Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Article En | MEDLINE | ID: mdl-33318008

Mycobacterium abscessus exhibits Arr (ADP-ribosyltransferase)-dependent rifampin resistance. In apparent contrast, rifabutin (RBT) has demonstrated promising activity in M. abscessus infection models, implying that RBT might not be inactivated by Arr. RBT susceptibility testing of M. abscessusΔarr revealed a strongly decreased MIC. Our findings suggest that the efficacy of RBT might be enhanced by rendering RBT resilient to Arr-dependent modification or by blocking M. abscessus Arr activity.


Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , ADP Ribose Transferases , Humans , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium abscessus/genetics , Rifabutin/pharmacology , Rifampin/pharmacology
2.
Am J Respir Cell Mol Biol ; 61(2): 198-208, 2019 08.
Article En | MEDLINE | ID: mdl-30741559

We established a murine model of multiwall carbon nanotube (MWCNT)-elicited chronic granulomatous disease that bears similarities to human sarcoidosis pathology, including alveolar macrophage deficiency of peroxisome proliferator-activated receptor γ (PPARγ). Because lymphocyte reactivity to mycobacterial antigens has been reported in sarcoidosis, we hypothesized that addition of mycobacterial ESAT-6 (early secreted antigenic target protein 6) to MWCNT might exacerbate pulmonary granulomatous pathology. MWCNTs with or without ESAT-6 peptide 14 were instilled by the oropharyngeal route into macrophage-specific PPARγ-knockout (KO) or wild-type mice. Control animals received PBS or ESAT-6. Lung tissues, BAL cells, and BAL fluid were evaluated 60 days after instillation. PPARγ-KO mice receiving MWCNT + ESAT-6 had increased granulomas and significantly elevated fibrosis (trichrome staining) compared with wild-type mice or PPARγ-KO mice that received only MWCNT. Immunostaining of lung tissues revealed elevated fibronectin and Siglec F expression on CD11c+ infiltrating alveolar macrophages in the presence of MWCNT + ESAT-6 compared with MWCNT alone. Analyses of BAL fluid proteins indicated increased levels of transforming growth factor (TGF)-ß and the TGF-ß pathway mediator IL-13 in PPARγ-KO mice that received MWCNT + ESAT-6 compared with wild-type or PPARγ-KO mice that received MWCNT. Similarly, mRNA levels of matrix metalloproteinase 9, another requisite factor for TGF-ß production, was elevated in PPARγ-KO mice by MWCNT + ESAT-6. Analysis of ESAT-6 in lung tissues by mass spectrometry revealed ESAT-6 retention in lung tissues of PPARγ-KO but not wild-type mice. These data indicate that PPARγ deficiency promotes pulmonary ESAT-6 retention, exacerbates macrophage responses to MWCNT + ESAT-6, and intensifies pulmonary fibrosis. The present findings suggest that the model may facilitate understanding of the effects of environmental factors on sarcoidosis-associated pulmonary fibrosis.


Antigens, Bacterial/pharmacology , Bacterial Proteins/pharmacology , Macrophages, Alveolar/metabolism , PPAR gamma/deficiency , Pulmonary Fibrosis/microbiology , Sarcoidosis, Pulmonary/microbiology , Animals , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid , CD11 Antigens/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis/metabolism , Inflammation , Lung/pathology , Macrophages/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanotubes, Carbon/chemistry , PPAR gamma/genetics , Pulmonary Fibrosis/genetics , Sarcoidosis, Pulmonary/pathology
3.
Methods Mol Biol ; 1199: 141-54, 2014.
Article En | MEDLINE | ID: mdl-25103806

