro-bioreactor (µ-bioreactor) system as an HTP cultivation platform to screen E. coli expression clones producing representative protein candidates for biopharmaceutical applications. We evaluated the extent to which generated clones and condition screening results were transferable and comparable to results from fully controlled bioreactor systems operated in fed-batch mode at moderate or high cell densities. Direct comparison of 22 different production clones showed great transferability. We observed the same growth and expression characteristics, and identical clone rankings except one host-Fab-leader combination. This outcome demonstrates the explanatory power of HTP µ-bioreactor data and the suitability of this platform as a screening tool in upstream development of microbial systems. Fast, reliable, and transferable screening data significantly reduce experiments in fully controlled bioreactor systems and accelerate process development at lower cost.


Subject(s)
Bioreactors , Cell Engineering/trends , High-Throughput Screening Assays/methods , Protein Engineering/trends , Biomass , Escherichia coli/genetics , Humans , Recombinant Proteins/genetics
4.
Int J Mol Sci ; 21(19)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33007850

ABSTRACT

The engineering of T cells through expression of chimeric antigen receptors (CARs) against tumor-associated antigens (TAAs) has shown significant potential for use as an anti-cancer therapeutic. The development of strategies for flexible and modular CAR T systems is accelerating, allowing for multiple antigen targeting, precise programming, and adaptable solutions in the field of cellular immunotherapy. Moving beyond the fixed antigen specificity of traditional CAR T systems, the modular CAR T technology splits the T cell signaling domains and the targeting elements through use of a switch molecule. The activity of CAR T cells depends on the presence of the switch, offering dose-titratable response and precise control over CAR T cells. In this review, we summarize developments in universal or modular CAR T strategies that expand on current CAR T systems and open the door for more customizable T cell activity.


Subject(s)
Immunotherapy , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/immunology , Cell Engineering/trends , Humans , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
5.
J Hematol Oncol ; 13(1): 86, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616000

ABSTRACT

Chimeric antigen receptor T (CAR-T) cell therapy is regarded as an effective solution for relapsed or refractory tumors, particularly for hematological malignancies. Although the initially approved anti-CD19 CAR-T therapy has produced impressive outcomes, setbacks such as high relapse rates and resistance were experienced, driving the need to discover engineered CAR-T cells that are more effective for therapeutic use. Innovations in the structure and manufacturing of CAR-T cells have resulted in significant improvements in efficacy and persistence, particularly with the development of fourth-generation CAR-T cells. Paired with an immune modifier, the use of fourth-generation and next-generation CAR-T cells will not be limited because of cytotoxic effects and will be an efficient tool for overcoming the tumor microenvironment. In this review, we summarize the recent transformations in the ectodomain, transmembrane domain, and endodomain of the CAR structure, which, together with innovative manufacturing technology and improved cell sources, improve the prospects for the future development of CAR-T cell therapy.


Subject(s)
Cell Engineering/trends , Immunotherapy, Adoptive/trends , Receptors, Chimeric Antigen/genetics , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , CD28 Antigens/chemistry , CD28 Antigens/immunology , Chemotaxis, Leukocyte , Clinical Trials as Topic , Cytokines/metabolism , Genetic Vectors/genetics , Humans , Immunotherapy, Adoptive/methods , Lentivirus/genetics , Lymphoma, Large B-Cell, Diffuse/therapy , Neoplasms/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein Binding , Protein Domains , Protein Engineering , Receptors, Chemokine/immunology , Receptors, Chimeric Antigen/agonists , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Microenvironment
6.
G Chir ; 41(1): 50-65, 2020.
Article in English | MEDLINE | ID: mdl-32038013

ABSTRACT

The loss or deficiency of a tissue or organ is a serious health problem and severely affects the patients' quality of life. In the near future, an option for solving this problem will be the development of bio-derived extracellular matrices (ECM) from huPlastic Romaman or animal tissues and their usage for in vitro or in vivo cellular reseeding. Many researchers are working on this development. Nowadays, different decellularization protocols have been developed to manufacture ECMs but there is not yet a consensus about the most efficient procedures. This review aims to describe the state of the art in the field of decellularization of complex mucosal tissues, analizing and comparing the most recent and most valiant articles published about this procedure.


