Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Front Endocrinol (Lausanne) ; 12: 737276, 2021.
Article in English | MEDLINE | ID: mdl-34858326

ABSTRACT

Type 1 diabetes (T1D) is a disease that arises due to complex immunogenetic mechanisms. Key cell-cell interactions involved in the pathogenesis of T1D are activation of autoreactive T cells by dendritic cells (DC), migration of T cells across endothelial cells (EC) lining capillary walls into the islets of Langerhans, interaction of T cells with macrophages in the islets, and killing of ß-cells by autoreactive CD8+ T cells. Overall, pathogenic cell-cell interactions are likely regulated by the individual's collection of genetic T1D-risk variants. To accurately model the role of genetics, it is essential to build systems to interrogate single candidate genes in isolation during the interactions of cells that are essential for disease development. However, obtaining single-donor matched cells relevant to T1D is a challenge. Sourcing these genetic variants from human induced pluripotent stem cells (iPSC) avoids this limitation. Herein, we have differentiated iPSC from one donor into DC, macrophages, EC, and ß-cells. Additionally, we also engineered T cell avatars from the same donor to provide an in vitro platform to study genetic influences on these critical cellular interactions. This proof of concept demonstrates the ability to derive an isogenic system from a single donor to study these relevant cell-cell interactions. Our system constitutes an interdisciplinary approach with a controlled environment that provides a proof-of-concept for future studies to determine the role of disease alleles (e.g. IFIH1, PTPN22, SH2B3, TYK2) in regulating cell-cell interactions and cell-specific contributions to the pathogenesis of T1D.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Type 1/pathology , Induced Pluripotent Stem Cells/pathology , Cell Differentiation/physiology , Humans , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology
2.
Am J Physiol Cell Physiol ; 319(5): C825-C838, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32783656

ABSTRACT

Hypertension (HTN) is a polyfactorial disease that can manifest severe cardiovascular pathologies such as heart failure or stroke. Genome-wide association studies (GWAS) of HTN indicate that single-nucleotide polymorphisms (SNPs) contribute to increased risk for HTN and resistance to some HTN drug regimens (Hiltunen TP et al., J Am Heart Assoc 4: e001521, 2015; Le MT et al., PLoS One 8: e52062, 2013; McDonough CW et al., J Hypertens 31: 698-704, 2013; Vandell AG et al., Hypertension 60: 957-964, 2012). However, cellular mechanistic insights of such SNPs remain largely unknown. Using a bank of induced pluripotent stem cells (iPSCs) derived from patients with HTN and CRISPR/Cas9-mediated gene-editing approach, we investigated the effects of a female HTN risk-associated SNP (rs1154431) of the G protein-coupled estrogen receptor (GPER) (Bassuk SS, Manson JE., Clin Chem 60: 68-77, 2014) in vascular endothelial cells. Although GPER1 deletion reduced endothelial nitric oxide synthase (eNOS) activation in iPSC-derived endothelial cells (iECs), the polymorphism itself did not significantly affect eNOS and NO production in a comparison of isogenic hemizygous iECs expressing either normal (P16) or HTN-associated (L16) GPER. Interestingly, we demonstrate for the first time that GPER plays a role in regulation of adhesion molecule expression and monocyte adhesion to iECs. Moreover, the L16 iECs had higher expression of inflammation genes than P16 iECs, implying that the risk variant may affect carrier individuals through increased inflammatory activity. This study further indicates that iPSCs are a useful platform for exploring mechanistic insights underlying hypertension GWAS endeavors.


Subject(s)
Endothelial Cells/metabolism , Hypertension/genetics , Induced Pluripotent Stem Cells/metabolism , Polymorphism, Single Nucleotide , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Base Sequence , CRISPR-Cas Systems , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Cell Differentiation , Cell Engineering/methods , Endothelial Cells/pathology , Female , Gene Editing/methods , Gene Expression Regulation , Humans , Hypertension/metabolism , Hypertension/physiopathology , Induced Pluripotent Stem Cells/pathology , Models, Biological , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Primary Cell Culture , Receptors, Estrogen/deficiency , Receptors, G-Protein-Coupled/deficiency , Risk Factors , THP-1 Cells , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
3.
Cell Reprogram ; 22(4): 179-188, 2020 08.
Article in English | MEDLINE | ID: mdl-32608992