Bone marrow derived multipotent mesenchymal stem cells (MSCs) have the potential to differentiate into bone, cartilage, fat, and muscle cells and are being investigated for their utility in cell-based therapies. Stem cell transplantation therapy represents a novel and innovative approach with the promise to restore function to diseased or damaged heart muscle. Transplanted MSCs are expected to engraft, differentiate, and remodel in response to the surrounding cardiac microenvironment significantly changing the therapeutic approach for heart disease. Quantum Dots (QDs) offer an alternative to organic dyes and fluorescent proteins to label and track cells in vitro and in vivo. Here, we describe in vitro QD labeling of MSCs, MSC integration in a cardiomyocyte co-culture microenvironment, and a fluorescent recovery after photobleaching (FRAP) technique to assess functional cell-cell communication. FRAP techniques establish an optical record of dynamic cellular interactions with high spatial and temporal resolution and can be used to successfully evaluate dynamic changes in cellular coupling in multicellular preparations.


Cellular Microenvironment , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/cytology , Quantum Dots/metabolism , Animals , Cell Separation , Coculture Techniques , Fluorescence Recovery After Photobleaching , Rats , Staining and Labeling
4.
Methods Mol Biol ; 906: 199-210, 2012.
Article En | MEDLINE | ID: mdl-22791434

Mesenchymal stem cells (MSCs) are multipotent cells with the potential to differentiate into bone, -cartilage, adipose, and muscle cells. Adult derived MSCs are being actively investigated because of their potential to be utilized for therapeutic cell-based transplantation. Methods to track MSCs in vivo are -limited, preventing long-term functional studies of transplanted cells. Quantum Dots (QDs) offer an alternative to organic dyes and fluorescent proteins to label and track cells in vitro and in vivo. Nanoparticles are resistant to chemical and metabolic degradation, demonstrating long-term photostability. Here, we describe the technique to label MSCs with QDs and demonstrate intracellular QD distribution in the labeled MSCs with laser scanning confocal fluorescent microscopy.


Cell Tracking/methods , Mesenchymal Stem Cells/cytology , Quantum Dots , Cell Culture Techniques , Microscopy, Confocal , Staining and Labeling
5.
Front Physiol ; 3: 220, 2012.
Article En | MEDLINE | ID: mdl-22737133

Connexin 43 (Cx43), the principal gap junction protein in vascular smooth muscle cells (VSMCs), regulates movement of ions and other signaling molecules through gap junction intercellular communication (GJIC) and plays important roles in maintaining normal vessel function; however, many of the signaling mechanisms controlling Cx43 in VSMCs are not clearly described. The goal of this study was to investigate mechanisms of Cx43 regulation with respect to VSMC proliferation. Treatment of rat primary VSMCs with the cAMP analog 8Br-cAMP, the soluble guanylate cyclase (sGC) stimulator BAY 41-2272 (BAY), or the Cx inducer diallyl disulfide (DADS) significantly reduced proliferation after 72 h compared with vehicle controls. Bromodeoxyuridine uptake revealed reduction (p < 0.05) in DNA synthesis after 6 h and flow cytometry showed reduced (40%) S-phase cell numbers after 16 h in DADS-treated cells compared with vehicle controls. Cx43 expression significantly increased after 270 min treatment with 8Br-cAMP, 8Br-cGMP, BAY or DADS. Inhibition of PKA, PKG or PKC reversed 8Br-cAMP-stimulated increases in Cx43 expression, whereas only PKG or PKC inhibition reversed 8Br-cGMP- and BAY-stimulated increases in total Cx43. Interestingly, stimulation of Cx43 expression by DADS was not dependent on PKA, PKG or PKC. Using fluorescence recovery after photobleaching, only 8Br-cAMP or DADS increased GJIC with 8Br-cAMP mediated by PKC and DADS mediated by PKG. Further, DADS significantly increased phosphorylation at MAPK-sensitive Serine (Ser)255 and Ser279, the cell cycle regulatory kinase-sensitive Ser262 and PKC-sensitive Ser368 after 30 min while 8Br-cAMP significantly increased phosphorylation only at Ser279 compared with controls. This study demonstrates that 8Br-cAMP- and DADS-enhanced GJIC rather than Cx43 expression and/or phosphorylation plays important roles in the regulation of VSMC proliferation and provides new insights into the growth-regulatory capacities of Cx43 in VSM.

