Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 54
1.
J Biomech ; 128: 110770, 2021 11 09.
Article En | MEDLINE | ID: mdl-34628203

Duchenne muscular dystrophy (DMD) is still an incurable muscle degenerative disease; thus, numerous studies focused on novel therapeutic approaches. However, a simple assay of muscle function restoration remains needed. Herein, we used an oscillatory shear rheometer to evaluate changes in rheological properties of mouse muscles (tibialis anterior, TA) and their restoration upon autologous cell therapy by comparing the viscoelastic properties of normal, diseased and treated muscles. Amplitude sweep tests of muscle samples were performed under 20% compression over a range of shear strain between 0.01 and 2% and frequency of 1 rad/s. The samples were tested in plane-plane geometry and horizontal myofiber alignment. Typical linear viscoelastic region (LVER) patterns were found for each muscle type. For healthy muscles, a broad LVER between shear deformations (γ) of 0.013-0.62% was observed. The LVER of DMD mdx/SCID muscles was found at 0.14% to 0.46% shear deformation, and no shear dependence of storage (G') and loss (G") moduli at γ range changing from 0.034% to 0.26% was found for transplanted tissues. G'LVER and G"LVER moduli of healthy muscles were significantly higher than G'LVER and G"LVER of dystrophic tissues. Additionally, muscle resistance assessment by rheometer indicated that muscles transplanted with stem cells restored elastic properties to levels close to those of healthy muscles. Interestingly, histological staining and rheological data indicate that the loss factor is strongly related to structural changes of examined muscles.


Muscular Dystrophy, Duchenne , Animals , Cell- and Tissue-Based Therapy , Disease Models, Animal , Mice , Mice, Inbred mdx , Mice, SCID , Muscle, Skeletal , Muscular Dystrophy, Duchenne/therapy
2.
J Biotechnol ; 341: 103-112, 2021 Nov 20.
Article En | MEDLINE | ID: mdl-34560160

Integrative non-viral vectors such as transposons engineered to mediate targeted gene transfer into safe harbor sites in the genome may be a promising approach for the production of therapeutic proteins or for gene therapy in an efficient and secure way. In this context, we designed and evaluated two strategies for targeting the nuclear ribosomal DNA (rDNA) loci. One approach relied on the co-location of the transposase and transposon near transcriptionally active rDNA copies using a nucleolar localization signal (NoLS). Another one consisted of targeting the 18S-coding region in the rDNA loci using a NoLS-FokI-dCas9 endonuclease to perform targeted transgene knock-in. We show that integration into the rDNA of Chinese hamster ovary (CHO) cells can be achieved at a high frequency using the piggyBac transposon system, indicating that the rDNA is highly accessible for transposition. Consistently, rDNA-targeted transposition events were most frequently obtained when both the piggyBac transposon DNA and the transposase were nucleoli-targeted, yielding cells displaying stable and homogeneous expression of the transgene. This approach thus provides an alternative strategy to improve targeted transgene delivery and protein expression using CHO cells.


DNA Transposable Elements , Transposases , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Transposable Elements/genetics , DNA, Ribosomal , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Transposases/genetics , Transposases/metabolism
3.
Biotechnol Bioeng ; 117(12): 3628-3638, 2020 12.
Article En | MEDLINE | ID: mdl-32779734

A clonally derived (or "monoclonal") cell line is a cell population derived from a single progenitor cell. Clonally derived cell lines are required for many biotechnological applications. For instance, recombinant mammalian cells used to produce therapeutic proteins are expected by regulatory authorities to be clonally derived. Assurance of clonal derivation (or "clonality") is usually obtained from the characterization of the procedure used for cell cloning, for instance by assessing the success rate of single-cell sorting but not by assessing the cell line itself. We have developed a method to assess clonal derivation directly from the genetic makeup of cells. The genomic test of clonality is based on whole-genome sequencing and statistical analysis of single nucleotide variants. This approach quantifies the clonal fractions present in nonclonal samples and it provides a measure of the probability that a cell line is derived from a single cell. Upon experimental validation of the test, we show that it is highly accurate and that it can robustly detect minor clonal fractions of as little as 1% of the cell population. Moreover, we find that it is applicable to various cell line development protocols. This approach can simplify development protocols and shorten timelines while ensuring clonal derivation with high confidence.