ABSTRACT

Since it is extremely difficult to establish an animal model for human chromosomal abnormalities, induced pluripotent stem cells (iPSCs) provide a powerful alternative to study underlying mechanisms of these disorders and identify potential therapeutic interventions. In this study we established iPSCs from a young girl with a hemizygous deletion of Xq27.3-q28 who exhibited global developmental delay and intellectual disability from early in infancy. The deletion site on the X chromosome includes Fragile X Mental Retardation 1 (FMR1), the gene responsible for fragile X syndrome, which likely contributes to the patient's neurodevelopmental abnormalities. The FMR1 gene was expressed in approximately half of the iPSC clones we generated while it was absent in the other half due to the random inactivation of normal and abnormal X chromosomes. The normal or absent expression pattern of the FMR1 gene was not altered when the iPSCs were differentiated into neural progenitor cells (NPCs). Moreover, chromosome reactivating reagents such as 5-aza-2-deoxycytidine, trichostatin A, and UNC0638, were tested in an attempt to reactivate the suppressed FMR1 gene in affected iPSC-NPCs. The affected and control isogenic iPSCs developed in this study are ideal models with which to identify downstream consequences caused by the Xq27.3-q28 deletion and also to provide tools for high-throughput screening to identify compounds potentially improving the well-being of this patient population.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation , Cells, Cultured , Child, Preschool , Chromosome Deletion , Chromosomes, Human, X/genetics , Decitabine/pharmacology , Developmental Disabilities/drug therapy , Developmental Disabilities/genetics , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Intellectual Disability/drug therapy , Intellectual Disability/genetics , Quinazolines/pharmacology
4.
Biotechnol Bioeng ; 117(3): 816-831, 2020 03.
Article in English | MEDLINE | ID: mdl-31814110

ABSTRACT

Intracellular delivery of functional proteins is of great interest for basic biological research as well as for clinical applications. Transfection is the most commonly used method, however, it is not applicable to large-scale manipulation and inefficient in important cell types implicated in biomedical applications, such as epithelial, immune and pluripotent stem cells. In this study, we explored a bacterial type III secretion system (Bac-T3SS)-mediated proteofection method to overcome these limitations. An attenuated Pseudomonas aeruginosa vector was constructed, which has features of low toxicity, high T3SS activity, and self-limiting growth. Compared to the method of transfection, the Bac-T3SS showed significantly higher efficiencies of Cre recombinase translocation and target site recombination for hard-to-transfect human cell lines. Furthermore, through the delivery of ß-lactamase in live animals, we demonstrated the feasibility and biosafety of in vivo application of the Bac-T3SS. This study provided an efficient and low-cost proteofection strategy for laboratory use as well as for application in large-scale cell manipulations.


Subject(s)
Integrases/genetics , Recombinant Fusion Proteins/genetics , Transfection/methods , Type III Secretion Systems/genetics , Animals , Bacterial Proteins/genetics , Bioreactors , Cell Line , Genetic Vectors/genetics , Humans , Mice , Pluripotent Stem Cells , Pseudomonas aeruginosa/genetics , Recombinant Fusion Proteins/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism
5.
Sci Adv ; 5(8): eaaw4597, 2019 08.
Article in English | MEDLINE | ID: mdl-31489369

ABSTRACT

The mitochondrial permeability transition pore (MPTP) has resisted molecular identification. The original model of the MPTP that proposed the adenine nucleotide translocator (ANT) as the inner membrane pore-forming component was challenged when mitochondria from Ant1/2 double null mouse liver still had MPTP activity. Because mice express three Ant genes, we reinvestigated whether the ANTs comprise the MPTP. Liver mitochondria from Ant1, Ant2, and Ant4 deficient mice were highly refractory to Ca2+-induced MPTP formation, and when also given cyclosporine A (CsA), the MPTP was completely inhibited. Moreover, liver mitochondria from mice with quadruple deletion of Ant1, Ant2, Ant4, and Ppif (cyclophilin D, target of CsA) lacked Ca2+-induced MPTP formation. Inner-membrane patch clamping in mitochondria from Ant1, Ant2, and Ant4 triple null mouse embryonic fibroblasts showed a loss of MPTP activity. Our findings suggest a model for the MPTP consisting of two distinct molecular components: The ANTs and an unknown species requiring CypD.


Subject(s)
Adenine Nucleotides/genetics , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Transmembrane Permeability-Driven Necrosis/genetics , Peptidyl-Prolyl Isomerase F/genetics , Sequence Deletion/genetics , Animals , Cells, Cultured , Female , Male , Mice , Mice, Knockout , Mitochondrial Permeability Transition Pore
6.
Nature ; 571(7766): 515-520, 2019 07.
Article in English | MEDLINE | ID: mdl-31341297

ABSTRACT

The mitochondrial ADP/ATP carrier (AAC) is a major transport protein of the inner mitochondrial membrane. It exchanges mitochondrial ATP for cytosolic ADP and controls cellular production of ATP. In addition, it has been proposed that AAC mediates mitochondrial uncoupling, but it has proven difficult to demonstrate this function or to elucidate its mechanisms. Here we record AAC currents directly from inner mitochondrial membranes from various mouse tissues and identify two distinct transport modes: ADP/ATP exchange and H+ transport. The AAC-mediated H+ current requires free fatty acids and resembles the H+ leak via the thermogenic uncoupling protein 1 found in brown fat. The ADP/ATP exchange via AAC negatively regulates the H+ leak, but does not completely inhibit it. This suggests that the H+ leak and mitochondrial uncoupling could be dynamically controlled by cellular ATP demand and the rate of ADP/ATP exchange. By mediating two distinct transport modes, ADP/ATP exchange and H+ leak, AAC connects coupled (ATP production) and uncoupled (thermogenesis) energy conversion in mitochondria.


Subject(s)
Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Protons , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Coenzymes/metabolism , Fatty Acids/metabolism , Ion Transport , Male , Mice , Oxygen Consumption
7.
Hum Mol Genet ; 28(14): 2365-2377, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31267131

ABSTRACT

MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.


Subject(s)
Membrane Proteins/genetics , Muscular Diseases/drug therapy , Myoblasts/drug effects , Selective Serotonin Reuptake Inhibitors/therapeutic use , Sertraline/therapeutic use , Animals , Cell Line , Cell Movement , Cell Proliferation , Citalopram/pharmacology , Citalopram/therapeutic use , Drosophila/drug effects , Drosophila/genetics , Drug Evaluation, Preclinical , Humans , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Mutation , Myoblasts/metabolism , Receptor, Notch1/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Signal Transduction , Zebrafish/genetics , Zebrafish/metabolism
8.
Stem Cell Res ; 37: 101434, 2019 05.
Article in English | MEDLINE | ID: mdl-30999275

ABSTRACT

Patient-derived induced pluripotent stem cells (iPSCs) have become a promising resource for exploring genetics of complex diseases, discovering new drugs, and advancing regenerative medicine. Increasingly, laboratories are creating their own banks of iPSCs derived from diverse donors. However, there are not yet standardized guidelines for qualifying these cell lines, i.e., distinguishing between bona fide human iPSCs, somatic cells, and imperfectly reprogrammed cells. Here, we report the establishment of a panel of 30 iPSCs from CD34+ peripheral blood mononuclear cells, of which 10 were further differentiated in vitro into all three germ layers. We characterized these different cell types with commonly used pluripotent and lineage specific markers, and showed that NES, TUBB3, and OTX2 cannot be reliably used as ectoderm differentiation markers. Our work highlights the importance of marker selection in iPSC authentication, and the need for the field to establish definitive standard assays.


Subject(s)
Antigens, Differentiation/metabolism , Biomarkers/metabolism , Cell Differentiation , Ectoderm/metabolism , Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/metabolism , Cells, Cultured , Ectoderm/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/cytology
9.
J Cell Biol ; 218(5): 1491-1502, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30914420

ABSTRACT

Mitochondria contain cardiolipin (CL), an organelle-specific phospholipid that carries four fatty acids with a strong preference for unsaturated chains. Unsaturation is essential for the stability and for the function of mitochondrial CL. Surprisingly, we found tetrapalmitoyl-CL (TPCL), a fully saturated species, in the testes of humans and mice. TPCL was absent from other mouse tissues but was the most abundant CL species in testicular germ cells. Most intriguingly, TPCL was not localized in mitochondria but was in other cellular membranes even though mitochondrial CL was the substrate from which TPCL was synthesized. During spermiogenesis, TPCL became associated with the acrosome, a sperm-specific organelle, along with a subset of authentic mitochondrial proteins, including Ant4, Suox, and Spata18. Our data suggest that mitochondria-derived membranes are assembled into the acrosome, challenging the concept that this organelle is strictly derived from the Golgi apparatus and revealing a novel function of mitochondria.