Subject(s)
Extracellular Matrix , Tissue Engineering/trends , Animals , Cell Engineering/methods , Cell Engineering/trends , Humans , Mucous Membrane , Quality of Life , Tissue Engineering/methods
7.
SLAS Technol ; 24(4): 359-372, 2019 08.
Article in English | MEDLINE | ID: mdl-30917060

ABSTRACT

The next generation of therapies is moving beyond the use of small molecules and proteins to using whole cells. Compared with the interactions of small-molecule drugs with biomolecules, which can largely be understood through chemistry, cell therapies act in a chemical and physical world and can actively adapt to that world, amplifying complexity but also the potential for truly breakthrough impact. Although there has been success in introducing targeting proteins into cells to achieve a therapeutic effect, for example, chimeric antigen receptor (CAR) T cells, our ability to engineer cells is generally limited to introducing proteins, but not modulating large-scale traits or structures of cellular "machines," which play critical roles in disease. Example traits include the ability to secrete compounds, deform through tissue, adhere to surrounding cells, apply force to phagocytose targets, or move through extracellular matrix. There is an opportunity to increase the efficacy of cell therapies through the use of quantitative automation tools, to analyze, sort, and select rare cells with beneficial traits. Combined with methods of genetic or epigenetic mutagenesis to create diversity, such approaches can enable the directed cellular evolution of new therapeutically optimal populations of cells and uncover genetic underpinnings of these optimal traits.


Subject(s)
Cell Engineering/methods , Cell Separation/methods , Cell- and Tissue-Based Therapy/methods , Automation, Laboratory/instrumentation , Automation, Laboratory/methods , Cell Engineering/trends , Cell Separation/trends , Cell- and Tissue-Based Therapy/trends , Humans , Microfluidics/instrumentation , Microfluidics/methods
8.
Nano Lett ; 19(3): 1701-1705, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30773888

ABSTRACT

Mesenchymal stem cell (MSC) has been increasingly applied to cancer therapy because of its tumor-tropic capability. However, short retention at target tissue and limited payload option hinder the progress of MSC-based cancer therapy. Herein, we proposed a hybrid spheroid/nanomedicine system, comprising MSC spheroid entrapping drug-loaded nanocomposite, to address these limitations. Spheroid formulation enhanced MSC's tumor tropism and facilitated loading of different types of therapeutic payloads. This system acted as an active drug delivery platform seeking and specifically targeting glioblastoma cells. It enabled effective delivery of combinational protein and chemotherapeutic drugs by engineered MSC and nanocomposite, respectively. In an in vivo migration model, the hybrid spheroid showed higher nanocomposite retention in the tumor tissue compared with the single MSC approach, leading to enhanced tumor inhibition in a heterotopic glioblastoma murine model. Taken together, this system integrates the merits of cell- and nanoparticle- mediated drug delivery with the tumor-homing characteristics of MSC to advance targeted combinational cancer therapy.


Subject(s)
Drug Delivery Systems , Glioblastoma/drug therapy , Mesenchymal Stem Cells/chemistry , Spheroids, Cellular/transplantation , Cell Engineering/trends , Cell Movement/drug effects , Combined Modality Therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mesenchymal Stem Cells/cytology , Nanomedicine/trends , Spheroids, Cellular/chemistry , Viral Tropism/drug effects
9.
Molecules ; 24(1)2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30621193

ABSTRACT

Click chemistry has great potential for use in binding between nucleic acids, lipids, proteins, and other molecules, and has been used in many research fields because of its beneficial characteristics, including high yield, high specificity, and simplicity. The recent development of copper-free and less cytotoxic click chemistry reactions has allowed for the application of click chemistry to the field of medicine. Moreover, metabolic glycoengineering allows for the direct modification of living cells with substrates for click chemistry either in vitro or in vivo. As such, click chemistry has become a powerful tool for cell transplantation and drug delivery. In this review, we describe some applications of click chemistry for cell engineering in cell transplantation and for drug delivery in the diagnosis and treatment of diseases.


Subject(s)
Cell Engineering/trends , Cell Transplantation/trends , Click Chemistry/trends , Drug Delivery Systems/trends , Humans , Metabolic Engineering
14.
Curr Opin Genet Dev ; 52: 100-108, 2018 10.
Article in English | MEDLINE | ID: mdl-29957586

ABSTRACT

Widespread treatment of human diseases with gene therapies necessitates the development of gene transfer vectors that integrate genetic information effectively, safely and economically. Indeed, significant efforts have been devoted to engineer novel tools that (i) achieve high-level stable gene transfer at low toxicity to the host cell; (ii) induce low levels of genotoxicity and possess a `safe' integration profile with a high proportion of integrations into safe genomic locations; and (iii) are associated with acceptable cost per treatment, and scalable/exportable vector production to serve large numbers of patients. Two decades after the discovery of the Sleeping Beauty (SB) transposon, it has been transformed into a vector system that is fulfilling these requirements. Here we review recent developments in vectorization of SB as a tool for gene therapy, and highlight clinical development of the SB system towards hematopoietic stem cell gene therapy and cancer immunotherapy.


Subject(s)
DNA Transposable Elements/genetics , Hematopoietic Stem Cells/cytology , Immunotherapy/trends , Transposases/genetics , Animals , Cell Engineering/trends , Humans
15.
Front Immunol ; 9: 233, 2018.
Article in English | MEDLINE | ID: mdl-29497421

ABSTRACT

T regulatory cells, a specialized subset of T cells, are key players in modulating antigen (Ag)-specific immune responses in vivo. Inducible T regulatory type 1 (Tr1) cells are characterized by the co-expression of CD49b and lymphocyte-activation gene 3 (LAG-3) and the ability to secrete IL-10, TGF-ß, and granzyme (Gz) B, in the absence of IL-4 and IL-17. The chief mechanisms by which Tr1 cells control immune responses are secretion of IL-10 and TGF-ß and killing of myeloid cells via GzB. Tr1 cells, first described in peripheral blood of patients who developed tolerance after HLA-mismatched fetal liver hematopoietic stem cell transplantation, have been proven to modulate inflammatory and effector T cell responses in several immune-mediated diseases. The possibility to generate and expand Tr1 cells in vitro in an Ag-specific manner has led to their clinical use as cell therapy in patients. Clinical grade protocols to generate or to enrich and expand Tr1 cell medicinal products have been established. Proof-of-concept clinical trials with Tr1 cell products have demonstrated the safety and the feasibility of this approach and indicated some clinical benefit. In the present review, we provide an overview on protocols established to induce/expand Tr1 cells in vitro for clinical application and on results obtained in Tr1 cell-based clinical trials. Moreover, we will discuss a recently developed protocol to efficient convert human CD4+ T cells into a homogeneous population of Tr1-like cells by lentiviral vector-mediated IL-10 gene transfer.


Subject(s)
Cell Engineering/methods , Immune Tolerance , Immunotherapy/methods , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Biological Factors/immunology , Biological Factors/metabolism , Biological Factors/therapeutic use , Cell Culture Techniques/methods , Cell Engineering/trends , Cell Line , Cell Transplantation/methods , Clinical Trials as Topic , Disease Models, Animal , Gene Transfer Techniques , Humans , Integrin alpha2/immunology , Integrin alpha2/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , T-Lymphocytes, Regulatory/transplantation , Lymphocyte Activation Gene 3 Protein
16.
Pediatr Nephrol ; 33(7): 1105-1111, 2018 07.
Article in English | MEDLINE | ID: mdl-28589209

ABSTRACT

End-stage kidney disease (ESKD) is one of the most prevalent diseases in the world with significant morbidity and mortality. Current modes of renal replacement therapy include dialysis and renal transplantation. Although dialysis is an acceptable mode of renal replacement therapy, it does have its shortcomings, which include poorer life expectancy compared with renal transplantation, risk of infections and vascular thrombosis, lack of vascular access and absence of biosynthetic functions of the kidney. Renal transplantation, in contrast, is the preferred option of renal replacement therapy, with improved morbidity and mortality rates and quality of life, compared with dialysis. Renal transplantation, however, may not be available to all patients with ESKD. Some of the key factors limiting the availability and efficiency of renal transplantation include shortage of donor organs and the constant risk of rejection with complications associated with over-immunosuppression respectively. This review focuses chiefly on the potential roles of bioengineering in overcoming limitations in renal transplantation via the development of cell-based bioartificial dialysis devices as bridging options before renal transplantation, and the development of new sources of organs utilizing cell and organ engineering.


Subject(s)
Bioartificial Organs , Cell Engineering/methods , Kidney Failure, Chronic/surgery , Kidney Transplantation/methods , Renal Replacement Therapy/instrumentation , Biomedical Technology/methods , Cell Engineering/trends , Child , Humans , Kidney Transplantation/trends , Quality of Life , Renal Replacement Therapy/methods
17.
Adv Exp Med Biol ; 1016: 3-27, 2017.
Article in English | MEDLINE | ID: mdl-29130151

ABSTRACT

Over the past few decades the ability to edit human cells has revolutionized modern biology and medicine. With advances in genome editing methodologies, gene delivery and cell-based therapeutics targeted at treatment of genetic disease have become a reality that will become more and more essential in clinical practice. Modifying specific mutations in eukaryotic cells using CRISPR-Cas systems derived from prokaryotic immune systems has allowed for precision in correcting various disease mutations. Furthermore, delivery of genetic payloads by employing viral tropism has become a crucial and effective mechanism for delivering genes and gene editing systems into cells. Lastly, cells modified ex vivo have tremendous potential and have shown effective in studying and treating a myriad of diseases. This chapter seeks to highlight and review important progress in the realm of the editing of human cells using CRISPR-Cas systems, the use of viruses as vectors for gene therapy, and the application of engineered cells to study and treat disease.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Genetic Vectors/genetics , Viruses/genetics , Cell Engineering/trends , Genetic Therapy/trends , Humans
18.
Biotechnol J ; 12(8)2017 Aug.
Article in English | MEDLINE | ID: mdl-28497498

ABSTRACT

The ability to preserve stem cells/cells with minimal damage for short and long periods of time is essential for advancements in biomedical therapies and biotechnology. New methods of cell banking are continuously needed to provide effective damage prevention to cells. This paper puts forward a solution to the problem of the low viability of cells during cryopreservation in a traditional suspension and storage by developing innovative multiple emulsion-based carriers for the encapsulation and cryopreservation of cells. During freezing-thawing processes, irreversible damage to cells occurs as a result of the formation of ice crystals, cell dehydration, and the toxicity of cryoprotectant. The proposed method was effective due to the "flexible" protective structure of multiple emulsions, which was proven by a high cell survival rate, above 90%. Results make new contributions in the fields of cell engineering and biotechnology and contribute to the development of methods for banking biological material.


Subject(s)
Cell Survival/drug effects , Cryopreservation/methods , Cryoprotective Agents/chemistry , Mesenchymal Stem Cells/cytology , Cell Engineering/trends , Cryoprotective Agents/pharmacology , Emulsions/chemistry , Emulsions/pharmacology , Freezing , Mesenchymal Stem Cells/drug effects
19.
Transfusion ; 57(5): 1115-1121, 2017 May.
Article in English | MEDLINE | ID: mdl-28380265

ABSTRACT

T cells play a key role in the adaptive immune response, and the ability to manipulate T cells for therapeutic uses has advanced in the past decade. Infusion of expanded or engineered T cells can potentially be used to treat cancer, viral infections, graft-versus-host disease, and organ transplant rejection. The role that blood banks play in the manufacture and distribution of T-cell therapeutics is still being defined. Given the regulatory framework of blood banks, they are well positioned to collect raw material for manufacture of T-cell therapies and to distribute finished product to hospitals in support of clinical trials or eventually for licensed products. A deeper level of involvement in manufacture of T-cell therapeutics is also possible, although that requires more substantial investment in physical facilities and personnel with the regulatory and scientific expertise to prepare and produce cellular therapy products. Examples of physical infrastructure needed would be a laboratory with a clean room for culture of T cells, specialized equipment for expansion of the cells, and adequate administrative and storage support space. Processes that would need to be developed to produce T-cell therapeutics would include development of standard operating procedures and an appropriate quality assurance program. As blood banks consider supporting this novel class of therapies, they will need to weigh capital and expertise requirements with the benefits of providing a novel therapy and the potential of growth for their operations.


Subject(s)
Immunotherapy/methods , T-Lymphocytes/immunology , Blood Banks/trends , Cell Engineering/methods , Cell Engineering/trends , Genetic Engineering , Humans , Laboratories/standards , T-Lymphocytes/transplantation
20.
Biosci Biotechnol Biochem ; 80(7): 1243-53, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27305282

ABSTRACT

Cell surface display of proteins/peptides has been established based on mechanisms of localizing proteins to the cell surface. In contrast to conventional intracellular and extracellular (secretion) expression systems, this method, generally called an arming technology, is particularly effective when using yeasts as a host, because the control of protein folding that is often required for the preparation of proteins can be natural. This technology can be employed for basic and applied research purposes. In this review, I describe various strategies for the construction of engineered yeasts and provide an outline of the diverse applications of this technology to industrial processes such as the production of biofuels and chemicals, as well as bioremediation and health-related processes. Furthermore, this technology is suitable for novel protein engineering and directed evolution through high-throughput screening, because proteins/peptides displayed on the cell surface can be directly analyzed using intact cells without concentration and purification. Functional proteins/peptides with improved or novel functions can be created using this beneficial, powerful, and promising technique.


Subject(s)
Cell Engineering/methods , Gene Expression Regulation, Fungal , Membrane Proteins/genetics , Saccharomyces cerevisiae/genetics , Biodegradation, Environmental , Biofuels , Cell Engineering/trends , High-Throughput Screening Assays , Mating Factor/genetics , Mating Factor/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Peptide Library , Protein Folding , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...