Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 37(12): 2531-2547, 2022 12.
Article in English | MEDLINE | ID: mdl-36214465

ABSTRACT

Understanding how obesity-induced metabolic stress contributes to synovial joint tissue damage is difficult because of the complex role of metabolism in joint development, maintenance, and repair. Chondrocyte mitochondrial dysfunction is implicated in osteoarthritis (OA) pathology, which motivated us to study the mitochondrial deacetylase enzyme sirtuin 3 (Sirt3). We hypothesized that combining high-fat-diet (HFD)-induced obesity and cartilage Sirt3 loss at a young age would impair chondrocyte mitochondrial function, leading to cellular stress and accelerated OA. Instead, we unexpectedly found that depleting cartilage Sirt3 at 5 weeks of age using Sirt3-flox and Acan-CreERT2 mice protected against the development of cartilage degeneration and synovial hyperplasia following 20 weeks of HFD. This protection was associated with increased cartilage glycolysis proteins and reduced mitochondrial fatty acid metabolism proteins. Seahorse-based assays supported a mitochondrial-to-glycolytic shift in chondrocyte metabolism with Sirt3 deletion. Additional studies with primary murine juvenile chondrocytes under hypoxic and inflammatory conditions showed an increased expression of hypoxia-inducible factor (HIF-1) target genes with Sirt3 deletion. However, Sirt3 deletion impaired chondrogenesis using a murine bone marrow stem/stromal cell pellet model, suggesting a context-dependent role of Sirt3 in cartilage homeostasis. Overall, our data indicate that Sirt3 coordinates HFD-induced changes in mature chondrocyte metabolism that promote OA. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Cell Respiration , Chondrocytes , Chondrogenesis , Diet, High-Fat , Mitochondria , Osteoarthritis , Sirtuin 3 , Animals , Mice , Chondrocytes/metabolism , Diet, High-Fat/adverse effects , Mitochondria/metabolism , Obesity/genetics , Obesity/metabolism , Osteoarthritis/etiology , Osteoarthritis/genetics , Sirtuin 3/genetics , Sirtuin 3/metabolism
2.
Cancers (Basel) ; 14(9)2022 May 05.
Article in English | MEDLINE | ID: mdl-35565426

ABSTRACT

Oxidative phosphorylation is an active metabolic pathway in cancer. Atovaquone is an oral medication that inhibits oxidative phosphorylation and is FDA-approved for the treatment of malaria. We investigated its potential anti-cancer properties by measuring cell proliferation in 2D culture. The clinical formulation of atovaquone, Mepron, was given to mice with ovarian cancers to monitor its effects on tumor and ascites. Patient-derived cancer stem-like cells and spheroids implanted in NSG mice were treated with atovaquone. Atovaquone inhibited the proliferation of cancer cells and ovarian cancer growth in vitro and in vivo. The effect of atovaquone on oxygen radicals was determined using flow and imaging cytometry. The oxygen consumption rate (OCR) in adherent cells was measured using a Seahorse XFe96 Extracellular Flux Analyzer. Oxygen consumption and ATP production were inhibited by atovaquone. Imaging cytometry indicated that the majority of the oxygen radical flux triggered by atovaquone occurred in the mitochondria. Atovaquone decreased the viability of patient-derived cancer stem-like cells and spheroids implanted in NSG mice. NMR metabolomics showed shifts in glycolysis, citric acid cycle, electron transport chain, phosphotransfer, and metabolism following atovaquone treatment. Our studies provide the mechanistic understanding and preclinical data to support the further investigation of atovaquone's potential as a gynecologic cancer therapeutic.

3.
Annu Rev Chem Biomol Eng ; 13: 255-278, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35320695

ABSTRACT

The emergence of human pluripotent stem cell (hPSC) technology over the past two decades has provided a source of normal and diseased human cells for a wide variety of in vitro and in vivo applications. Notably, hPSC-derived cardiomyocytes (hPSC-CMs) are widely used to model human heart development and disease and are in clinical trials for treating heart disease. The success of hPSC-CMs in these applications requires robust, scalable approaches to manufacture large numbers of safe and potent cells. Although significant advances have been made over the past decade in improving the purity and yield of hPSC-CMs and scaling the differentiation process from 2D to 3D, efforts to induce maturation phenotypes during manufacturing have been slow. Process monitoring and closed-loop manufacturing strategies are just being developed. We discuss recent advances in hPSC-CM manufacturing, including differentiation process development and scaling and downstream processes as well as separation and stabilization.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Cell Differentiation , Humans
4.
J Proteome Res ; 20(10): 4646-4654, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34499502