Subject(s)
Acrosome/metabolism , Cardiolipins/metabolism , Golgi Apparatus/metabolism , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Spermatogenesis , Animals , Humans , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Proteome/analysis
10.
Curr Stem Cell Rep ; 4(4): 299-309, 2018.
Article in English | MEDLINE | ID: mdl-30524939

ABSTRACT

PURPOSE OF REVIEW: Muscular dystrophies (MDs) are a spectrum of muscle disorders, which are caused by a number of gene mutations. The studies of MDs are limited due to lack of appropriate models, except for Duchenne muscular dystrophy (DMD), myotonic dystrophy type 1 (DM1), facioscapulohumeral muscular dystrophy (FSHD), and certain type of limb-girdle muscular dystrophy (LGMD). Human induced pluripotent stem cell (iPSC) technologies are emerging to offer a useful model for mechanistic studies, drug discovery, and cell-based therapy to supplement in vivo animal models. This review will focus on current applications of iPSC as disease models of MDs for studies of pathogenic mechanisms and therapeutic development. RECENT FINDINGS: Many and more human disease-specific iPSCs have been or being established, which carry the natural mutation of MDs with human genomic background. These iPSCs can be differentiated into specific cell types affected in a particular MDs such as skeletal muscle progenitor cells, skeletal muscle fibers, and cardiomyocytes. Human iPSCs are particularly useful for studies of the pathogenicity at the early stage or developmental phase of MDs. High-throughput screening using disease-specific human iPSCs has become a powerful technology in drug discovery. While MD iPSCs have been generated for cell-based replacement therapy, recent advances in genome editing technologies enabled correction of genetic mutations in these cells in culture, raising hope for in vivo genome therapy, which offers a fundamental cure for these daunting inherited MDs. SUMMARY: Human disease-specific iPSC models for MDs are emerging as an additional tool to current disease models for elucidating disease mechanisms and developing therapeutic intervention.

11.
Mol Ther ; 26(11): 2617-2630, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30274788

ABSTRACT

Myotonic dystrophy type 1 (DM1) is caused by a CTG nucleotide repeat expansion within the 3' UTR of the Dystrophia Myotonica protein kinase gene. In this study, we explored therapeutic genome editing using CRISPR/Cas9 via targeted deletion of expanded CTG repeats and targeted insertion of polyadenylation signals in the 3' UTR upstream of the CTG repeats to eliminate toxic RNA CUG repeats. We found paired SpCas9 or SaCas9 guide RNA induced deletion of expanded CTG repeats. However, this approach incurred frequent inversion in both the mutant and normal alleles. In contrast, the insertion of polyadenylation signals in the 3' UTR upstream of the CTG repeats eliminated toxic RNA CUG repeats, which led to phenotype reversal in differentiated neural stem cells, forebrain neurons, cardiomyocytes, and skeletal muscle myofibers. We concluded that targeted insertion of polyadenylation signals in the 3' UTR is a viable approach to develop therapeutic genome editing for DM1.


Subject(s)
Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Neural Stem Cells/physiology , Trinucleotide Repeat Expansion/genetics , 3' Untranslated Regions , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Gene Editing/methods , Genetic Therapy/methods , HEK293 Cells , Humans , Muscle, Skeletal/growth & development , Myocytes, Cardiac/physiology , Myotonic Dystrophy/pathology , Myotonic Dystrophy/therapy , Neurons/physiology , RNA 3' Polyadenylation Signals/genetics , RNA, Guide, Kinetoplastida , Transfection
12.
Stem Cells Transl Med ; 7(7): 551-558, 2018 07.
Article in English | MEDLINE | ID: mdl-29730892

ABSTRACT

All-trans retinoic acid (ATRA) or mesenchymal stem cells (MSCs) have been shown to promote lung tissue regeneration in animal models of emphysema. However, the reparative effects of the combination of the two and the role of p70S6 kinase-1 (p70S6k1) activation in the repair process have not been defined. Twenty-one days after intratracheal instillation of porcine pancreatic elastase (PPE), MSC and/or 10 days of ATRA treatment was initiated. Thirty-two days later, static lung compliance (Cst), mean linear intercepts (MLIs), and alveolar surface area (S) were measured. After PPE, mice demonstrated increased values of Cst and MLI, and decreased S values. Both ATRA and MSC transfer were individually effective in improving these outcomes while the combination of ATRA and MSCs was even more effective. The combination of p70S6k1-/- MSCs transfer followed by ATRA demonstrated only modest effects, and rapamycin treatment of recipients with wild-type (WT) MSCs and ATRA failed to show any effect. However, transfer of p70S6k1 over-expressing-MSCs together with ATRA resulted in further improvements over those seen following WT MSCs together with ATRA. ATRA activated p70S6k1 in MSCs in vitro, which was completely inhibited by rapamycin. Tracking of transferred MSCs following ATRA revealed enhanced accumulation and extended survival of MSCs in recipient lungs following PPE but not vehicle instillation. These data suggest that in MSCs, p70S6k1 activation plays a critical role in ATRA-enhanced lung tissue repair, mediated in part by prolonged survival of transferred MSCs. p70S6k1-activated MSCs may represent a novel therapeutic approach to reverse the lung damage seen in emphysema. Stem Cells Translational Medicine 2018;7:551-558.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Pulmonary Emphysema/therapy , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Female , Lung/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Pancreatic Elastase/toxicity , Phosphorylation , Pulmonary Emphysema/etiology , Regeneration , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Tissue Engineering , Tretinoin/pharmacology , Tretinoin/therapeutic use
13.
Sci Rep ; 8(1): 5039, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29567975

ABSTRACT

Isocitrate dehydrogenase (IDH) 2 participates in the TCA cycle and catalyzes the conversion of isocitrate to α-ketoglutarate and NADP+ to NADPH. In the mitochondria, IDH2 also plays a key role in protecting mitochondrial components from oxidative stress by supplying NADPH to both glutathione reductase (GSR) and thioredoxin reductase 2 (TXNRD2). Here, we report that loss of Idh2 accelerates age-related hearing loss, the most common form of hearing impairment, in male mice. This was accompanied by increased oxidative DNA damage, increased apoptotic cell death, and profound loss of spiral ganglion neurons and hair cells in the cochlea of 24-month-old Idh2-/- mice. In young male mice, loss of Idh2 resulted in decreased NADPH redox state and decreased activity of TXNRD2 in the mitochondria of the inner ear. In HEI-OC1 mouse inner ear cell lines, knockdown of Idh2 resulted in a decline in cell viability and mitochondrial oxygen consumption. This was accompanied by decreased NADPH redox state and decreased activity of TXNRD2 in the mitochondria of the HEI-OC1 cells. Therefore, IDH2 functions as the principal source of NADPH for the mitochondrial thioredoxin antioxidant defense and plays an essential role in protecting hair cells and neurons against oxidative stress in the cochlea of male mice.


Subject(s)
Aging/genetics , Hearing Loss/genetics , Isocitrate Dehydrogenase/genetics , Thioredoxin Reductase 2/genetics , Aging/pathology , Animals , Apoptosis/genetics , Cell Survival/genetics , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Glutathione Reductase/genetics , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , NADP/metabolism , Neurons/metabolism , Neurons/pathology , Oxidative Stress/genetics , Oxygen Consumption/genetics , Spiral Ganglion/metabolism , Spiral Ganglion/pathology
14.
PLoS One ; 13(3): e0194895, 2018.
Article in English | MEDLINE | ID: mdl-29579079

