Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 14: 1211026, 2023.
Article in English | MEDLINE | ID: mdl-37608896

ABSTRACT

Introduction: Small-cell-lung-cancer (SCLC) has the worst prognosis of all lung cancers because of a high incidence of relapse after therapy. While lung cancer is the second most common malignancy in the US, only about 10% of cases of lung cancer are SCLC, therefore, it is categorized as a rare and recalcitrant disease. Therapeutic discovery for SCLC has been challenging and the existing pre-clinical models often fail to recapitulate actual tumor pathophysiology. To address this, we developed a bioengineered 3-dimensional (3D) SCLC co-culture organoid model as a phenotypic tool to study SCLC tumor kinetics and SCLC-fibroblast interactions after chemotherapy. Method: We used functionalized alginate microbeads as a scaffold to mimic lung alveolar architecture and co-cultured SCLC cell lines with primary adult lung fibroblasts (ALF). We found that SCLCs in the model proliferated extensively, invaded the microbead scaffold and formed tumors within just 7 days. We compared the bioengineered tumors with patient tumors and found them to recapitulate the pathology and immunophenotyping of the patient tumors. When treated with standard chemotherapy drugs, etoposide and cisplatin, we observed that some of the cells survived the chemotherapy and reformed the tumor in the organoid model. Result and Discussion: Co-culture of the SCLC cells with ALFs revealed that the fibroblasts play a key role in inducing faster and more robust SCLC cell regrowth in the model. This is likely due to a paracrine effect, as conditioned media from the same fibroblasts could also support this accelerated regrowth. This model can be used to study cell-cell interactions and the response to chemotherapy in SCLC and is also scalable and amenable to high throughput phenotypic or targeted drug screening to find new therapeutics for SCLC.

2.
bioRxiv ; 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36711908

ABSTRACT

Small-cell-lung-cancer (SCLC) has the worst prognosis of all lung cancers because of a high incidence of relapse after therapy. We developed a bioengineered 3-dimensional (3D) SCLC co-culture organoid as a phenotypic tool to study SCLC tumor kinetics and SCLC-fibroblast interactions during relapse. We used functionalized alginate microbeads as a scaffold to mimic lung alveolar architecture and co-cultured SCLC cell lines with primary adult lung fibroblasts (ALF). We found that SCLCs in the model proliferated extensively, invaded the microbead scaffold and formed tumors within just 7 days. We compared the bioengineered tumors with patient tumors and found them to recapitulate the pathology and immunophenotyping of the patient tumors better than the PDX model developed from the same SCLC cell line. When treated with standard chemotherapy drugs, etoposide and cisplatin, the organoid recapitulated relapse after chemotherapy. Co-culture of the SCLC cells with ALFs revealed that the fibroblasts play a key role in inducing faster and more robust SCLC cell regrowth in the model. This was a paracrine effect as conditioned medium from the same fibroblasts was responsible for this accelerated cell regrowth. This model is also amenable to high throughput phenotypic or targeted drug screening to find new therapeutics for SCLC.

3.
Circulation ; 145(18): 1412-1426, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35089805

ABSTRACT

BACKGROUND: Human pluripotent stem cell (hPSC)-derived cardiomyocytes (hPSC-CMs) have tremendous promise for application in cardiac regeneration, but their translational potential is limited by an immature phenotype. We hypothesized that large-scale manufacturing of mature hPSC-CMs could be achieved through culture on polydimethylsiloxane (PDMS)-lined roller bottles and that the transplantation of these cells would mediate better structural and functional outcomes than with conventional immature hPSC-CM populations. METHODS: We comprehensively phenotyped hPSC-CMs after in vitro maturation for 20 and 40 days on either PDMS or standard tissue culture plastic substrates. All hPSC-CMs were generated from a transgenic hPSC line that stably expressed a voltage-sensitive fluorescent reporter to facilitate in vitro and in vivo electrophysiological studies, and cardiomyocyte populations were also analyzed in vitro by immunocytochemistry, ultrastructure and fluorescent calcium imaging, and bulk and single-cell transcriptomics. We next compared outcomes after the transplantation of these populations into a guinea pig model of myocardial infarction using end points including histology, optical mapping of graft- and host-derived action potentials, echocardiography, and telemetric electrocardiographic monitoring. RESULTS: We demonstrated the economic generation of >1×108 mature hPSC-CMs per PDMS-lined roller bottle. Compared with their counterparts generated on tissue culture plastic substrates, PDMS-matured hPSC-CMs exhibited increased cardiac gene expression and more mature structural and functional properties in vitro. More important, intracardiac grafts formed with PDMS-matured myocytes showed greatly enhanced structure and alignment, better host-graft electromechanical integration, less proarrhythmic behavior, and greater beneficial effects on contractile function. CONCLUSIONS: We describe practical methods for the scaled generation of mature hPSC-CMs and provide the first evidence that the transplantation of more mature cardiomyocytes yields better outcomes in vivo.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Animals , Cell Differentiation , Cell Line , Guinea Pigs , Humans , Myocytes, Cardiac/metabolism , Plastics/metabolism , Pluripotent Stem Cells/metabolism
4.
Nat Commun ; 12(1): 3155, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039977

ABSTRACT

Compact cardiomyocytes that make up the ventricular wall of the adult heart represent an important therapeutic target population for modeling and treating cardiovascular diseases. Here, we established a differentiation strategy that promotes the specification, proliferation and maturation of compact ventricular cardiomyocytes from human pluripotent stem cells (hPSCs). The cardiomyocytes generated under these conditions display the ability to use fatty acids as an energy source, a high mitochondrial mass, well-defined sarcomere structures and enhanced contraction force. These ventricular cells undergo metabolic changes indicative of those associated with heart failure when challenged in vitro with pathological stimuli and were found to generate grafts consisting of more mature cells than those derived from immature cardiomyocytes following transplantation into infarcted rat hearts. hPSC-derived atrial cardiomyocytes also responded to the maturation cues identified in this study, indicating that the approach is broadly applicable to different subtypes of the heart. Collectively, these findings highlight the power of recapitulating key aspects of embryonic and postnatal development for generating therapeutically relevant cell types from hPSCs.


Subject(s)
Cell Culture Techniques/methods , Heart Failure/therapy , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Disease Models, Animal , Embryo, Mammalian , Embryonic Development/physiology , Heart Atria/cytology , Heart Atria/embryology , Heart Failure/pathology , Heart Ventricles/cytology , Heart Ventricles/embryology , Heart Ventricles/pathology , Humans , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocytes, Cardiac/physiology , Rats
5.
Cell Rep ; 29(11): 3488-3505.e9, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825831

ABSTRACT

Progressive organ fibrosis accounts for one-third of all deaths worldwide, yet preclinical models that mimic the complex, progressive nature of the disease are lacking, and hence, there are no curative therapies. Progressive fibrosis across organs shares common cellular and molecular pathways involving chronic injury, inflammation, and aberrant repair resulting in deposition of extracellular matrix, organ remodeling, and ultimately organ failure. We describe the generation and characterization of an in vitro progressive fibrosis model that uses cell types derived from induced pluripotent stem cells. Our model produces endogenous activated transforming growth factor ß (TGF-ß) and contains activated fibroblastic aggregates that progressively increase in size and stiffness with activation of known fibrotic molecular and cellular changes. We used this model as a phenotypic drug discovery platform for modulators of fibrosis. We validated this platform by identifying a compound that promotes resolution of fibrosis in in vivo and ex vivo models of ocular and lung fibrosis.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Pulmonary Fibrosis/drug therapy , Small Molecule Libraries/pharmacology , Animals , Cell Line , Cells, Cultured , Drug Discovery/methods , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta/metabolism
6.
Curr Protoc Stem Cell Biol ; 46(1): e56, 2018 08.
Article in English | MEDLINE | ID: mdl-29927098

ABSTRACT

This unit describes a protocol for generation of lung organoids. A lung organoid is a 3D cell/hydrogel composite that resembles the morphology and cellular composition of the human distal lung. These tissue-engineered constructs provide an in vitro model of human lung and are best suited for disease modeling applications. The organoid generation methodology is flexible, allowing for easy scalability in the number of organoids produced and in the ability to accommodate a wide range of cell types. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Bioengineering/methods , Imaging, Three-Dimensional , Lung/physiology , Precision Medicine , Alginates/chemistry , Animals , Bioreactors , Collagen Type I/metabolism , Fibroblasts/cytology , Microspheres , Organoids/physiology , Rats , Static Electricity
7.
Stem Cells Transl Med ; 6(2): 622-633, 2017 02.
Article in English | MEDLINE | ID: mdl-28191779

ABSTRACT

Stem cell technologies, especially patient-specific, induced stem cell pluripotency and directed differentiation, hold great promise for changing the landscape of medical therapies. Proper exploitation of these methods may lead to personalized organ transplants, but to regenerate organs, it is necessary to develop methods for assembling differentiated cells into functional, organ-level tissues. The generation of three-dimensional human tissue models also holds potential for medical advances in disease modeling, as full organ functionality may not be necessary to recapitulate disease pathophysiology. This is specifically true of lung diseases where animal models often do not recapitulate human disease. Here, we present a method for the generation of self-assembled human lung tissue and its potential for disease modeling and drug discovery for lung diseases characterized by progressive and irreversible scarring such as idiopathic pulmonary fibrosis (IPF). Tissue formation occurs because of the overlapping processes of cellular adhesion to multiple alveolar sac templates, bioreactor rotation, and cellular contraction. Addition of transforming growth factor-ß1 to single cell-type mesenchymal organoids resulted in morphologic scarring typical of that seen in IPF but not in two-dimensional IPF fibroblast cultures. Furthermore, this lung organoid may be modified to contain multiple lung cell types assembled into the correct anatomical location, thereby allowing cell-cell contact and recapitulating the lung microenvironment. Our bottom-up approach for synthesizing patient-specific lung tissue in a scalable system allows for the development of relevant human lung disease models with the potential for high throughput drug screening to identify targeted therapies. Stem Cells Translational Medicine 2017;6:622-633.


Subject(s)
Cell Culture Techniques , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Induced Pluripotent Stem Cells/pathology , Lung/pathology , Organoids/pathology , Tissue Engineering/methods , Bioreactors , Cell Culture Techniques/instrumentation , Cell Differentiation , Cell Lineage , Cells, Cultured , Fibroblasts/drug effects , Humans , Idiopathic Pulmonary Fibrosis/physiopathology , Induced Pluripotent Stem Cells/drug effects , Lung/physiopathology , Organoids/drug effects , Phenotype , Time Factors , Tissue Engineering/instrumentation , Transforming Growth Factor beta1/pharmacology
8.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L186-L195, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27941077

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a common complication of premature birth. The histopathology of BPD is characterized by an arrest of alveolarization with fibroblast activation. The Wnt/ß-catenin signaling pathway is important in early lung development. When Wnt signaling is active, phosphorylation of ß-catenin by tyrosine kinases at activating sites, specifically at tyrosine 489 (Y489), correlates with nuclear localization of ß-catenin. We examined fetal lung tissue, lung tissue from term newborns, and lung tissue from infants who died with BPD; we found nuclear ß-catenin phosphorylation at Y489 in epithelial and mesenchymal cells in fetal tissue and BPD tissue, but not in the lungs of term infants. Using a 3D human organoid model, we found increased nuclear localization of ß-catenin phosphorylated at Y489 (p-ß-cateninY489) after exposure to alternating hypoxia and hyperoxia compared with organoids cultured in normoxia. Exogenous stimulation of the canonical Wnt pathway in organoids was sufficient to cause nuclear localization of p-ß-cateninY489 in normoxia and mimicked the pattern of α-smooth muscle actin (α-SMA) expression seen with fibroblastic activation from oxidative stress. Treatment of organoids with a tyrosine kinase inhibitor prior to cyclic hypoxia-hyperoxia inhibited nuclear localization of p-ß-cateninY489 and prevented α-SMA expression by fibroblasts. Posttranslational phosphorylation of ß-catenin is a transient feature of normal lung development. Moreover, the persistence of p-ß-cateninY489 is a durable marker of fibroblast activation in BPD and may play an important role in BPD disease pathobiology.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Protein Processing, Post-Translational , beta Catenin/metabolism , Actins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dasatinib/pharmacology , Fibroblasts/drug effects , Humans , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Infant, Newborn , Lung/drug effects , Lung/growth & development , Lung/metabolism , Lung/pathology , Organoids/drug effects , Organoids/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
9.
Am J Physiol Lung Cell Mol Physiol ; 310(10): L889-98, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26968771

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a leading complication of premature birth and occurs primarily in infants delivered during the saccular stage of lung development. Histopathology shows decreased alveolarization and a pattern of fibroblast proliferation and differentiation to the myofibroblast phenotype. Little is known about the molecular pathways and cellular mechanisms that define BPD pathophysiology and progression. We have developed a novel three-dimensional human model of the fibroblast activation associated with BPD, and using this model we have identified the Notch pathway as a key driver of fibroblast activation and proliferation in response to changes in oxygen. Fetal lung fibroblasts were cultured on sodium alginate beads to generate lung organoids. After exposure to alternating hypoxia and hyperoxia, the organoids developed a phenotypic response characterized by increased α-smooth muscle actin (α-SMA) expression and other genes known to be upregulated in BPD and also demonstrated increased expression of downstream effectors of the Notch pathway. Inhibition of Notch with a γ-secretase inhibitor prevented the development of the pattern of cellular proliferation and α-SMA expression in our model. Analysis of human autopsy tissue from the lungs of infants who expired with BPD demonstrated evidence of Notch activation within fibrotic areas of the alveolar septae, suggesting that Notch may be a key driver of BPD pathophysiology.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Signal Transduction , Alginates/chemistry , Bronchopulmonary Dysplasia/metabolism , Cell Culture Techniques , Cell Hypoxia , Cells, Cultured , Culture Media/chemistry , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Receptors, Notch/metabolism
10.
J Biophotonics ; 8(7): 555-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25264131

ABSTRACT

The development of new methods for the early diagnosis of cartilage disease could offer significant improvement in patient care. Raman spectroscopy is an emerging biomedical technology with unique potential to recognize disease tissues, though difficulty in obtaining the samples needed to train a diagnostic and excessive signal noise could slow its development into a clinical tool. In the current report we detail the use of principal component analysis--linear discriminant analysis (PCA-LDA) on spectra from pairs of materials modeling cartilage disease to create multiple spectral scoring metrics, which could limit the reliance on primary training data for identifying disease in low signal-to-noise-ratio (SNR) Raman spectra. Our proof-of-concept experiments show that combinations of these model-metrics has the potential to improve the classification of low-SNR Raman spectra from human normal and osteoarthritic (OA) cartilage over a single metric trained with spectra from the same healthy and OA tissues. Scatter plot showing the PCA-LDA derived human-disease-metric scores versus rat-model-metric scores for 7656 low signal-to-noise spectra from healthy (blue) and osteoarthritic (red) cartilage. Light vertical and horizontal lines represent the optimized single metric classification boundary. Dark diagonal line represents the classification of boundary resulting from the optimized combination of the two metrics.


Subject(s)
Cartilage, Articular , Image Processing, Computer-Assisted/methods , Osteoarthritis/diagnosis , Signal-To-Noise Ratio , Spectrum Analysis, Raman , Animals , Discriminant Analysis , Humans , Male , Multivariate Analysis , Principal Component Analysis , Rats
11.
Nano Lett ; 13(3): 1041-6, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23360394

ABSTRACT

The pathway of interfacial self-assembly of large-scale, highly ordered 2D nanoparticle/polymer monolayer or bilayer arrays from a toluene solution at an air/water interface was investigated using grazing-incidence small-angle scattering at a synchrotron source. Interfacial-assembly of the ordered nanoparticle/polymer array was found to occur through two stages: formation of an incipient randomly close-packed interfacial monolayer followed by compression of the monolayer to form a close-packed lattice driven by solvent evaporation from the polymer. Because the nanoparticles are hydrophobic, they localize exclusively to the polymer-air interface during self-assembly, creating a through thickness asymmetric film as confirmed by X-ray reflectivity. The interfacial self-assembly approach can be extended to form binary NP/polymer arrays. It is anticipated that by understanding the interfacial self-assembly pathway, this simple evaporative procedure could be conducted as a continuous process amenable to large area nanoparticle-based manufacturing needed for emerging energy technologies.

12.
Adv Healthc Mater ; 1(3): 348-53, 2012 May.
Article in English | MEDLINE | ID: mdl-23184753

ABSTRACT

Mesoporous silica nanoparticle-supported lipid bilayers, or "protocells", exhibit a high loading capacity, enhanced colloidal stability, and peptide-directed, cell-specific uptake, making them especially well-suited for targeted delivery of protein toxins to cancer. Protocells loaded with ricin toxin A-chain (RTA) and targeted to hepatocellular carcinoma cause complete cell death at 30 pM of RTA without affecting the viability of control hepatocytes.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Lipid Bilayers/chemistry , Nanocapsules/chemistry , Ricin/administration & dosage , Ricin/chemistry , Silicon Dioxide/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Humans , Materials Testing , Porosity
13.
ACS Nano ; 6(3): 2174-88, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22309035

ABSTRACT

The therapeutic potential of small interfering RNAs (siRNAs) is severely limited by the availability of delivery platforms that protect siRNA from degradation, deliver it to the target cell with high specificity and efficiency, and promote its endosomal escape and cytosolic dispersion. Here we report that mesoporous silica nanoparticle-supported lipid bilayers (or "protocells") exhibit multiple properties that overcome many of the limitations of existing delivery platforms. Protocells have a 10- to 100-fold greater capacity for siRNA than corresponding lipid nanoparticles and are markedly more stable when incubated under physiological conditions. Protocells loaded with a cocktail of siRNAs bind to cells in a manner dependent on the presence of an appropriate targeting peptide and, through an endocytic pathway followed by endosomal disruption, promote delivery of the silencing nucleotides to the cytoplasm. The expression of each of the genes targeted by the siRNAs was shown to be repressed at the protein level, resulting in a potent induction of growth arrest and apoptosis. Incubation of control cells that lack expression of the antigen recognized by the targeting peptide with siRNA-loaded protocells induced neither repression of protein expression nor apoptosis, indicating the precise specificity of cytotoxic activity. In terms of loading capacity, targeting capabilities, and potency of action, protocells provide unique attributes as a delivery platform for therapeutic oligonucleotides.


Subject(s)
Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Nanoparticles/chemistry , Peptides/metabolism , RNA, Small Interfering/metabolism , Silicon Dioxide/chemistry , Transfection/methods , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation , Gene Silencing , Humans , Models, Molecular , Molecular Conformation , Porosity , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...