ABSTRACT

Human pluripotent stem-cell-derived cardiomyocytes (hPSC-CMs) show immense promise for patient-specific disease modeling, cardiotoxicity screening, and regenerative therapy development. However, thus far, hPSC-CMs in culture have not recapitulated the structural or functional properties of adult CMs in vivo. To gain global insight into hPSC-CM biology, we established a multiomics method for analyzing the hPSC-CM metabolome and proteome from the same cell culture, creating multidimensional profiles of hPSC-CMs. Specifically, we developed a sequential extraction to capture metabolites and proteins from the same hPSC-CM monolayer cultures and analyzed these extracts using high-resolution mass spectrometry. Using this method, we annotated 205 metabolites/lipids and 4319 proteins from 106 cells with high reproducibility. We further integrated the proteome and metabolome measurements to create network profiles of molecular phenotypes for hPSC-CMs. Out of 310 pathways identified using metabolomics and proteomics, 40 pathways were considered significantly overrepresented (false-discovery-rate-corrected p ≤ 0.05). Highly populated pathways included those involved in protein synthesis (ribosome, spliceosome), ATP generation (oxidative phosphorylation), and cardiac muscle contraction. This multiomics method achieves a deep coverage of metabolites and proteins, creating a multidimensional view of the hPSC-CM phenotype, which provides a strong technological foundation to advance the understanding of hPSC-CM biology. Raw data are available in the MassIVE repository with identifier MSV000088010.


Subject(s)
Myocytes, Cardiac , Proteomics , Cell Differentiation , Humans , Metabolomics , Reproducibility of Results
5.
Adv Healthc Mater ; 10(21): e2100995, 2021 11.
Article in English | MEDLINE | ID: mdl-34459150

ABSTRACT

Spatiotemporally controlled presentation of morphogens and elaborate modulation of signaling pathways elicit pattern formation during development. Though this process is critical for proper organogenesis, unraveling the mechanisms of developmental biology have been restricted by challenges associated with studying human embryos. Human pluripotent stem cells (hPSCs) have been used to model development in vitro, however difficulties in precise spatiotemporal control of the cellular microenvironment have limited the utility of this model in exploring mechanisms of pattern formation. Here, a simple and versatile method is presented to spatially pattern hPSC differentiation in 2-dimensional culture via localized morphogen adsorption on substrates. Morphogens including bone morphogenetic protein 4 (BMP4), activin A, and WNT3a are patterned to induce localized mesendoderm, endoderm, cardiomyocyte (CM), and epicardial cell (EpiC) differentiation from hPSCs and hPSC-derived progenitors. Patterned CM and EpiC co-differentiation allows investigation of intercellular interactions in a spatially controlled manner and demonstrate improved alignment of CMs in proximity to EpiCs. This approach provides a platform for the controlled and systematic study of early pattern formation. Moreover, this study provides a facile approach to generate 2D patterned hPSC-derived tissue structures for modeling disease and drug interactions.


Subject(s)
Pluripotent Stem Cells , Cell Differentiation , Cellular Microenvironment , Humans , Myocytes, Cardiac , Tissue Engineering
6.
Tissue Eng Part C Methods ; 27(5): 322-336, 2021 05.
Article in English | MEDLINE | ID: mdl-33843289

ABSTRACT

Reverse transcription, quantitative polymerase chain reaction (RT-qPCR) is a powerful technique to quantify gene expression by transcript abundance. Expression of target genes is normalized to expression of stable reference genes to account for sample preparation variability. Thus, the identification and validation of stably expressed reference genes is crucial for making accurate, quantitative, statistical conclusions in gene expression studies. Traditional housekeeping genes identified decades ago based on high and relatively stable expression are often used, although many have shown these to not be valid, particularly in highly dynamic systems such as stem cell differentiation. In this study we outline a rational approach to identify stable reference genes valid throughout human pluripotent stem cell (hPSC) differentiation to hPSC-derived cardiomyocytes (hPSC-CMs). Several publicly available transcriptomic data sets were analyzed to identify genes with low variability in expression throughout differentiation. These putative novel reference genes were subsequently validated in RT-qPCR analyses to assess their stability under various perturbations, including maturation during extended culture, lactate purification, and various differentiation efficiencies. Expression in hPSC-CMs was also compared with whole human heart tissue. A core set of three novel reference genes (EDF1, DDB1, and ZNF384) exhibited robust stability across the conditions tested, whereas expression of the traditional housekeeping genes tested (ACTB, B2M, GAPDH, and RPL13A) varied significantly under these conditions. Impact statement This article presents an unbiased method for the selection and validation of novel reference genes for real-time quantitative polymerase chain reaction normalization using data from RNA sequencing datasets. This method identified more robust and stable reference genes for gene expression studies during human pluripotent stem cell differentiation to cardiomyocytes than commonly used reference genes. This study also provides a roadmap for identifying reference genes for assessing gene expression during other dynamic cellular processes, including stem cell differentiation to other cell types.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Cell Differentiation/genetics , Genes, Essential , Humans , Real-Time Polymerase Chain Reaction , Reference Standards
7.
Sci Rep ; 10(1): 21244, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277521

ABSTRACT

Breast cancer metastasis occurs via blood and lymphatic vessels. Breast cancer cells 'educate' lymphatic endothelial cells (LECs) to support tumor vascularization and growth. However, despite known metabolic alterations in breast cancer, it remains unclear how lymphatic endothelial cell metabolism is altered in the tumor microenvironment and its effect in lymphangiogenic signaling in LECs. We analyzed metabolites inside LECs in co-culture with MCF-7, MDA-MB-231, and SK-BR-3 breast cancer cell lines using [Formula: see text] nuclear magnetic resonance (NMR) metabolomics, Seahorse, and the spatial distribution of metabolic co-enzymes using optical redox ratio imaging to describe breast cancer-LEC metabolic crosstalk. LECs co-cultured with breast cancer cells exhibited cell-line dependent altered metabolic profiles, including significant changes in lactate concentration in breast cancer co-culture. Cell metabolic phenotype analysis using Seahorse showed LECs in co-culture exhibited reduced mitochondrial respiration, increased reliance on glycolysis and reduced metabolic flexibility. Optical redox ratio measurements revealed reduced NAD(P)H levels in LECs potentially due to increased NAD(P)H utilization to maintain redox homeostasis. [Formula: see text]-labeled glucose experiments did not reveal lactate shuttling into LECs from breast cancer cells, yet showed other [Formula: see text] signals in LECs suggesting internalized metabolites and metabolic exchange between the two cell types. We also determined that breast cancer co-culture stimulated lymphangiogenic signaling in LECs, yet activation was not stimulated by lactate alone. Increased lymphangiogenic signaling suggests paracrine signaling between LECs and breast cancer cells which could have a pro-metastatic role.


Subject(s)
Breast Neoplasms/metabolism , Endothelial Cells/metabolism , Metabolomics/methods , Cell Line, Tumor , Coculture Techniques , Female , Humans , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , MCF-7 Cells , Oxidation-Reduction , Signal Transduction/genetics , Signal Transduction/physiology
8.
Sci Rep ; 9(1): 6065, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988341

ABSTRACT

Cardiovascular mechanical stresses trigger physiological and pathological cellular reactions including secretion of Transforming Growth Factor ß1 ubiquitously in a latent form (LTGF-ß1). While complex shear stresses can activate LTGF-ß1, the mechanisms underlying LTGF-ß1 activation remain unclear. We hypothesized that different types of shear stress differentially activate LTGF-ß1. We designed a custom-built cone-and-plate device to generate steady shear (SS) forces, which are physiologic, or oscillatory shear (OSS) forces characteristic of pathologic states, by abruptly changing rotation directions. We then measured LTGF-ß1 activation in platelet releasates. We modeled and measured flow profile changes between SS and OSS by computational fluid dynamics (CFD) simulations. We found a spike in shear rate during abrupt changes in rotation direction. OSS activated TGF-ß1 levels significantly more than SS at all shear rates. OSS altered oxidation of free thiols to form more high molecular weight protein complex(es) than SS, a potential mechanism of shear-dependent LTGF-ß1 activation. Increasing viscosity in platelet releasates produced higher shear stress and higher LTGF-ß1 activation. OSS-generated active TGF-ß1 stimulated higher pSmad2 signaling and endothelial to mesenchymal transition (EndoMT)-related genes PAI-1, collagen, and periostin expression in endothelial cells. Overall, our data suggest variable TGF-ß1 activation and signaling occurs with competing blood flow patterns in the vasculature to generate complex shear stress, which activates higher levels of TGF-ß1 to drive vascular remodeling.


Subject(s)
Models, Cardiovascular , Regional Blood Flow/physiology , Stress, Physiological , Transforming Growth Factor beta1/metabolism , Vascular Remodeling/physiology , Blood Platelets/metabolism , Cell Adhesion Molecules/metabolism , Collagen/metabolism , Computer Simulation , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Healthy Volunteers , Hemodynamics/physiology , Human Umbilical Vein Endothelial Cells , Humans , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction/physiology , Smad2 Protein/metabolism
9.
Biotechnol J ; 14(8): e1800725, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30927511