6.
PLoS One ; 7(6): e38454, 2012.
Article En | MEDLINE | ID: mdl-22715383

The phenotype of somatic cells has recently been found to be reversible. Direct reprogramming of one cell type into another has been achieved with transduction and over expression of exogenous defined transcription factors emphasizing their role in specifying cell fate. To discover early and novel endogenous transcription factors that may have a role in adult-derived stem cell acquisition of a cardiomyocyte phenotype, mesenchymal stem cells from human and mouse bone marrow and rat liver were co-cultured with neonatal cardiomyocytes as an in vitro cardiogenic microenvironment. Cell-cell communications develop between the two cell types as early as 24 hrs in co-culture and are required for elaboration of a myocardial phenotype in the stem cells 8-16 days later. These intercellular communications are associated with novel Ca(2+) oscillations in the stem cells that are synchronous with the Ca(2+) transients in adjacent cardiomyocytes and are detected in the stem cells as early as 24-48 hrs in co-culture. Early and significant up-regulation of Ca(2+)-dependent effectors, CAMTA1 and RCAN1 ensues before a myocardial program is activated. CAMTA1 loss-of-function minimizes the activation of the cardiac gene program in the stem cells. While the expression of RCAN1 suggests involvement of the well-characterized calcineurin-NFAT pathway as a response to a Ca(2+) signal, the CAMTA1 up-regulated expression as a response to such a signal in the stem cells was unknown. Cell-cell communications between the stem cells and adjacent cardiomyocytes induce Ca(2+) signals that activate a myocardial gene program in the stem cells via a novel and early Ca(2+)-dependent intermediate, up-regulation of CAMTA1.


Adult Stem Cells/metabolism , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Adult , Adult Stem Cells/cytology , Animals , Calcium-Binding Proteins/genetics , Cell Line , Coculture Techniques , Humans , Mice , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Rats , Trans-Activators/genetics , Up-Regulation/physiology
7.
J Nanobiotechnology ; 5: 9, 2007 Nov 07.
Article En | MEDLINE | ID: mdl-17988386

BACKGROUND: Mesenchymal stem cells (MSCs) are multipotent cells with the potential to differentiate into bone, cartilage, fat and muscle cells and are being investigated for their utility in cell-based transplantation therapy. Yet, adequate methods to track transplanted MSCs in vivo are limited, precluding functional studies. Quantum Dots (QDs) offer an alternative to organic dyes and fluorescent proteins to label and track cells in vitro and in vivo. These nanoparticles are resistant to chemical and metabolic degradation, demonstrating long term photostability. Here, we investigate the cytotoxic effects of in vitro QD labeling on MSC proliferation and differentiation and use as a cell label in a cardiomyocyte co-culture. RESULTS: A dose-response to QDs in rat bone marrow MSCs was assessed in Control (no-QDs), Low concentration (LC, 5 nmol/L) and High concentration (HC, 20 nmol/L) groups. QD yield and retention, MSC survival, proinflammatory cytokines, proliferation and DNA damage were evaluated in MSCs, 24 -120 hrs post QD labeling. In addition, functional integration of QD labeled MSCs in an in vitro cardiomyocyte co-culture was assessed. A dose-dependent effect was measured with increased yield in HC vs. LC labeled MSCs (93 +/- 3% vs. 50% +/- 15%, p < 0.05), with a larger number of QD aggregates per cell in HC vs. LC MSCs at each time point (p < 0.05). At 24 hrs >90% of QD labeled cells were viable in all groups, however, at 120 hrs increased apoptosis was measured in HC vs. Control MSCs (7.2% +/- 2.7% vs. 0.5% +/- 0.4%, p < 0.05). MCP-1 and IL-6 levels doubled in HC MSCs when measured 24 hrs after QD labeling. No change in MSC proliferation or DNA damage was observed in QD labeled MSCs at 24, 72 and 120 hrs post labeling. Finally, in a cardiomyocyte co-culture QD labeled MSCs were easy to locate and formed functional cell-to-cell couplings, assessed by dye diffusion. CONCLUSION: Fluorescent QDs label MSC effectively in an in vitro co-culture model. QDs are easy to use, show a high yield and survival rate with minimal cytotoxic effects. Dose-dependent effects suggest limiting MSC QD exposure.

8.
J Surg Res ; 142(2): 263-7, 2007 Oct.
Article En | MEDLINE | ID: mdl-17719065

BACKGROUND: Cellular cardiomyoplasty is emerging as a potentially novel therapeutic option for heart failure and typically involves direct intramyocardial injection of donor cells into a beating heart. Yet, limited rates of cell engraftment remain an obstacle to be overcome before cell therapy is fully recognized. Mechanical and biological mechanisms may account for observed donor cell loss. This study examines acute mechanical loss during intramyocardial injections in beating and arrested hearts. MATERIALS AND METHODS: A porcine cardiopulmonary bypass model was used. Animals underwent either beating (n = 5) or arrested (n = 5) intramyocardial injections into the left ventricle. Fluorescent microspheres were used in lieu of cells because they are biologically inert. Thirty minutes after delivery, animals were euthanized. Microspheres in cardiac and peripheral tissues were quantified using flow cytometry. RESULTS: Approximately 10% of microspheres were retained within the site of injection in both groups. There was no statistical difference between microsphere retention rates in either the beating or the arrested heart group. Microspheres were found in peripheral organs, pericardial fluid, and the delivery device. CONCLUSIONS: The majority of microspheres injected intramyocardially are lost in both beating and arrested hearts. Cardiac standstill does not enhance microsphere retention. Possible mechanisms include leakage from the injection site and washout via the cardiac venous/lymphatic system. Delivery strategy will need to be modified if more cells are to be retained within the target organ.


Cardiomyoplasty , Cell Transplantation , Heart Arrest, Induced , Heart/physiology , Myocardial Contraction , Animals , Cardiopulmonary Bypass , Female , Fluorescence , Heart Ventricles , Injections , Male , Microspheres , Models, Animal , Sus scrofa
9.
Cell Cycle ; 6(13): 1565-9, 2007 Jul 01.
Article En | MEDLINE | ID: mdl-17603299

Heart failure is a major cause of premature death and disability in the United States. Stem cell therapy has attracted great interest for the treatment of myocardial infarction and heart failure. Some tissue-specific adult-derived stem cells demonstrate plasticity in that they are multipotent, react to inductive signals provided by a new micro-environment, and acquire the phenotype of cells endogenous to the new micro-environment. The mechanism through which this phenotype is acquired is unknown. We have demonstrated that a liver-derived clonal stem cell line, WB F344, differentiate into cardiomyocytes in vivo and in vitro. Using a coculture model of neonatal heart cells and WB F344 cells, we have found that cytosolic communication between the two cell types results in calcium-induced transcription of cardiac transcription factors and appears to usher in the cardiac phenotype. Functional gap junctions and IP3 receptors appear to be required for this process. We propose that the observed low frequency of stem cell differentiation into cardiomyocytes when transplanted into the injured heart is due, in part, to their inability to establish functioning intercellular communications with healthy cardiomyocytes and receive instructive signals needed to activate a cardiac gene program.


Adult Stem Cells/cytology , Calcium Signaling/physiology , Cell Differentiation/physiology , Liver/cytology , Myocardium/cytology , Animals , Biological Therapy/methods , Cell Communication , Cells, Cultured , Coculture Techniques , Humans , Myocytes, Cardiac/cytology , Phenotype , Stem Cell Transplantation , Transcription, Genetic
10.
Proc Natl Acad Sci U S A ; 104(10): 3877-82, 2007 Mar 06.
Article En | MEDLINE | ID: mdl-17360446

The mechanisms underlying stem cell acquisition of a cardiac phenotype are unresolved. We studied early events during the acquisition of a cardiac phenotype by a cloned adult liver stem cell line (WB F344) in a cardiac microenvironment. WB F344 cells express a priori the transcription factors GATA4 and SRF, connexin 43 in the cell membrane, and myoinositol 1,4,5-triphosphate receptor in the perinuclear region. Functional cell-cell communication developed between WB F344 cells and adjacent cocultured cardiomyocytes in 24 h. De novo cytoplasmic [Ca(2+)](c) and nuclear [Ca(2+)](nu) oscillations appeared in WB F344 cells, synchronous with [Ca(2+)](i) transients in adjacent cardiomyocytes. The [Ca(2+)] oscillations in the WB F344 cells, but not those in the cardiomyocytes, were eliminated by a gap junction uncoupler and reappeared with its removal. By 24 h, WB F344 cells began expressing the cardiac transcription factors Nkx2.5, Tbx5, and cofactor myocardin; cardiac proteins 24 h later; and a sarcomeric pattern 4-6 days later. Myoinositol 1,4,5-triphosphate receptor inhibition suppressed WB F344 cell [Ca(2+)](nu) oscillations but not [Ca(2+)](c) oscillations, and L-type calcium channel inhibition eliminated [Ca(2+)] oscillations in cardiomyocytes and WB F344 cells. The use of these inhibitors was associated with a decrease in Nkx2.5, Tbx5, and myocardin expression in the WB F344 cells. Our findings suggest that signals from cardiomyocytes diffuse through shared channels, inducing [Ca(2+)] oscillations in the WB F344 cells. We hypothesize that the WB F344 cell [Ca(2+)](nu) oscillations activate the expression of a cardiac specifying gene program, ushering in a cardiac phenotype.


Calcium/metabolism , Liver/cytology , Myocardium/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Nucleus/metabolism , Connexin 43/biosynthesis , Cytoplasm/metabolism , Gap Junctions , Nuclear Proteins/biosynthesis , Phenotype , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Trans-Activators/biosynthesis
11.
Article En | MEDLINE | ID: mdl-17946843

Adult-derived stem cells have recently been found to respond in vivo to inductive signals from the microenvironment and to differentiate into a phenotype that is characteristic of cells in that microenvironment. We examined the differentiation potential of an adult liver stem cell line (WBF344) in a cardiac microenvironment in vitro. WBF344 cells were established from a single cloned non-parenchymal epithelial cell isolated from a normal male adult rat liver. Genetically modified, WBF344 cells that express beta-galactosidase, green fluorescent protein (GFP) or mitochondrial red fluorescent protein (DsRed) were co-cultured with rat neonatal cardiac cells. After 4-14 days, we identified WBF344-derived cardiomyocytes that were elongated, binucleated and expressed the cardiac specific proteins cardiac troponin T, cardiac troponin I and N cadherin. These WBF344-derived cardiomyocytes also exhibited myofibrils, sarcomeres, and a nascent sarcoplasmic reticulum. Furthermore, rhythmically beating WBF344-derived cardiomyocytes displayed "cardiac-like" calcium transients similar to the surrounding neonatal cardiomyocytes. Fluorescent recovery after photobleaching demonstrated that WBF344-derived cardiomyocytes were electrically coupled with adjacent neonatal cardiomyocytes through gap junctions (GJs). Collectively, these results support the conclusion that these adult-derived liver stem cells respond to signals generated in a cardiac microenvironment in vitro acquiring a cardiomyocyte phenotype and function. The identification of micro-environmental signals that appear to cross germ layer and species specificities should prove valuable in understanding the regulation of normal development and stem cell differentiation in vivo.