Clone Cells , Polymorphism, Single Nucleotide , Whole Genome Sequencing , Animals , Biological Products , CHO Cells , Cricetulus
4.
Metab Eng ; 60: 77-86, 2020 07.
Article En | MEDLINE | ID: mdl-32247827

Maintaining a metabolic steady state is essential for an organism's fitness and survival when confronted with environmental stress, and metabolic imbalance can be reversed by exposing the organism to fasting. Here, we attempted to apply this physiological principle to mammalian cell cultures to improve cellular fitness and consequently their ability to express recombinant proteins. We showed that transient vitamin B5 deprivation, an essential cofactor of central cellular metabolism, can quickly and irreversibly affect mammalian cell growth and division. A selection method was designed that relies on mammalian cell dependence on vitamin B5 for energy production, using the co-expression of the B5 transporter SLC5A6 and a gene of interest. We demonstrated that vitamin B5 selection persistently activates peroxisome proliferator-activated receptors (PPAR), a family of transcription factors involved in energy homeostasis, thereby altering lipid metabolism, improving cell fitness and therapeutic protein production. Thus, stable PPAR activation may constitute a cellular memory of past deprivation state, providing increased resistance to further potential fasting events. In other words, our results imply that cultured cells, once exposed to metabolic starvation, may display an improved metabolic fitness as compared to non-exposed cells, allowing increased resistance to cellular stress.


Homeostasis , Pantothenic Acid/deficiency , Pantothenic Acid/metabolism , Recombinant Proteins/biosynthesis , Animals , CHO Cells , Cell Division , Cells, Cultured , Cricetinae , Cricetulus , Energy Metabolism , Genetic Vectors , Lipid Metabolism/physiology , PPAR alpha/biosynthesis , PPAR alpha/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Stress, Physiological , Symporters/metabolism
5.
Biotechnol Bioeng ; 117(4): 1101-1116, 2020 04.
Article En | MEDLINE | ID: mdl-31956982

Despite extensive research conducted to increase protein production from Chinese hamster ovary (CHO) cells, cellular bottlenecks often remain, hindering high yields. In this study, a transcriptomic analysis led to the identification of 32 genes that are consistently upregulated in high producer clones and thus might mediate high productivity. Candidate genes were associated with functions such as signaling, protein folding, cytoskeleton organization, and cell survival. We focused on two engineering targets, Erp27, which binds unfolded proteins and the Erp57 disulfide isomerase in the endoplasmic reticulum, and Foxa1, a pioneering transcription factor involved in organ development. Erp27 moderate overexpression increased production of an easy-to-express antibody, whereas Erp27 and Erp57 co-overexpression increased cell density, viability, and the yield of difficult-to-express proteins. Foxa1 overexpression increased cell density, cell viability, and easy- and difficult-to-express protein yields, whereas it decreased reactive oxygen species late in fed-batch cultures. Foxa1 overexpression upregulated two other candidate genes that increased the production of difficult- and/or easy-to-express proteins, namely Ca3, involved in protecting cells from oxidative stress, and Tagap, involved in signaling and cytoskeleton remodeling. Overall, several genes allowing to overcome CHO cell bottlenecks were identified, including Foxa1, which mediated multiple favorable metabolic changes that improve therapeutic protein yields.


Cell Engineering/methods , Hepatocyte Nuclear Factor 3-alpha , Recombinant Proteins , Animals , CHO Cells , Cell Survival , Cricetinae , Cricetulus , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Protein Folding , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Biotechnol Bioeng ; 117(4): 1117-1126, 2020 04.
Article En | MEDLINE | ID: mdl-31956990

In this study, we assessed the importance of cytoskeleton organization in the mammalian cells used to produce therapeutic proteins. Two cytoskeletal genes, Actin alpha cardiac muscle 1 (ACTC1) and a guanosine triphosphate GTPase-activating protein (TAGAP), were found to be upregulated in highly productive therapeutic protein-expressing Chinese hamster ovary (CHO) cells selected by the deprivation of vitamin B5. We report here that the overexpression of the ACTC1 protein was able to improve significantly recombinant therapeutic production, as well as to decrease the levels of toxic lactate metabolic by-products. ACTC1 overexpression was accompanied by altered as well as decreased polymerized actin, which was associated with high protein production by CHO cell cultured in suspension. We suggest that the depolymerization of actin and the possible modulation of integrin signaling, as well as changes in basal metabolism, may be driving the increase of protein secretion by CHO cells.