ABSTRACT

Cardiomyocytes (CMs) generated from human pluripotent stem cells (hPSCs) are immature in their structure and function, limiting their potential in disease modeling, drug screening, and cardiac cellular therapies. Prior studies have demonstrated that coculture of hPSC-derived CMs with other cardiac cell types, including endothelial cells (ECs), can accelerate CM maturation. To address whether the CM differentiation stage at which ECs are introduced affects CM maturation, the authors coculture hPSC-derived ECs with hPSC-derived cardiac progenitor cells (CPCs) and CMs and analyze the molecular and functional attributes of maturation. ECs have a more significant effect on acceleration of maturation when cocultured with CPCs than with CMs. EC coculture with CPCs increases CM size, expression of sarcomere, and ion channel genes and proteins, the presence of intracellular membranous extensions, and chronotropic response compared to monoculture. Maturation is accelerated with an increasing EC:CPC ratio. This study demonstrates that EC incorporation at the CPC stage of CM differentiation expedites CM maturation, leading to cells that may be better suited for in vitro and in vivo applications of hPSC-derived CMs.


Subject(s)
Coculture Techniques/methods , Endothelial Cells/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/cytology , Adrenergic beta-Agonists/pharmacology , Cell Differentiation , Cell Size , Endothelial Cells/drug effects , Gene Expression Regulation , Humans , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Troponin C/metabolism , Troponin I/metabolism
10.
Ann Biomed Eng ; 46(1): 37-47, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29022110

ABSTRACT

Ever-increasing demand for bone grafts necessitates the realization of clinical implementation of bone tissue engineered constructs. The predominant hurdle to implementation remains to be securing FDA approval, based on the lack of viable methods for the rigorous monitoring of said constructs. The study presented herein details a method for such monitoring based on the shifting metabolism of mesenchymal stem cells (MSCs) as they differentiate into osteoblasts. To that end, rat MSCs seeded on 85% porous spunbonded poly(L-lactic acid) scaffolds were cultured in flow perfusion bioreactors with baseline or osteoinductive media, and levels of key physio-metabolic markers (oxygen, glucose, osteoprotegerin, and osteocalcin) were monitored throughout culture. Comparison of these non-destructively obtained values and current standard destructive analyses demonstrated key trends useful for the concurrent real-time monitoring of construct cellularity and maturation. Principle among these is the elucidation of the ratio of the rates of oxygen uptake to glucose consumption as a powerful quality marker. This ratio, supported on a physiological basis, has been shown herein to be reliable in the determination of both construct maturation (defined as osteoblastic differentiation and accompanying mineralization) and construct cellularity. Supplementary monitoring of OPG and OCN are shown to provide further validation of such metrics.


Subject(s)
Bone and Bones/metabolism , Mesenchymal Stem Cells/metabolism , Tissue Engineering , Animals , Bioreactors , Cells, Cultured , Culture Media/analysis , Glucose/metabolism , Male , Osteocalcin/metabolism , Osteoprotegerin/metabolism , Oxygen Consumption , Rats, Wistar , Tissue Scaffolds
11.
Biosens Bioelectron ; 90: 443-449, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-27825527

ABSTRACT

As the field of tissue engineering progresses ever-further toward realizing clinical implementation of tissue-engineered constructs for wound regeneration, perhaps the most significant hurdle remains the establishment of non-destructive means for real-time in vitro assessment. In order to address this barrier, the study presented herein established the viability of the development of correlations between metabolic rates (specifically oxygen uptake, glucose consumption, and lactate production) and the cellularity of tissue-engineered cultures comprised of rat mesenchymal stem cells dynamically seeded on 85% porous nonwoven spunbonded poly(l-lactic acid) fiber mesh scaffolds. Said scaffolds were cultured for up to 21 days in a flow perfusion bioreactor system wherein α-MEM (supplemented with 10% fetal bovine serum and 1% antibiotic-antimycotic) was perfused directly through each scaffold at low flow rates (~0.15mL/min). Metabolite measurements were obtained intermittently through the use of a fiber-optic probe (for the case of oxygen) and biochemical assays (for glucose and lactate). Such measurements were subsequently correlated with cellularity data obtained utilizing current-standard destructive means. The resulting correlations, all exhibiting high R2 values, serve as a proof-on-concept for the use of metabolic data for the determination of scaffold cellularity in real-time non-destructively. This study can be easily adapted for use with various cell types, media formulations, and potentially different bioreactor systems. Implementation of more advanced in situ measurement devices could be easily accommodated to allow for true real-time, on-line metabolite monitoring and cellularity estimation.


Subject(s)
Biosensing Techniques , Glucose/isolation & purification , Lactic Acid/isolation & purification , Metabolome , Oxygen/isolation & purification , Animals , Bioreactors , Cattle , Glucose/metabolism , Lactic Acid/metabolism , Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Rats , Regeneration , Tissue Engineering/methods , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL