Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
J Ovarian Res ; 17(1): 66, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38504307

BACKGROUND: Quiescin sulfhydryl oxidase 2 (QSOX2) is a flavin adenine dinucleotide-dependent sulfhydryl oxidase that is known to be involved in protein folding, cell growth regulation, and redox state modification through oxidative activities. Earlier studies demonstrated the tissue and cellular localization of QSOX2 in the male reproductive tract, as well as the highly-regulated mechanism of QSOX2 protein synthesis and expression through the coordinated action of testosterone and epididymal-enriched amino acid, glutamate. However, the presence and the functions of QSOX2 in female reproduction are unknown. In this study, we applied the Cre-loxP gene manipulation system to generate the heterozygous and homozygous Qsox2 knockout mice and examined its effects on ovarian function. RESULTS: We demonstrated that QSOX2 was detected in the follicle-supporting cells (granulosa and cumulus cells) of ovarian follicles of all stages but was absent in the corpus luteum, suggesting its supportive role in folliculogenesis. In comparison with reproductive organogenesis in wild-type mice, there was no difference in testicular and epididymal structure in male Qsox2 knockout; however, Qsox2 knockout disrupted the regular ovulation process in female mice as a drastic decrease in the formation of the corpus luteum was detected, and no pregnancy was achieved when mating males with homozygous Qsox2 knockout females. RNAseq analyses further revealed that Qsox2 knockout altered critical signaling pathways and genes that are responsible for maintaining ovarian functions. CONCLUSION: Our data demonstrated for the first time that Qsox2 is critical for ovarian function in mice.


Granulosa Cells , Oxidoreductases , Tamoxifen , Female , Mice , Male , Animals , Granulosa Cells/metabolism , Tamoxifen/pharmacology , Tamoxifen/metabolism , Ovary , Ovulation , Mice, Knockout
2.
Am J Pathol ; 193(6): 755-768, 2023 06.
Article En | MEDLINE | ID: mdl-36868469

miR-194 is abundantly expressed in hepatocytes, and its depletion increases hepatic resistance to acetaminophen-induced acute injuries. In this study, the biological role of miR-194 in cholestatic liver injury was investigated by using miR-194/miR-192 cluster liver-specific knockout (LKO) mice, in which no liver injuries or metabolic disorders were predisposed. Bile duct ligation (BDL) and 1-naphthyl isothiocyanate (ANIT) were applied to LKO and matched control wild-type (WT) mice to induce hepatic cholestasis. Periportal liver damage, mortality rate, and liver injury biomarkers in LKO mice were significantly less than in WT mice after BDL and ANIT injection. Intrahepatic bile acid level was significantly lower in the LKO liver within 48 hours of BDL- and ANIT-induced cholestasis compared with WT. Western blot analysis showed that ß-catenin (CTNNB1) signaling and genes involved in cellular proliferation were activated in BDL- and ANIT-treated mice. The expression levels of cholesterol 7 alpha-hydroxylase (CYP7A1), pivotal in bile synthesis, and its upstream regulator hepatocyte nuclear factor 4α were reduced in primary LKO hepatocytes and liver tissues compared with WT. The knockdown of miR-194 using miRNA inhibitors reduced CYP7A1 expression in WT hepatocytes. In contrast, the knockdown of CTNNB1 and overexpression of miR-194, but not miR-192, in LKO hepatocytes and AML12 cells increased CYP7A1 expression. In conclusion, the results suggest that the loss of miR-194 ameliorates cholestatic liver injury and may suppress CYP7A1 expression via activation of CTNNB1 signaling.


Cholestasis , Liver Diseases , Mice , Animals , beta Catenin/metabolism , Cholestasis/genetics , Cholestasis/metabolism , Liver Diseases/metabolism , Hepatocytes/metabolism , Bile Acids and Salts/metabolism , Cholesterol/metabolism
3.
Stem Cell Res ; 69: 103068, 2023 06.
Article En | MEDLINE | ID: mdl-36933359

Pathogenic variants of the WFS1 gene can cause recessive-inherited Wolfram syndrome or dominant-inherited Wolfram-like syndrome with optic atrophy and hearing impairment. Using the Sendai virus delivery system, we generated induced pluripotent stem cells from the peripheral blood mononuclear cells of a female patient with the WFS1 pathogenic variant c.2051C > T (p.Ala684Val). The resulting induced pluripotent stem cells exhibited a normal karyotype and pluripotency, as confirmed using immunofluorescence staining, and differentiated into three germ layers in vivo. This cellular model provides a useful platform for investigating the pathogenic mechanisms of both blindness and deafness related to WFS1 variants.


Hearing Loss , Induced Pluripotent Stem Cells , Wolfram Syndrome , Humans , Female , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Hearing Loss/genetics , Wolfram Syndrome/genetics , Wolfram Syndrome/pathology , Mutation
4.
Stem Cell Res ; 64: 102903, 2022 10.
Article En | MEDLINE | ID: mdl-36075118

Pathogenic variants of OPA1 have been associated with autosomal dominant optic atrophy (DOA), leading to optic, auditory, and other sensorineural neuropathies and myopathies. Using the Sendai virus delivery system, we generated induced pluripotent stem cells from the peripheral blood mononuclear cells of a female patient with the OPA1 pathogenic variant c.1468T>C (p.Cys490Arg). The resulting induced pluripotent stem cells exhibited a normal karyotype and pluripotency, as confirmed using immunofluorescence staining, and differentiated into three germ layers in vivo. This cellular model is a useful platform for investigating the pathogenic mechanisms of both blindness and deafness related to OPA1 variants.


Hearing Loss , Induced Pluripotent Stem Cells , Optic Atrophy, Autosomal Dominant , Humans , Female , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Optic Atrophy, Autosomal Dominant/pathology , Hearing Loss/genetics , GTP Phosphohydrolases/genetics
5.
Biomaterials ; 285: 121471, 2022 06.
Article En | MEDLINE | ID: mdl-35490561

Dissemination is an ominous feature of cancer to cause poor prognosis. Formation of tumor satellites is the first step, which is closely interdependent on the factors originating from surrounding niche. Because of lacking appropriate modeling, most studies focusing on cancer-environmental interaction depend on the static pathological analyses of specimens. Using the biomimicking system capable of inducing tumor satellite formation in vitro, the niche factors were explored for their influence on cancer budding. Our results demonstrated the real-time dynamic of tumor satellite formation of head and neck squamous cell carcinoma (HNSCC) that was significantly affected by these niche factors. Hypoxia reduced the incidence and migratory distance of HNSCC tumor satellites via decreasing the nuclear localization of intracellular domain of E-cadherin (iEcad) and ß-catenin. Three-dimensional (3D) architecture and collagen were essential for mediating hypoxia effects on nuclear translocation and signaling transduction of iEcad and ß-catenin in the HNSCC cells. The findings were further confirmed by the compatible in vivo findings of the cancer specimens. This approach revealed the effects of oxygen and extracellular matrices on tumor satellites. The cellular behaviors and molecular dynamics were along a phenotypic spectrum that conferred unique change for specific cancer populations in response to the altered microenvironment. The current study provides a platform to recapitulate the process of cancer budding, and a way to investigate the mutual interaction between the cancer cells and their surrounding microenvironment.


Head and Neck Neoplasms , beta Catenin , Cadherins , Cell Line, Tumor , Collagen , Humans , Hypoxia , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment/physiology
6.
Gut ; 71(9): 1876-1891, 2022 09.
Article En | MEDLINE | ID: mdl-34933915

BACKGROUND AND OBJECTIVES: Liver fibrosis (LF) occurs following chronic liver injuries. Currently, there is no effective therapy for LF. Recently, we identified thioredoxin domain containing 5 (TXNDC5), an ER protein disulfide isomerase (PDI), as a critical mediator of cardiac and lung fibrosis. We aimed to determine if TXNDC5 also contributes to LF and its potential as a therapeutic target for LF. DESIGN: Histological and transcriptome analyses on human cirrhotic livers were performed. Col1a1-GFPTg , Alb-Cre;Rosa26-tdTomato and Tie2-Cre/ERT2;Rosa26-tdTomato mice were used to determine the cell type(s) where TXNDC5 was induced following liver injury. In vitro investigations were conducted in human hepatic stellate cells (HSCs). Col1a2-Cre/ERT2;Txndc5fl/fl (Txndc5cKO ) and Alb-Cre;Txndc5fl/fl (Txndc5Hep-cKO ) mice were generated to delete TXNDC5 in HSCs and hepatocytes, respectively. Carbon tetrachloride treatment and bile duct ligation surgery were employed to induce liver injury/fibrosis in mice. The extent of LF was quantified using histological, imaging and biochemical analyses. RESULTS: TXNDC5 was upregulated markedly in human and mouse fibrotic livers, particularly in activated HSC at the fibrotic foci. TXNDC5 was induced by transforming growth factor ß1 (TGFß1) in HSCs and it was both required and sufficient for the activation, proliferation, survival and extracellular matrix production of HSC. Mechanistically, TGFß1 induces TXNDC5 expression through increased ER stress and ATF6-mediated transcriptional regulation. In addition, TXNDC5 promotes LF by redox-dependent JNK and signal transducer and activator of transcription 3 activation in HSCs through its PDI activity, activating HSCs and making them resistant to apoptosis. HSC-specific deletion of Txndc5 reverted established LF in mice. CONCLUSIONS: ER protein TXNDC5 promotes LF through redox-dependent HSC activation, proliferation and excessive extracellular matrix production. Targeting TXNDC5, therefore, could be a potential novel therapeutic strategy to ameliorate LF.


Hepatic Stellate Cells , Liver Cirrhosis , Animals , Carbon Tetrachloride/adverse effects , Carbon Tetrachloride/metabolism , Fibrosis , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Protein Disulfide-Isomerases/adverse effects , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
7.
Stem Cell Res Ther ; 12(1): 508, 2021 09 22.
Article En | MEDLINE | ID: mdl-34551822

BACKGROUND: The c.G6055A (p.G2019S) mutation in leucine-rich repeat kinase 2 (LRRK2) is the most prevalent genetic cause of Parkinson's disease (PD). CRISPR/Cas9-mediated genome editing by homology-directed repair (HDR) has been applied to correct the mutation but may create small insertions and deletions (indels) due to double-strand DNA breaks. Adenine base editors (ABEs) could convert targeted A·T to G·C in genomic DNA without double-strand breaks. However, the correction efficiency of ABE in LRRK2 c.G6055A (p.G2019S) mutation remains unknown yet. This study aimed to compare the mutation correction efficiencies and off-target effects between HDR and ABEs in induced pluripotent stem cells (iPSCs) carrying LRRK2 c.G6055A (p.G2019S) mutation. METHODS: A set of mutation-corrected isogenic lines by editing the LRRK2 c.G6055A (p.G2019S) mutation in a PD patient-derived iPSC line using HDR or ABE were established. The mutation correction efficacies, off-target effects, and indels between HDR and ABE were compared. Comparative transcriptomic and proteomic analyses between the LRRK2 p.G2019S iPSCs and isogenic control cells were performed to identify novel molecular targets involved in LRRK2-parkinsonism pathways. RESULTS: ABE had a higher correction rate (13/53 clones, 24.5%) than HDR (3/47 clones, 6.4%). Twenty-seven HDR clones (57.4%), but no ABE clones, had deletions, though 14 ABE clones (26.4%) had off-target mutations. The corrected isogenic iPSC-derived dopaminergic neurons exhibited reduced LRRK2 kinase activity, decreased phospho-α-synuclein expression, and mitigated neurite shrinkage and apoptosis. Comparative transcriptomic and proteomic analysis identified different gene expression patterns in energy metabolism, protein degradation, and peroxisome proliferator-activated receptor pathways between the mutant and isogenic control cells. CONCLUSIONS: The results of this study envision that ABE could directly correct the pathogenic mutation in iPSCs for reversing disease-related phenotypes in neuropathology and exploring novel pathophysiological targets in PD.


Induced Pluripotent Stem Cells , Parkinson Disease , Dopaminergic Neurons , Gene Editing , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation , Parkinson Disease/genetics , Parkinson Disease/therapy , Phenotype , Proteomics
8.
Biochim Biophys Acta Mol Cell Res ; 1868(6): 119011, 2021 05.
Article En | MEDLINE | ID: mdl-33716172

WD repeat and HMG-box DNA binding protein 1 (Wdhd1) is the mouse ortholog of budding yeast Chromosome Transmission Fidelity 4 (CTF4), the protein product of which integrates the MCM2-7 helicase and DNA polymerase α/primase complex to initiate DNA replication. Previous work in fruit flies, Xenopus egg extracts, and human cell lines suggest that Wdhd1 is required for efficient DNA synthesis. However, rigorous in vivo functional studies on Wdhd1 in mammals are unavailable. In the present study, we have successfully generated a Wdhd1 null allele in mice through CRISPR/Cas9-mediated genome editing to investigate the role of Wdhd1 in embryogenesis in vivo. We characterized Wdhd1 expression using quantitative reverse-transcription polymerase chain reaction, and assessed embryonic cell proliferation by histology in both pre- and peri-implantation embryos. While Wdhd1 heterozygous mutant mice were grossly normal and fertile, we observed a reduction in cell proliferation by the gastrulation stage in Wdhd1 homozygous null mutant embryos which severely hampered their growth and viability. These results indicate that Wdhd1 plays a major role in cell proliferation during embryogenesis in mice.


DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development , Animals , CRISPR-Cas Systems , Cell Culture Techniques , Cell Line , Cell Proliferation , Fertility , Gastrulation , Gene Editing , Loss of Function Mutation , Mice
9.
Transplantation ; 105(4): 785-795, 2021 04 01.
Article En | MEDLINE | ID: mdl-32976366

BACKGROUND: Hepatocyte transplantation has been extensively investigated as an alternative to orthotopic liver transplantation. However, its application in routine clinical practice has been restricted because of low initial engraftment and subsequent repopulation. METHODS: Using mice as a model, we have developed a minimally invasive and nontoxic preconditioning strategy based on preadministration of antibodies against hepsin to increase donor hepatocyte retention and engraftment rate. RESULTS: Liver sinusoid diameters decreased significantly with antihepsin pretreatment, and graft cell numbers increased nearly 2-fold in the recipients' liver parenchyma for 20 days after hepatocyte transplantation. Postoperative complications such as hepatic ischemia injury or apparent immune cell accumulation were not observed in recipients. In a hemophilia B mouse model, antihepsin preconditioning enhanced the expression and clotting activity of coagulation factor IX (FIX) to nearly 2-fold that of immunoglobulin G-treated controls and maintained higher plasma FIX clotting activity relative to the prophylactic range for 50 days after hepatocyte transplantation. Antihepsin pretreatment combined with adeno-associated virus-transduced donor hepatocytes expressing human FIX-Triple, a hyperfunctional FIX variant, resulted in plasma FIX levels similar to those associated with mild hemophilia, which protected hemophilia B mice from major bleeding episodes for 50 days after transplantation. Furthermore, antihepsin pretreatment and repeated transplantation resulted in extending the therapeutic period by 30 days relative to the immunoglobulin G control. CONCLUSIONS: Thus, this antihepsin strategy improved the therapeutic effect of hepatocyte transplantation in mice with tremendous safety and minimal invasion. Taken together, we suggest that preconditioning with antihepsin may have clinical applications for liver cell therapy.


Antibodies, Neutralizing/pharmacology , Graft Survival , Hemophilia A/surgery , Hepatocytes/transplantation , Liver Transplantation , Serine Endopeptidases/immunology , Transplantation Conditioning , Animals , Blood Coagulation , Cell Survival , Disease Models, Animal , Factor IX/genetics , Factor IX/metabolism , Hemophilia A/blood , Hemophilia A/genetics , Hemophilia A/immunology , Hepatocytes/immunology , Hepatocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
10.
PLoS One ; 14(7): e0219221, 2019.
Article En | MEDLINE | ID: mdl-31260508

Cul4b-null (Cul4bΔ/Y) mice undergo growth arrest and degeneration during the early embryonic stages and die at E9.5. The pathogenic causes of this lethality remain incompletely characterized. However, it has been hypothesized that the loss of Cul4b function in extraembryonic tissues plays a key role. In this study, we investigated possible causes of death for Cul4b-null embryos, particularly in regard to the role of embryonic Cul4b. First, we show that the loss of embryonic Cul4b affects the growth of the inner cell mass in vitro and delays epiblast development during the gastrulation period at E6.5~E7.5 in vivo, as highlighted by the absence of the epiblastic transcription factor Brachyury from E6.5~E7.5. Additionally, at E7.5, strong and laterally expanded expression of Eomes and Fgf8 signaling was detected. Sectioning of these embryos showed disorganized primitive streak layer cells. Second, we observed that Mash2-expressing cells were present in the extraembryonic tissues of Cul4b-deficient embryos at E6.5 but were absent at E7.5. In addition, the loss of Cul4b resulted in decreased expression of cyclin proteins, which are required for the cell cycle transition from G1 to S. Taken together, these observations suggest that the embryonic expression of Cul4b is important for epiblast growth during E6.5~E7.5, and the loss of Cul4b results in either delayed growth of the epiblast or defective localization of primitive streak layer cells. As a result, the signaling activity mediated by the epiblast for subsequent ectoplacental cone development is affected, with the potential to induce growth retardation and lethality in Cul4bΔ/Y embryos.


Cullin Proteins/physiology , Gastrulation/physiology , Germ Layers/embryology , Primitive Streak/embryology , Animals , Blastocyst Inner Cell Mass/metabolism , Embryo, Mammalian , Female , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Heterozygote , Male , Mice , Mice, Knockout , Models, Animal , T-Box Domain Proteins/metabolism
11.
FASEB J ; 33(4): 5571-5584, 2019 04.
Article En | MEDLINE | ID: mdl-30640520

The clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 technology facilitates somatic genome editing to reveal cooperative genetic interactions at the cellular level without extensive breeding between different mutant animals. Here we propose a transgenic inducible Cas9 effector-CRISPR mutagen ( ICE CRIM) mouse model in which CRISPR/Cas9-mediated somatic mutagenesis events can occur in response to Cre expression. The well-known tumor suppressor gene, Trp53, and 2 important DNA mismatch repair genes, Mlh1 and Msh2, were selected to be our somatic mutagenesis targets. Amplicon-based sequencing was performed to validate the editing efficiency and to identify the mutant allelic series. Crossed with various Cre lines, the Trp53 ICE CRIM alleles were activated to generate targeted cancer gene somatic or germ line mutant variants. We provide experimental evidence to show that an activated ICE CRIM can mutate both targeted alleles within a cell. Simultaneous disruption of multiple genes was also achieved when there were multiple single-guide RNA expression cassettes embedded within an activated ICE CRIM. Our mouse model can be used to generate mutant pools in vivo, which enables a functional screen to be performed in situ. Our results also provide evidence to support a monoclonal origin of hematopoietic neoplasms and to indicate that DNA mismatch repair deficiency accelerates tumorigenesis in Trp53 mutant genetic background.-Fan, H.-H., Yu, I.-S., Lin, Y.-H., Wang, S.-Y., Liaw, Y.-H., Chen, P.-L., Yang, T.-L., Lin, S.-W., Chen, Y.-T. P53 ICE CRIM mouse: a tool to generate mutant allelic series in somatic cells and germ lines for cancer studies.


CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Mutation/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Alleles , Animals , Gene Editing/methods , Gene Targeting/methods , Germ Cells , Mice , Mice, Transgenic/genetics , Mutagenesis/genetics , Oncogenes/genetics , RNA, Guide, Kinetoplastida/genetics
12.
Stem Cell Res ; 33: 51-55, 2018 12.
Article En | MEDLINE | ID: mdl-30316039

Recessive mutations in the GJB2 gene are the most common genetic cause of hearing loss in humans. By using the Sendai-virus delivery system, we generated induced pluripotent stem cells (iPSCs) from the peripheral blood mononuclear cells of a female patient with the p.V37I (c.109G > A) mutation, a GJB2 mutation highly prevalent in the Asian population. The resulting iPSCs had a normal karyotype. The iPSCs also showed pluripotency, as confirmed by immunofluorescence staining, and differentiated into the three germ layers in vivo. This cellular model will provide a useful platform for investigating the pathogenic mechanisms of deafness related to GJB2 mutations.


Induced Pluripotent Stem Cells/metabolism , Adult , Female , Hearing Loss/genetics , Humans , Mutation
13.
Biomaterials ; 154: 134-146, 2018 Feb.
Article En | MEDLINE | ID: mdl-29128842

Head and neck squamous cell carcinoma (HNSCC) is a worldwide leading malignancy with poor prognoses. Aggressive HNSCC is manifested by forming tumor satellites in the invasive front, which is closely associated with epithelial-mesenchymal transition, local invasion, and metastasis. Limited by the pathological analyses of static cancer specimens conducted in most previous investigations, the dynamic processes and the decisive factors of tumor satellite formation in HNSCC cannot be monitored and studied. The establishment of a system to recapitulate the phenomenon in vitro may be instrumental to explicitly address the question. In this study, we explored the feasibility of establishing an in vitro system to induce tumor satellite formation of different HNSCC cells by applying a system composed of three-dimensional collagen scaffolds. The real-time dynamic process of tumor satellite formation could be monitored in detail, and the pivotal factors accounting for HNSCC tumor satellite formation were evaluated. E-cadherin remodeling, vimentin aggregation, invadopodia formation, and extracellular matrix degradation occurred in the HNSCC cells when they were cultured in an environment created with a low calcium concentration and steric collagen scaffolds, which altogether contributed to tumor satellite formation and spreading. The phenomena of HNSCC cell transition observed in the current system were confirmed in vivo with compatible findings in surgical specimens. Through the use of this system, we illustrated a novel method to study tumor satellite formation in HNSCC. It may serve as a platform for further investigation of underlying mechanisms for tumor satellites of cancer.


Collagen/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Head and Neck Neoplasms/pathology , Tissue Scaffolds/chemistry , Cadherins/chemistry , Cadherins/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Humans , Protein Domains , Pseudopodia/drug effects , Pseudopodia/metabolism
14.
Am J Sports Med ; 46(3): 713-727, 2018 03.
Article En | MEDLINE | ID: mdl-29211970

BACKGROUND: Insufficient cell numbers still present a challenge for articular cartilage repair. Converting heterotopic auricular chondrocytes by extracellular matrix may be the solution. HYPOTHESIS: Specific extracellular matrix may convert the phenotype of auricular chondrocytes toward articular cartilage for repair. STUDY DESIGN: Controlled laboratory study. METHODS: For in vitro study, rabbit auricular chondrocytes were cultured in monolayer for several passages until reaching status of dedifferentiation. Later, they were transferred to chondrogenic type II collagen (Col II)-coated plates for further cell conversion. Articular chondrogenic profiles, such as glycosaminoglycan deposition, articular chondrogenic gene, and protein expression, were evaluated after 14-day cultivation. Furthermore, 3-dimensional constructs were fabricated using Col II hydrogel-associated auricular chondrocytes, and their histological and biomechanical properties were analyzed. For in vivo study, focal osteochondral defects were created in the rabbit knee joints, and auricular Col II constructs were implanted for repair. RESULTS: The auricular chondrocytes converted by a 2-step protocol expressed specific profiles of chondrogenic molecules associated with articular chondrocytes. The histological and biomechanical features of converted auricular chondrocytes became similar to those of articular chondrocytes when cultivated with Col II 3-dimensional scaffolds. In an in vivo animal model of osteochondral defects, the treated group (auricular Col II) showed better cartilage repair than did the control groups (sham, auricular cells, and Col II). Histological analyses revealed that cartilage repair was achieved in the treated groups with abundant type II collagen and glycosaminoglycans syntheses rather than elastin expression. CONCLUSION: The study confirmed the feasibility of applying heterotopic chondrocytes for cartilage repair via extracellular matrix-induced cell conversion. CLINICAL RELEVANCE: This study proposes a feasible methodology to convert heterotopic auricular chondrocytes for articular cartilage repair, which may serve as potential alternative sources for cartilage repair.


Cartilage, Articular/surgery , Chondrocytes/transplantation , Knee Joint/surgery , Tissue Engineering , Animals , Cells, Cultured , Chondrogenesis , Collagen Type II/metabolism , Ear Auricle/cytology , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Hydrogels , Rabbits
15.
Mater Sci Eng C Mater Biol Appl ; 82: 69-79, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-29025676

The 3D bioprinting and cell/tissue printing techniques open new possibilities for future applications. To facilitate the 3D bioprinting process, a large amount of living cells are required. Induced pluripotent stem cells (iPSCs) represent a promising cell source for bioprinting. However, the maintenance and expansion of undifferentiated iPSCs are expensive and time consuming. Therefore, in this study a culture method to obtain a sufficient amount of healthy and undifferentiated iPSCs in a short-term period was established. The iPSCs could be passaged for twice on tissue culture polystyrene (TCPS) dish with the conditional medium and could adapt to the feeder-free environment. Feeder-free dishes were further prepared from chitosan, chitosan-hyaluronan, silk fibroin, and polyurethane (PU1 and PU2) two-dimensional substrates. The iPSCs cultured on the chitosan substrates showed a higher proliferation rate without losing the stemness feature. Among the different materials, PU2 could be prepared as a thermoresponsive hydrogel, which was a potential ink for 3D bioprinting. The iPSCs cultured on PU2 substrates well survived when further embedded in PU2 hydrogel. Moreover, PU2 hydrogel printed with iPSCs remained structural integrity. The use of PU2 hydrogel to embed iPSCs reduced the injury to iPSCs by shear stress. These results indicate that iPSCs could be expanded on chitosan or PU2 membranes without the feeder layer and then printed in PU2 hydrogel. The combination of these steps could offer a new possibility for future applications of iPSC-based 3D bioprinting in tissue engineering.


Bioprinting/methods , Induced Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Chitosan/chemistry , Fibroins/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Induced Pluripotent Stem Cells/metabolism , Polystyrenes/chemistry , Printing, Three-Dimensional
16.
Theranostics ; 7(18): 4577-4590, 2017.
Article En | MEDLINE | ID: mdl-29158846

Rationale: Reducing cardiomyocyte death and enhancing their proliferation after myocardial infarction is perhaps the single largest challenge for cardiac tissue regeneration. Survivin (SVV) is the smallest member of the inhibitor of apoptosis (IAP) family but plays two important roles; inhibiting caspase-9 activation in the intrinsic apoptosis pathway, and regulating microtubule dynamics and chromosome segregation during cell division. Genetic depletion of cardiac SVV leads to incomplete cardiomyocyte division and abnormal heart development. However, the function of SVV in adult hearts after myocardial infarction remains unclear. Methods: A homozygous inducible cardiomyocyte-specific SVV knockout transgenic mouse model was established through crossbreeding SVVflox/flox and αMHC-MCM transgenic mice. Adult mice received consecutive intraperitoneal injection of tamoxifen to induce genetic removal of SVV in cardiomyocytes. A SVV overexpressing model was established via local delivery of SVV in wild-type mouse hearts. Results: We found that 30.82% of cardiomyocytes in the peri-infarct region of SVV knockout mice were apoptotic, significantly higher than the 22.18% in control mice. In addition, ejection fraction was 29.00±0.40% in knockout mice compared to 38.04±0.50% in control mice 21 days after myocardial infarction. On the contrary, locally overexpressing SVV in the heart improved cardiac functions. Unexpectedly, we found that altering the subcellular localization of SVV overexpression produced different outcomes. Overexpression of SVV in the cytoplasm decreased cardiomyocyte apoptosis, whereas overexpression of SVV in the nucleus enhanced cardiac regeneration. The ejection fraction of mice overexpressing SVV was 36.58±0.91%, significantly higher than 28.18±1.70% in the GFP control group. Apoptotic cardiomyocytes were only 4.63% in mouse overexpressing cytosolic SVV, compared to 9.31% in the GFP group, and activation of caspase-3 was also reduced. Moreover, mice overexpressing NLS-SVV exhibited a better ejection fraction (36.19±1.02%,) than GFP controls (26.69±0.75%). NLS-SVV enhanced H3P-positive cardiomyocytes in the border zone to 0.28%, compared to only 0.08% in GFP group, through interacting with Aurora B. Conclusions: We demonstrate the importance of SVV subcellular localization in regulating post-MI cardiac repair and regeneration. We hope that this will open new translational approaches through targeted delivery of SVV.


Inhibitor of Apoptosis Proteins/metabolism , Myocytes, Cardiac/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Disease Models, Animal , Inhibitor of Apoptosis Proteins/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Repressor Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Survivin
17.
Data Brief ; 15: 545-561, 2017 Dec.
Article En | MEDLINE | ID: mdl-29071292

Tumor satellite formation is an indicator of cancer invasiveness and correlates with recurrence, metastasis, and poorer prognosis. By analyzing pathological specimens, tumor satellites formed at the tumor-host interface reflect the phenomena of epithelial-mesenchymal transition. It is impossible to reveal the dynamic processes and the decisive factors of tumor satellite formation using clinicopathological approaches alone. Therefore, establishment of an in vitro system to monitor the phenomena is important to explicitly elucidate underlying mechanisms. In this study, we explored the feasibility of creating an in vitro three-dimensional collagen culture system to recapitulate the process of tumor satellite formation. This data presented here are referred to the research article (Chen et al., 2017) [1]. Using this model, the dynamic process of tumor satellite formation could be recapitulated in different types of human cancer cells. Induced by calcium deprivation, the treated cells increased the incidence and migratory distance of tumor satellites. E-cadherin internalization and invadopodia formation were enhanced by calcium deprivation and were associated with cellular dynamic change during tumor satellite formation. The data confirmed the utility of this culture system to recapitulate dynamic cellular alteration and to explore the potential mechanisms of tumor satellite formation.

18.
Nucleic Acids Res ; 45(18): 10492-10503, 2017 Oct 13.
Article En | MEDLINE | ID: mdl-28985359

Telomerase is highly expressed in cancer and embryonic stem cells (ESCs) and implicated in controlling genome integrity, cancer formation and stemness. Previous studies identified that Krüppel-like transcription factor 4 (KLF4) activates telomerase reverse transcriptase (TERT) expression and contributes to the maintenance of self-renewal in ESCs. However, little is known about how KLF4 regulates TERT expression. Here, we discover poly(ADP-ribose) polymerase 1 (PARP1) as a novel KLF4-interacting partner. Knockdown of PARP1 reduces TERT expression and telomerase activity not only in cancer cells, but also in human and mouse ESCs. Recruitment of KLF4 to TERT promoter is reduced in PARP1-suppressed cells. The poly(ADP-ribose) polymerase activity is dispensable, while the oligo(ADP-ribose) polymerase activity is required for the PARP1- and KLF4-mediated TERT activation. Repression of Parp1 in mouse ESCs decreases expression of pluripotent markers and induces differentiation. These results suggest that PARP1 recruits KLF4 to activate telomerase expression and stem cell pluripotency, indicating a positive regulatory role of the PARP1-KLF4 complex in telomerase expression in cancer and stem cells.


Embryonic Stem Cells/metabolism , Kruppel-Like Transcription Factors/physiology , Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1/physiology , Telomerase/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Telomerase/metabolism
19.
Cancer Res ; 77(22): 6083-6096, 2017 11 15.
Article En | MEDLINE | ID: mdl-28939680

Genotoxicity-induced hair loss from chemotherapy and radiotherapy is often encountered in cancer treatment, and there is a lack of effective treatment. In growing hair follicles (HF), quiescent stem cells (SC) are maintained in the bulge region, and hair bulbs at the base contain rapidly dividing, yet genotoxicity-sensitive transit-amplifying cells (TAC) that maintain hair growth. How genotoxicity-induced HF injury is repaired remains unclear. We report here that HFs mobilize ectopic progenitors from distinct TAC compartments for regeneration in adaptation to the severity of dystrophy induced by ionizing radiation (IR). Specifically, after low-dose IR, keratin 5+ basal hair bulb progenitors, rather than bulge SCs, were quickly activated to replenish matrix cells and regenerated all concentric layers of HFs, demonstrating their plasticity. After high-dose IR, when both matrix and hair bulb cells were depleted, the surviving outer root sheath cells rapidly acquired an SC-like state and fueled HF regeneration. Their progeny then homed back to SC niche and supported new cycles of HF growth. We also revealed that IR induced HF dystrophy and hair loss and suppressed WNT signaling in a p53- and dose-dependent manner. Augmenting WNT signaling attenuated the suppressive effect of p53 and enhanced ectopic progenitor proliferation after genotoxic injury, thereby preventing both IR- and cyclophosphamide-induced alopecia. Hence, targeted activation of TAC-derived progenitor cells, rather than quiescent bulge SCs, for anagen HF repair can be a potential approach to prevent hair loss from chemotherapy and radiotherapy. Cancer Res; 77(22); 6083-96. ©2017 AACR.


Alopecia/metabolism , Cell Proliferation , Hair Follicle/metabolism , Stem Cells/metabolism , Alopecia/etiology , Alopecia/physiopathology , Animals , Antineoplastic Agents, Alkylating/adverse effects , Cyclophosphamide/adverse effects , Female , Gene Expression , Hair Follicle/cytology , Keratin-5/genetics , Keratin-5/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Radiation, Ionizing , Regeneration , Stem Cells/cytology , Wnt Signaling Pathway/genetics
20.
Am J Sports Med ; 45(13): 3128-3142, 2017 Nov.
Article En | MEDLINE | ID: mdl-28892654

BACKGROUND: To avoid complicated procedures requiring in vitro chondrocyte expansion for cartilage repair, the development of a culture-free, 1-stage approach combining platelet-rich fibrin (PRF) and autologous cartilage grafts may be the solution. PURPOSE: To develop a feasible 1-step procedure to combine PRF and autologous cartilage grafts for articular chondral defects. STUDY DESIGN: Controlled laboratory study Methods: The chemotactic effects of PRF on chondrocytes harvested from the primary culture of rabbit cartilage were evaluated in vitro and ex vivo. The rabbit chondrocytes were cultured with different concentrations of PRF media and evaluated for their cell proliferation, chondrogenic gene expression, cell viability, and extracellular matrix synthesis abilities. For the in vivo study, the chondral defects were created on established animal models of rabbits. The gross anatomy, histology, and objective scores were evaluated to validate the treatment results. RESULTS: PRF improved the chemotaxis, proliferation, and viability of the cultured chondrocytes. The gene expression of the chondrogenic markers, including type II collagen and aggrecan, revealed that PRF induced the chondrogenic differentiation of cultured chondrocytes. PRF increased the formation and deposition of the cartilaginous matrix produced by cultured chondrocytes. The efficacy of PRF on cell viability was comparable with that of fetal bovine serum. In animal disease models, morphologic, histological, and objectively quantitative evaluation demonstrated that PRF combined with cartilage granules was feasible in facilitating chondral repair. CONCLUSION: PRF enhances the migration, proliferation, viability, and differentiation of chondrocytes, thus showing an appealing capacity for cartilage repair. The data altogether provide evidence to confirm the feasibility of 1-stage, culture-free method of combining PRF and autologous cartilage graft for repairing articular chondral defects. CLINICAL RELEVANCE: The single-stage, culture-free method of combining PRF and autologous cartilage is useful for repairing articular chondral defects. These advantages benefit clinical translation by simplifying and potentiating the efficacy of autologous cartilage transplantation.


Cartilage, Articular/surgery , Chondrocytes/transplantation , Platelet-Rich Fibrin , Aggrecans/genetics , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Chondrogenesis/genetics , Collagen Type II/genetics , Gene Expression , Models, Animal , Rabbits , Transplantation, Autologous
...