Actin Cytoskeleton , Actins , Recombinant Proteins , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/genetics , Actins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Biotechnol Bioeng ; 117(2): 466-485, 2020 02.
Article En | MEDLINE | ID: mdl-31631325

The Chinese hamster ovary (CHO) cells used to produce biopharmaceutical proteins are known to contain type-C endogenous retrovirus (ERV) sequences in their genome and to release retroviral-like particles. Although evidence for their infectivity is missing, this has raised safety concerns. As the genomic origin of these particles remained unclear, we characterized type-C ERV elements at the genome, transcriptome, and viral particle RNA levels. We identified 173 type-C ERV sequences clustering into three functionally conserved groups. Transcripts from one type-C ERV group were full-length, with intact open reading frames, and cognate viral genome RNA was loaded into retroviral-like particles, suggesting that this ERV group may produce functional viruses. CRISPR-Cas9 genome editing was used to disrupt the gag gene of the expressed type-C ERV group. Comparison of CRISPR-derived mutations at the DNA and RNA level led to the identification of a single ERV as the main source of the release of RNA-loaded viral particles. Clones bearing a Gag loss-of-function mutation in this ERV showed a reduction of RNA-containing viral particle release down to detection limits, without compromising cell growth or therapeutic protein production. Overall, our study provides a strategy to mitigate potential viral particle contaminations resulting from ERVs during biopharmaceutical manufacturing.


CHO Cells/virology , Endogenous Retroviruses , Mutagenesis, Site-Directed/methods , RNA, Viral , Virion/genetics , Animals , CRISPR-Cas Systems , Cricetinae , Cricetulus , Drug Contamination/prevention & control , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Editing , Genome, Viral/genetics , Loss of Function Mutation/genetics , RNA, Viral/genetics , RNA, Viral/metabolism
8.
Stem Cell Res ; 41: 101619, 2019 12.
Article En | MEDLINE | ID: mdl-31683098

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease caused by the lack of dystrophin in muscle fibers that is currently without curative treatment. Mesoangioblasts (MABs) are multipotent progenitor cells that can differentiate to a myogenic lineage and that can be used to express Dystrophin upon transplantation into muscles, in autologous gene therapy approaches. However, their fate in the muscle environment remains poorly characterized. Here, we investigated the differentiation fate of MABs following their transplantation in DMD murine muscles using a mass cytometry strategy. This allowed the identification and isolation of a fraction of MAB-derived cells presenting common properties with satellite muscle stem cells. This analysis also indicated that most cells did not undergo a myogenic differentiation path once in the muscle environment, limiting their capacity to restore dystrophin expression in transplanted muscles. We therefore assessed whether MAB treatment with cytokines and growth factors prior to engraftment may improve their myogenic fate. We identified a combination of such signals that ameliorates MABs capacity to undergo myogenic differentiation in vivo and to restore dystrophin expression upon engraftment in myopathic murine muscles.


Cell Differentiation , Multipotent Stem Cells , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Disease Models, Animal , Mice , Mice, Inbred mdx , Mice, SCID , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/transplantation
9.
DNA Repair (Amst) ; 82: 102691, 2019 10.
Article En | MEDLINE | ID: mdl-31476574

CRISPR technologies greatly foster genome editing in mammalian cells through site-directed DNA double strand breaks (DSBs). However, precise editing outcomes, as mediated by homologous recombination (HR) repair, are typically infrequent and outnumbered by undesired genome alterations. By using knockdown and overexpression studies in Chinese hamster ovary (CHO) cells as well as characterizing repaired DNA junctions, we found that efficient HR-mediated genome editing depends on alternative end-joining (alt-EJ) DNA repair activities, a family of incompletely characterized DNA repair pathways traditionally considered to oppose HR. This dependency was influenced by the CRISPR nuclease type and the DSB-to-mutation distance, but not by the DNA sequence surrounding the DSBs or reporter cell line. We also identified elevated Mre11 and Pari, and low Rad51 expression levels as the most rate-limiting factors for HR in CHO cells. Counteracting these three bottlenecks improved precise genome editing by up to 75%. Altogether, our study provides novel insights into the complex interplay of alt-EJ and HR repair pathways, highlighting their relevance for developing improved genome editing strategies.


DNA End-Joining Repair/genetics , Gene Editing , Homologous Recombination/genetics , Animals , CHO Cells , CRISPR-Associated Protein 9/metabolism , Cricetinae , Cricetulus , Deoxyribonucleases, Type II Site-Specific/metabolism
10.
Mol Ther ; 26(4): 1093-1108, 2018 04 04.
Article En | MEDLINE | ID: mdl-29503200

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.


Cell- and Tissue-Based Therapy , DNA Transposable Elements , Gene Transfer Techniques , Genetic Vectors/genetics , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Myoblasts/transplantation , Animals , Cell Line , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Dystrophin/genetics , Fluorescent Antibody Technique , Gene Dosage , Gene Expression , Gene Order , Genes, Reporter , Male , Mice , Mice, Inbred mdx , Mice, SCID , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Phenotype , Transgenes , Transplantation, Autologous
11.
Biotechnol Bioeng ; 114(8): 1791-1802, 2017 08.
Article En | MEDLINE | ID: mdl-28214330

We developed a method for the fast sorting and selection of mammalian cells expressing and secreting a protein at high levels. This procedure relies on cell capture using an automated microfluidic device handling antibody-coupled magnetic microparticles and on a timed release of the cells from the microparticles after capture. Using clinically compatible materials and procedures, we show that this approach is able to discriminate between cells that truly secrete high amounts of a protein from those that just display it at high levels on their surface without properly releasing it. When coupled to a cell colony imaging and picking device, this approach allowed the identification of CHO cell clones secreting a therapeutic protein at high levels that were not achievable without the cell sorting procedure. Biotechnol. Bioeng. 2017;114: 1791-1802. © 2017 Wiley Periodicals, Inc.


CHO Cells/cytology , CHO Cells/metabolism , Cell Separation/methods , Magnetite Nanoparticles/chemistry , Recombinant Proteins/metabolism , Animals , CHO Cells/radiation effects , Cricetulus , Magnetite Nanoparticles/radiation effects , Staining and Labeling/methods
12.
Biotechnol Bioeng ; 114(2): 384-396, 2017 02.
Article En | MEDLINE | ID: mdl-27575535

Untargeted plasmid integration into mammalian cell genomes remains a poorly understood and inefficient process. The formation of plasmid concatemers and their genomic integration has been ascribed either to non-homologous end-joining (NHEJ) or homologous recombination (HR) DNA repair pathways. However, a direct involvement of these pathways has remained unclear. Here, we show that the silencing of many HR factors enhanced plasmid concatemer formation and stable expression of the gene of interest in Chinese hamster ovary (CHO) cells, while the inhibition of NHEJ had no effect. However, genomic integration was decreased by the silencing of specific HR components, such as Rad51, and DNA synthesis-dependent microhomology-mediated end-joining (SD-MMEJ) activities. Genome-wide analysis of the integration loci and junction sequences validated the prevalent use of the SD-MMEJ pathway for transgene integration close to cellular genes, an effect shared with matrix attachment region (MAR) DNA elements that stimulate plasmid integration and expression. Overall, we conclude that SD-MMEJ is the main mechanism driving the illegitimate genomic integration of foreign DNA in CHO cells, and we provide a recombination engineering approach that increases transgene integration and recombinant protein expression in these cells. Biotechnol. Bioeng. 2017;114: 384-396. © 2016 The Authors. Biotechnology and Bioengineering published by Wiley Periodicals, Inc.


Chromatin/genetics , Genetic Engineering/methods , Matrix Attachment Regions/genetics , Recombinant Proteins/genetics , Recombination, Genetic/genetics , Animals , Antibodies/chemistry , Antibodies/genetics , Antibodies/metabolism , CHO Cells , Cricetinae , Cricetulus , Gene Knockdown Techniques , Humans , Plasmids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transgenes/genetics
13.
Nucleic Acids Res ; 44(6): e56, 2016 Apr 07.
Article En | MEDLINE | ID: mdl-26657630

DNA double stranded breaks (DSBs) are one of the most deleterious types of DNA lesions. The main pathways responsible for repairing these breaks in eukaryotic cells are homologous recombination (HR) and non-homologous end-joining (NHEJ). However, a third group of still poorly characterized DSB repair pathways, collectively termed microhomology-mediated end-joining (MMEJ), relies on short homologies for the end-joining process. Here, we constructed GFP reporter assays to characterize and distinguish MMEJ variant pathways, namely the simple MMEJ and the DNA synthesis-dependent (SD)-MMEJ mechanisms. Transfection of these assay vectors in Chinese hamster ovary (CHO) cells and characterization of the repaired DNA sequences indicated that while simple MMEJ is able to mediate relatively efficient DSB repair if longer microhomologies are present, the majority of DSBs were repaired using the highly error-prone SD-MMEJ pathway. To validate the involvement of DNA synthesis in the repair process, siRNA knock-down of different genes proposed to play a role in MMEJ were performed, revealing that the knock-down of DNA polymerase θ inhibited DNA end resection and repair through simple MMEJ, thus favoring the other repair pathway. Overall, we conclude that this approach provides a convenient assay to study MMEJ-related DNA repair pathways.


Biological Assay , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/genetics , DNA/metabolism , Animals , Base Sequence , CHO Cells , Cricetulus , DNA/chemistry , DNA/genetics , DNA-Directed DNA Polymerase/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homologous Recombination , Molecular Sequence Data , Plasmids/chemistry , Plasmids/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Homology, Nucleic Acid , Transfection , DNA Polymerase theta
14.
Cell Cycle ; 14(17): 2853-61, 2015.
Article En | MEDLINE | ID: mdl-26125600

Eukaryotic cells respond to DNA breaks, especially double-stranded breaks (DSBs), by activating the DNA damage response (DDR), which encompasses DNA repair and cell cycle checkpoint signaling. The DNA damage signal is transmitted to the checkpoint machinery by a network of specialized DNA damage-recognizing and signal-transducing molecules. However, recent evidence suggests that DNA repair proteins themselves may also directly contribute to the checkpoint control. Here, we investigated the role of homologous recombination (HR) proteins in normal cell cycle regulation in the absence of exogenous DNA damage. For this purpose, we used Chinese Hamster Ovary (CHO) cells expressing the Fluorescent ubiquitination-based cell cycle indicators (Fucci). Systematic siRNA-mediated knockdown of HR genes in these cells demonstrated that the lack of several of these factors alters cell cycle distribution, albeit differentially. The knock-down of MDC1, Rad51 and Brca1 caused the cells to arrest in the G2 phase, suggesting that they may be required for the G2/M transition. In contrast, inhibition of the other HR factors, including several Rad51 paralogs and Rad50, led to the arrest in the G1/G0 phase. Moreover, reduced expression of Rad51B, Rad51C, CtIP and Rad50 induced entry into a quiescent G0-like phase. In conclusion, the lack of many HR factors may lead to cell cycle checkpoint activation, even in the absence of exogenous DNA damage, indicating that these proteins may play an essential role both in DNA repair and checkpoint signaling.


Cell Cycle Proteins/physiology , Cell Cycle/physiology , Homologous Recombination/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Recombinant Proteins
15.
Biotechnol J ; 10(7): 967-78, 2015 Jul.
Article En | MEDLINE | ID: mdl-26099730

Successful generation of high producing cell lines requires the generation of cell clones expressing the recombinant protein at high levels and the characterization of the clones' ability to maintain stable expression levels. The use of cis-acting epigenetic regulatory elements that improve this otherwise long and uncertain process has revolutionized recombinant protein production. Here we review and discuss new insights into the molecular mode of action of the matrix attachment regions (MARs) and ubiquitously-acting chromatin opening elements (UCOEs), i.e. cis-acting elements, and how these elements are being used to improve recombinant protein production. These elements can help maintain the chromatin environment of the transgene genomic integration locus in a transcriptionally favorable state, which increases the numbers of positive clones and the transgene expression levels. Moreover, the high producing clones tend to be more stable in long-term cultures even in the absence of selection pressure. Therefore, by increasing the probability of isolating a high producing clone, as well as by increasing transcription efficiency and stability, these elements can significantly reduce the time and cost required for producing large quantities of recombinant proteins.


CHO Cells , Epigenesis, Genetic , Recombinant Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/genetics , Cricetinae , Cricetulus , Gene Silencing , Matrix Attachment Regions/genetics , Recombinant Proteins/biosynthesis , Transgenes/genetics
16.
Liver Int ; 35(4): 1185-94, 2015 Apr.
Article En | MEDLINE | ID: mdl-25293436

BACKGROUND & AIMS: Knockout studies of the murine Nuclear Factor I-C (NFI-C) transcription factor revealed abnormal skin wound healing and growth of its appendages, suggesting a role in controlling cell proliferation in adult regenerative processes. Liver regeneration following partial hepatectomy (PH) is a well-established regenerative model whereby changes elicited in hepatocytes lead to their rapid and phased proliferation. Although NFI-C is highly expressed in the liver, no hepatic function was yet established for this transcription factor. This study aimed to determine whether NFI-C may play a role in hepatocyte proliferation and liver regeneration. METHODS: Liver regeneration and cell proliferation pathways following two-thirds PH were investigated in NFI-C knockout (ko) and wild-type (wt) mice. RESULTS: We show that the absence of NFI-C impaired hepatocyte proliferation because of plasminogen activator I (PAI-1) overexpression and the subsequent suppression of urokinase plasminogen activator (uPA) activity and hepatocyte growth factor (HGF) signalling, a potent hepatocyte mitogen. This indicated that NFI-C first acts to promote hepatocyte proliferation at the onset of liver regeneration in wt mice. The subsequent transient down regulation of NFI-C, as can be explained by a self-regulatory feedback loop with transforming growth factor beta 1 (TGF-ß1), may limit the number of hepatocytes entering the first wave of cell division and/or prevent late initiations of mitosis. CONCLUSION: NFI-C acts as a regulator of the phased hepatocyte proliferation during liver regeneration.


Cell Proliferation , Liver Regeneration , Liver/metabolism , NFI Transcription Factors/metabolism , Animals , Feedback, Physiological , Gene Expression Regulation , Genotype , Hepatectomy/methods , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Liver/pathology , Liver/physiopathology , Liver/surgery , Male , Mice, Inbred C57BL , Mice, Knockout , NFI Transcription Factors/deficiency , NFI Transcription Factors/genetics , Phenotype , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
17.
Stem Cell Res ; 13(3 Pt A): 390-403, 2014 Nov.
Article En | MEDLINE | ID: mdl-25310255

An emerging therapeutic approach for Duchenne muscular dystrophy is the transplantation of autologous myogenic progenitor cells genetically modified to express dystrophin. The use of this approach is challenged by the difficulty in maintaining these cells ex vivo while keeping their myogenic potential, and ensuring sufficient transgene expression following their transplantation and myogenic differentiation in vivo. We investigated the use of the piggyBac transposon system to achieve stable gene expression when transferred to cultured mesoangioblasts and into murine muscles. Without selection, up to 8% of the mesoangioblasts expressed the transgene from 1 to 2 genomic copies of the piggyBac vector. Integration occurred mostly in intergenic genomic DNA and transgene expression was stable in vitro. Intramuscular transplantation of mouse Tibialis anterior muscles with mesoangioblasts containing the transposon led to sustained myofiber GFP expression in vivo. In contrast, the direct electroporation of the transposon-donor plasmids in the mouse Tibialis muscles in vivo did not lead to sustained transgene expression despite molecular evidence of piggyBac transposition in vivo. Together these findings provide a proof-of-principle that piggyBac transposon may be considered for mesoangioblast cell-based therapies of muscular dystrophies.


Cell- and Tissue-Based Therapy/methods , Chromosomes, Artificial, Bacterial/metabolism , Gene Transfer Techniques , Muscle, Skeletal/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Microscopy, Fluorescence , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology
18.
Nanomedicine (Lond) ; 9(4): 393-406, 2014 Apr.
Article En | MEDLINE | ID: mdl-24910872

AIM: Atomic force microscopy nanoindentation of myofibers was used to assess and quantitatively diagnose muscular dystrophies from human patients. MATERIALS & METHODS: Myofibers were probed from fresh or frozen muscle biopsies from human dystrophic patients and healthy volunteers, as well as mice models, and Young's modulus stiffness values were determined. RESULTS: Fibers displaying abnormally low mechanical stability were detected in biopsies from patients affected by 11 distinct muscle diseases, and Young's modulus values were commensurate to the severity of the disease. Abnormal myofiber resistance was also observed from consulting patients whose muscle condition could not be detected or unambiguously diagnosed otherwise. DISCUSSION & CONCLUSION: This study provides a proof-of-concept that atomic force microscopy yields a quantitative read-out of human muscle function from clinical biopsies, and that it may thereby complement current muscular dystrophy diagnosis.


Microscopy, Atomic Force/methods , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Adolescent , Adult , Aged , Animals , Biomechanical Phenomena , Child , Elastic Modulus , Female , Humans , Male , Mice
19.
J Cell Sci ; 127(Pt 15): 3240-56, 2014 Aug 01.
Article En | MEDLINE | ID: mdl-24895400

Here, we identify a role for the matrilin-2 (Matn2) extracellular matrix protein in controlling the early stages of myogenic differentiation. We observed Matn2 deposition around proliferating, differentiating and fusing myoblasts in culture and during muscle regeneration in vivo. Silencing of Matn2 delayed the expression of the Cdk inhibitor p21 and of the myogenic genes Nfix, MyoD and Myog, explaining the retarded cell cycle exit and myoblast differentiation. Rescue of Matn2 expression restored differentiation and the expression of p21 and of the myogenic genes. TGF-ß1 inhibited myogenic differentiation at least in part by repressing Matn2 expression, which inhibited the onset of a positive-feedback loop whereby Matn2 and Nfix activate the expression of one another and activate myoblast differentiation. In vivo, myoblast cell cycle arrest and muscle regeneration was delayed in Matn2(-/-) relative to wild-type mice. The expression levels of Trf3 and myogenic genes were robustly reduced in Matn2(-/-) fetal limbs and in differentiating primary myoblast cultures, establishing Matn2 as a key modulator of the regulatory cascade that initiates terminal myogenic differentiation. Our data thus identify Matn2 as a crucial component of a genetic switch that modulates the onset of tissue repair.


Extracellular Matrix/metabolism , Matrilin Proteins/metabolism , Muscles/physiology , Myoblasts/physiology , Necrosis/therapy , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation/genetics , Elapid Venoms/administration & dosage , Humans , Matrilin Proteins/genetics , Mice , Mice, Knockout , Muscle Development/genetics , Muscles/pathology , Necrosis/chemically induced , Rats , Rats, Wistar , Regeneration/genetics , Time Factors
20.
J Neuroimmunol ; 270(1-2): 61-6, 2014 May 15.
Article En | MEDLINE | ID: mdl-24657030

Chronic-inflammatory demyelinating polyneuropathy (CIDP) is an immune-mediated disease with no known biomarkers for diagnosing the disease or assessing its prognosis. We performed transcriptional profiling microarray analysis on skin punch biopsies from 20 CIDP patients and 17 healthy controls to identify disease-associated gene expression changes. We demonstrate changes in expression of genes involved in immune and chemokine regulation, growth and repair. We also found a combination of two upregulated genes that can be proposed as a novel biomarker of the disorder.


Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/genetics , Transcriptome , Adult , Aged , Biomarkers/analysis , Biopsy , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Skin
...