ABSTRACT

Virus-mediated expression of defined transcription factor (TF) genes can effectively induce cellular reprogramming. However, sustained expression of the TFs often hinders pluripotent stem cell (PSC) differentiation into specific cell types, as each TF exerts its effect on PSCs for a defined period of time during differentiation. Here, we applied a bacterial type III secretion system (T3SS)-based protein delivery tool to directly translocate TFs in the form of protein into human PSCs. This transient protein delivery technique showed high delivery efficiency for hPSCs, and it avoids potential genetic alterations caused by the introduction of transgenes. In an established cardiomyocyte de novo differentiation procedure, five transcriptional factors, namely GATA4, MEF2C, TBX5, ESRRG and MESP1 (abbreviated as GMTEM), were translocated at various time points. By detecting the expression of cardiac marker genes (Nkx2.5 and cTnT), we found that GMTEM proteins delivered on mesoderm stage of the cardiomyocytes lineage differentiation significantly enhanced both the human ESC and iPSC differentiation into cardiomyocytes, while earlier or later delivery diminished the enhancing effect. Furthermore, all of the five factors were required to enhance the cardiac differentiation. This work provides a virus-free strategy of transient transcription factors delivery for directing human stem cell fate without jeopardizing genome integrity, thus safe for biomedical applications.


Subject(s)
Bacteria/genetics , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Type III Secretion Systems/genetics , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Plasmids/genetics , Plasmids/metabolism , Pseudomonas aeruginosa/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Biotechnol Adv ; 36(2): 482-493, 2018.
Article in English | MEDLINE | ID: mdl-29409784

ABSTRACT

A protein delivery tool based on bacterial type III secretion system (T3SS) has been broadly applied in biomedical researches. In this review, we summarize various applications of the T3SS-mediate protein delivery which enables translocation of proteins directly into mammalian cells without protein purification. Some of the remarkable advancements include delivery of antigens for therapeutic vaccines, nucleases for genome editing, transcription factors for cellular reprogramming and stem cells differentiation, and signaling molecules for post-translational proteomics studies. With continued improvement of the T3SS-mediated protein delivery tools, even wider application of the technology is anticipated.


Subject(s)
Bacterial Proteins , Drug Delivery Systems , Type III Secretion Systems , Animals , Humans , Models, Biological , Protein Transport
16.
J Leukoc Biol ; 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29437254

ABSTRACT

The IFN-stimulated gene ubiquitin-specific proteinase 18 (USP18) encodes a protein that negatively regulates T1 IFN signaling via stearic inhibition of JAK1 recruitment to the IFN-α receptor 2 subunit (IFNAR2). Here, we demonstrate that USP18 expression is induced by HIV-1 in a T1 IFN-dependent manner. Experimental depletion of USP18 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene editing results in a significant restriction of HIV-1 replication in an induced pluripotent stem cell (iPSC)-derived macrophage model. In the absence of USP18, macrophages have increased responsiveness to stimulation with T1 IFNs with prolonged phosphorylation of STAT1 and STAT2 and increased expression of IFN-stimulated genes that are key for antiviral responses. Interestingly, HIV-1 requires some signaling through the T1 IFN receptor to replicate efficiently because a neutralizing antibody that inhibits T1 IFN activity reduces HIV-1 replication rate in monocyte-derived macrophages. USP18 induction by HIV-1 tunes the IFN response to optimal levels allowing for efficient transcription from the HIV-1 LTR promoter while minimizing the T1 IFN-induced antiviral response that would otherwise restrict viral replication and spread. Finally, iPSC and CRISPR/Cas9 gene targeting offer a powerful tool to study host factors that regulate innate immune responses.

17.
Article in English | MEDLINE | ID: mdl-29093700

ABSTRACT

At least 57 independent loci within the human genome confer varying degrees of risk for the development of type 1 diabetes (T1D). The majority of these variants are thought to contribute to overall genetic risk by modulating host innate and adaptive immune responses, ultimately resulting in a loss of immunological tolerance to ß cell antigens. Early efforts to link specific risk variants with functional alterations in host immune responses have employed animal models or genotype-selected individuals from clinical bioresource banks. While some notable genotype:phenotype associations have been described, there remains an urgent need to accelerate the discovery of causal variants and elucidate the molecular mechanisms by which susceptible alleles alter immune functions. One significant limitation has been the inability to study human T1D risk loci on an isogenic background. The advent of induced pluripotent stem cells (iPSCs) and genome-editing technologies have made it possible to address a number of these outstanding questions. Specifically, the ability to drive multiple cell fates from iPSC under isogenic conditions now facilitates the analysis of causal variants in multiple cellular lineages. Bioinformatic analyses have revealed that T1D risk genes cluster within a limited number of immune signaling pathways, yet the relevant immune cell subsets and cellular activation states in which candidate risk genes impact cellular activities remain largely unknown. In this review, we summarize the functional impact of several candidate risk variants on host immunity in T1D and present an isogenic disease-in-a-dish model system for interrogating risk variants, with the goal of expediting precision therapeutics in T1D.

18.
Lab Invest ; 97(10): 1126-1132, 2017 10.
Article in English | MEDLINE | ID: mdl-28759008

ABSTRACT

Induced pluripotent stem cell (iPSC) technology was originally developed in 2006. Essentially, it converts somatic cells into pluripotent stem cells by transiently expressing a few transcriptional factors. Once generated, these iPSCs can differentiate into all the cell types of our body, theoretically, which has attracted great attention for clinical research including disease pathobiology studies. Could this technology then become an additional research or diagnostic tool widely available to practicing pathologists? Here we summarize progress in iPSC research toward disease pathobiology studies, its future potential, and remaining problems from a pathologist's perspective. A particular focus will be on introducing the effort to recapitulate disease-related morphological changes through three-dimensional culture of stem cells such as organoid differentiation.


Subject(s)
Induced Pluripotent Stem Cells , Pathology, Clinical , Stem Cell Research , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Pathologists
19.
Oncotarget ; 8(17): 28971-28989, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28423644

ABSTRACT

Many breast cancer deaths result from tumors acquiring resistance to available therapies. Thus, new therapeutic agents are needed for targeting drug-resistant breast cancers. Drug-refractory breast cancers include HER2+ tumors that have acquired resistance to HER2-targeted antibodies and kinase inhibitors, and "Triple-Negative" Breast Cancers (TNBCs) that lack the therapeutic targets Estrogen Receptor, Progesterone Receptor, and HER2. A significant fraction of TNBCs overexpress the HER2 family member Epidermal Growth Factor Receptor (EGFR). Thus agents that selectively kill EGFR+ and HER2+ tumors would provide new options for breast cancer therapy. We previously identified a class of compounds we termed Disulfide bond Disrupting Agents (DDAs) that selectively kill EGFR+ and HER2+ breast cancer cells in vitro and blocked the growth of HER2+ breast tumors in an animal model. DDA-dependent cytotoxicity was found to correlate with downregulation of HER1-3 and Akt dephosphorylation. Here we demonstrate that DDAs activate the Unfolded Protein Response (UPR) and that this plays a role in their ability to kill EGFR+ and HER2+ cancer cells. The use of breast cancer cell lines ectopically expressing EGFR or HER2 and pharmacological probes of UPR revealed all three DDA responses: HER1-3 downregulation, Akt dephosphorylation, and UPR activation, contribute to DDA-mediated cytotoxicity. Significantly, EGFR overexpression potentiates each of these responses. Combination studies with DDAs suggest that they may be complementary with EGFR/HER2-specific receptor tyrosine kinase inhibitors and mTORC1 inhibitors to overcome drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Unfolded Protein Response/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Nat Commun ; 8: 14477, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205519

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a common metabolic disorder in obese individuals. Adenine nucleotide translocase (ANT) exchanges ADP/ATP through the mitochondrial inner membrane, and Ant2 is the predominant isoform expressed in the liver. Here we demonstrate that targeted disruption of Ant2 in mouse liver enhances uncoupled respiration without damaging mitochondrial integrity and liver functions. Interestingly, liver specific Ant2 knockout mice are leaner and resistant to hepatic steatosis, obesity and insulin resistance under a lipogenic diet. Protection against fatty liver is partially recapitulated by the systemic administration of low-dose carboxyatractyloside, a specific inhibitor of ANT. Targeted manipulation of hepatic mitochondrial metabolism, particularly through inhibition of ANT, may represent an alternative approach in NAFLD and obesity treatment.


Subject(s)
Adenine Nucleotide Translocator 2/metabolism , Adenosine Triphosphate/metabolism , Fatty Liver/metabolism , Insulin Resistance , Mitochondria, Liver/metabolism , Protective Agents/metabolism , Adenine Nucleotide Translocator 2/genetics , Animals , Atractyloside/analogs & derivatives , Diet, High-Fat , Disease Models, Animal , Fatty Liver/therapy , Female , Glucose Clamp Technique , Hyperinsulinism , Lipid Metabolism , Lipogenesis , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondrial Membranes/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/therapy , Obesity/metabolism , Obesity/therapy , Pyruvic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL