Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Elife ; 102021 01 13.
Article in English | MEDLINE | ID: mdl-33438576

ABSTRACT

Generating mammalian cells with specific mitochondrial DNA (mtDNA)-nuclear DNA (nDNA) combinations is desirable but difficult to achieve and would be enabling for studies of mitochondrial-nuclear communication and coordination in controlling cell fates and functions. We developed 'MitoPunch', a pressure-driven mitochondrial transfer device, to deliver isolated mitochondria into numerous target mammalian cells simultaneously. MitoPunch and MitoCeption, a previously described force-based mitochondrial transfer approach, both yield stable isolated mitochondrial recipient (SIMR) cells that permanently retain exogenous mtDNA, whereas coincubation of mitochondria with cells does not yield SIMR cells. Although a typical MitoPunch or MitoCeption delivery results in dozens of immortalized SIMR clones with restored oxidative phosphorylation, only MitoPunch can produce replication-limited, non-immortal human SIMR clones. The MitoPunch device is versatile, inexpensive to assemble, and easy to use for engineering mtDNA-nDNA combinations to enable fundamental studies and potential translational applications.


Mitochondria are specialized structures within cells that generate vital energy and biological building blocks. Mitochondria have a double membrane and contain many copies of their own circular DNA (mitochondrial DNA), which include the blueprints to create just thirteen essential mitochondrial proteins. Like all genetic material, mitochondrial DNA can become damaged or mutated, and these changes can be passed on to offspring. Some of these alterations are linked to severe and debilitating diseases. Both the double membrane of the mitochondria and their high number of DNA copies make treating such diseases difficult. A successful therapy must be capable of correcting almost every copy of mitochondrial DNA. However, the multiple copies of mitochondrial DNA create a problem for genetic research as current techniques are unable to reliably introduce particular mitochondrial mutations to all types of human cells to investigate how they may alter cell function. Sercel, Patananan et al. have developed a method to deliver new mitochondria into thousands of cells at the same time. This technique, called MitoPunch, uses a pressure-driven device to propel mitochondria taken from donor cells into recipient cells without mitochondrial DNA to reestablish their function. Using human cancer cells and healthy skin cells that lack mitochondrial DNA, Sercel, Patananan et al. showed that cells that received mitochondria retained the new mitochondrial DNA. The technique uses readily accessible parts, meaning it can be performed quickly and inexpensively in any laboratory. It further only requires a small amount of donor starting material, meaning that even precious samples with limited material could be used as mitochondrial donors. This new technique has several important potential applications for mitochondrial DNA research. It could be used in the lab to create large numbers of cell lineswith known mutations in the mitochondrial DNA to establish new systems that test drugs or probe the interaction between mitochondrial and nuclear DNA. It could be used to study a broad spectrum of biological questions since mitochondrial function is essential for several processes required for life. Critically, it could also be used as a starting point to develop next-generation therapies capable of treating inherited mitochondrial genetic diseases in severely affected patients.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Mice
2.
Lab Chip ; 21(6): 1049-1060, 2021 03 21.
Article in English | MEDLINE | ID: mdl-33313615

ABSTRACT

In molecular and cellular biological research, cell isolation and sorting are required for accurate investigation of cell populations of specific physical or biological characteristics. By employing unique cell properties to distinguish between heterogeneous cell populations, rapid and accurate sorting with high efficiency is possible. Dielectrophoresis-based cell manipulation has significant promise for separation of cells based on their physical properties and is used in diverse areas ranging from cellular diagnostics to therapeutic applications. In this study, we present a microfluidic device that can achieve label-free and size-based cell separation with high size differential resolution from a mono-cellular population or complex sample matrices. It was realized by using the tunnel dielectrophoresis (TDEP) technique to manipulate the spatial position of individual cells three dimensionally with high resolution. Cells were processed in high speed flows in high ionic strength buffers. A mixture of different sizes of polystyrene micro-particles with a size difference as small as 1 µm can be separated with high purity (>90%). For the first time, high-pass, low-pass, and band-pass filtering within a mono-cellular mammalian cell population were demonstrated with a tunable bandwidth as small as 3 µm. In addition, leukocyte subtype separation was demonstrated by sorting monocytes out of peripheral blood mononuclear cells (PBMCs) from whole blood with high purity (>85%). Its ability to deliver real-time adjustable cut-off threshold size-based cell sorting and its capability to provide an arbitrary cell size pick-up band could potentially enable many research and clinical applications.


Subject(s)
Lab-On-A-Chip Devices , Leukocytes, Mononuclear , Animals , Cell Separation , Monocytes , Polystyrenes
3.
Cell Rep ; 33(13): 108562, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378680

ABSTRACT

Generating mammalian cells with desired mitochondrial DNA (mtDNA) sequences is enabling for studies of mitochondria, disease modeling, and potential regenerative therapies. MitoPunch, a high-throughput mitochondrial transfer device, produces cells with specific mtDNA-nuclear DNA (nDNA) combinations by transferring isolated mitochondria from mouse or human cells into primary or immortal mtDNA-deficient (ρ0) cells. Stable isolated mitochondrial recipient (SIMR) cells isolated in restrictive media permanently retain donor mtDNA and reacquire respiration. However, SIMR fibroblasts maintain a ρ0-like cell metabolome and transcriptome despite growth in restrictive media. We reprogrammed non-immortal SIMR fibroblasts into induced pluripotent stem cells (iPSCs) with subsequent differentiation into diverse functional cell types, including mesenchymal stem cells (MSCs), adipocytes, osteoblasts, and chondrocytes. Remarkably, after reprogramming and differentiation, SIMR fibroblasts molecularly and phenotypically resemble unmanipulated control fibroblasts carried through the same protocol. Thus, our MitoPunch "pipeline" enables the production of SIMR cells with unique mtDNA-nDNA combinations for additional studies and applications in multiple cell types.


Subject(s)
Cellular Reprogramming , Fibroblasts/metabolism , Gene Transfer Techniques , High-Throughput Screening Assays/methods , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/transplantation , Animals , Cell Differentiation , Cell Line , DNA, Mitochondrial/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Metabolome , Mice , Mice, Inbred C57BL , Transcriptome
4.
Sci Rep ; 9(1): 8457, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186484

ABSTRACT

The biology of tumor-derived exosomes (TEX) is only partially understood and much remains to be studied in order to define the effect that the tumor microenvironment or the activation of tumor cells exerts on their composition and functions. Increased expression and activity of toll-like receptor 4 (TLR4) in chronic infectious and inflammatory conditions is related with cancer progression: its activation induces an inflammatory signaling that increases the tumorigenic potential of cancer cells promoting their immune evasion. We investigated the immune modulatory properties of TEX released upon cell TLR4 activation, and we found that, although differences were observed depending on the type of the tumor, the treatment influences TEX composition and boosts their immunosuppressive ability. Our results suggest that the activation of TLR4 supports tumor progression by stimulating the release of more effective immunosuppressive exosomes, which allow tumor cells to escape immune surveillance and probably even play a role in the metastatic process.


Subject(s)
Exosomes/genetics , Neoplasms/genetics , Toll-Like Receptor 4/genetics , Tumor Microenvironment/immunology , Cell Line, Tumor , Exosomes/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/metabolism , MicroRNAs/genetics , Neoplasms/classification , Neoplasms/immunology , Neoplasms/pathology , Signal Transduction , Toll-Like Receptor 4/immunology , Tumor Microenvironment/genetics
5.
Sci Rep ; 8(1): 13325, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30190615

ABSTRACT

The predominant mechanism by which adipose mesenchymal stem cells (AMSCs) participate to tissue repair is through a paracrine activity and their communication with the inflammatory microenvironment is essential part of this process. This hypothesis has been strengthened by the recent discovery that stem cells release not only soluble factors but also extracellular vesicles, which elicit similar biological activity to the stem cells themselves. We demonstrated that the treatment with inflammatory cytokines increases the immunosuppressive and anti-inflammatory potential of AMSCs-derived exosomes, which acquire the ability to shift macrophages from M1 to M2 phenotype by shuttling miRNA regulating macrophages polarization. This suggests that the immunomodulatory properties of AMSCs-derived exosomes may be not constitutive, but are instead induced by the inflammatory microenvironment.


Subject(s)
Adipose Tissue/immunology , Cellular Microenvironment/immunology , Exosomes/immunology , Immune Tolerance , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Humans , Inflammation/immunology
6.
Mediators Inflamm ; 2017: 4814987, 2017.
Article in English | MEDLINE | ID: mdl-28634420

ABSTRACT

The purpose of this study is to characterize synovial fluid- (SF-) derived exosomes of patients with gonarthrosis comparing two methods of isolation and to investigate their immune regulatory properties. Extracellular vesicles (EVs) have been isolated from inflamed SF by polymer precipitation method and quantified by Exocet kit and by nanoparticle tracking analysis. Vesicles expressed all the specific exosomal markers by immunoblot and FACS. After isolation with Exoquick, a relevant contamination by immune complexes was detected, which required further magnetic bead-based purification to remove. SF-derived exosomes significantly stimulated the release of several inflammatory cytokines and chemokines and metalloproteinases by M1 macrophages but did not influence the expression of CD80 and CD86 costimulatory molecules. In conclusion, we characterized purified exosomes isolated from inflamed SF and demonstrate that purified exosomes are functionally active in their ability to stimulate the release of proinflammatory factors from M1 macrophages. Our data indicate that SF-derived exosomes from gonarthrosis patients play a role in disease progression.


Subject(s)
Exosomes/metabolism , Osteoarthritis/metabolism , Synovial Fluid/metabolism , Aged , Aged, 80 and over , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Exosomes/chemistry , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Female , Humans , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Male , Middle Aged
7.
Org Lett ; 17(14): 3608-11, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26144210

ABSTRACT

An analogue 2 of Brasilicardin A, 1 (BraA), a potent immunosuppressive and cytotoxic agent, was synthesized in which the natural tricyclic skeleton was replaced with a synthetically more accessible substituted tetrahydronaphthalene core. BraA, this analogue (BraL), and cyclosporine A were tested for their ability to inhibit the proliferation of human T cells upon CD3/CD28 activation. Although BraL did not impact T cell activation over the dose range tested, this study shows the inhibitory activity of BraA on human T cells for the first time.


Subject(s)
Aminoglycosides/chemical synthesis , Aminoglycosides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cyclosporine/chemistry , Immunosuppressive Agents/pharmacology , Naphthalenes/chemistry , T-Lymphocytes/drug effects , Aminoglycosides/chemistry , Antineoplastic Agents/chemistry , CD28 Antigens , CD3 Complex , Humans , Immunosuppressive Agents/chemistry , Molecular Structure
8.
Biomed Opt Express ; 4(9): 1618-25, 2013.
Article in English | MEDLINE | ID: mdl-24049682

ABSTRACT

Flow cytometry is a powerful tool for cell counting and biomarker detection in biotechnology and medicine especially with regards to blood analysis. Standard flow cytometers perform cell type classification both by estimating size and granularity of cells using forward- and side-scattered light signals and through the collection of emission spectra of fluorescently-labeled cells. However, cell surface labeling as a means of marking cells is often undesirable as many reagents negatively impact cellular viability or provide activating/inhibitory signals, which can alter the behavior of the desired cellular subtypes for downstream applications or analysis. To eliminate the need for labeling, we introduce a label-free imaging-based flow cytometer that measures size and cell protein concentration simultaneously either as a stand-alone instrument or as an add-on to conventional flow cytometers. Cell protein concentration adds a parameter to cell classification, which improves the specificity and sensitivity of flow cytometers without the requirement of cell labeling. This system uses coherent dispersive Fourier transform to perform phase imaging at flow speeds as high as a few meters per second.

9.
Immunology ; 122(4): 522-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17635609

ABSTRACT

Distinct CD4(+) T-cell epitopes within the same protein can be optimally processed and loaded into major histocompatibility complex (MHC) class II molecules in disparate endosomal compartments. The CD1 protein isoforms traffic to these same endosomal compartments as directed by unique cytoplasmic tail sequences, therefore we reasoned that antigen/CD1 chimeras containing the different CD1 cytoplasmic tail sequences could optimally target antigens to the MHC class II antigen presentation pathway. Evaluation of trafficking patterns revealed that all four human CD1-derived targeting sequences delivered antigen to the MHC class II antigen presentation pathway, to early/recycling, early/sorting and late endosomes/lysosomes. There was a preferential requirement for different CD1 targeting sequences for the optimal presentation of an MHC class II epitope in the following hierarchy: CD1b > CD1d = CD1c > > > CD1a or untargeted antigen. Therefore, the substitution of the CD1 ectodomain with heterologous proteins results in their traffic to distinct intracellular locations that intersect with MHC class II and this differential distribution leads to specific functional outcomes with respect to MHC class II antigen presentation. These findings may have implications in designing DNA vaccines, providing a greater variety of tools to generate T-cell responses against microbial pathogens or tumours.


Subject(s)
Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Chaperonin 10/immunology , Dose-Response Relationship, Immunologic , Endosomes/immunology , Green Fluorescent Proteins , HeLa Cells , Humans , Interferon-gamma/immunology , Mycobacterium leprae/immunology , Recombinant Fusion Proteins/immunology , Recombinant Proteins , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...