Adult Stem Cells/cytology , Adult Stem Cells/physiology , Calcium Signaling/physiology , Heart Conduction System/cytology , Heart Conduction System/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Cell Communication/physiology , Cell Differentiation , Cells, Cultured , Rats , Rats, Sprague-Dawley , Stem Cells
12.
J Electrocardiol ; 38(4 Suppl): 55-9, 2005 Oct.
Article En | MEDLINE | ID: mdl-16226075

Sudden cardiac death accounts for more than half of all cardiovascular deaths in the US, and a large proportion of these deaths are attributed to ischemia-induced ventricular fibrillation. As such, the mechanisms underlying the initiation and maintenance of these lethal rhythms are of significant clinical and scientific interest. In large animal hearts, regional ischemia induces two phases of ventricular arrhythmia. The first phase (1A) occurs between 5 and 7 min after arrest of perfusion. This phase is associated with membrane depolarization, a mild intracellular and extracellular acidification and a small membrane depolarization. A second phase (1B) of ventricular arrhythmia occurs between 20 and 30 minutes after arrest of perfusion. This phase occurs at a time when ischemia-induced K+ and pH changes are relatively stable. The arrhythmia is presumed to relate to the process of cell-to-cell electrical uncoupling because a rapid increase of tissue impedance precedes the onset of the arrhythmia. Of note is that tissue resistance is primarily determined by the conductance properties of the gap junctions accounting for cell-to-cell coupling. Impulse propagation in heart is determined by active and passive membrane properties. An important passive cable property that is modulated by ischemia is intercellular resistance and is determined primarily by gap junctional conductance. As such changes in Impulse propagation during myocardial ischemia are determined by contemporaneous changes in active and passive membrane properties. Cellular K loss, intracellular and extracellular acidosis and membrane depolarization are important factors decreasing excitatory currents, while the collapse of the extracellular compartment and cell-to-cell electrical uncoupling increase the resistance to current flow. The time-course of cellular coupling is closely linked to a number of physiological processes including depletion of ATP, and accumulation of intracellular Ca2+. Hence, interventions such as ischemic preconditioning attenuate the effect of subsequent ischemia, delay the onset of cell-to-cell electrical uncoupling and likewise delay the onset of ischemia-induced arrhythmia.


Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Connexins/metabolism , Heart Conduction System/physiopathology , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Animals , Arrhythmias, Cardiac/metabolism , Cell Communication , Electric Conductivity , Gap Junctions , Heart Conduction System/metabolism , Humans , Myocardial Ischemia/metabolism
13.
Am J Pathol ; 165(1): 135-45, 2004 Jul.
Article En | MEDLINE | ID: mdl-15215169

We examined the differentiation potential of an adult liver stem cell line (WB F344) in a cardiac microenvironment, ex vivo. WB F344 cells were established from a single cloned nonparenchymal epithelial cell isolated from a normal male adult rat liver. Genetically modified, WB F344 cells that express beta-galactosidase and green fluorescent protein or only beta-galactosidase were co-cultured with dissociated rat or mouse neonatal cardiac cells. After 4 to 14 days, WB F344-derived cardiomyocytes expressed cardiac-specific proteins and exhibited myofibrils, sarcomeres, and a nascent sarcoplasmic reticulum. Further, rhythmically beating WB F344-derived cardiomyocytes displayed calcium transients. Fluorescent recovery after photobleaching demonstrated that WB F344-derived cardiomyocytes were coupled with adjacent neonatal cardiomyocytes and other WB F344-derived cardiomyocytes. Fluorescence in situ hybridization experiments suggested that fusion between WB F344 cells and neonatal mouse cardiomyocytes did not take place. Collectively, these results support the conclusion that these adult-derived liver stem cells respond to signals generated in a cardiac microenvironment ex vivo acquiring a cardiomyocyte phenotype and function. The identification ex vivo of microenvironmental signals that appear to cross germ layer and species specificities should prove valuable in understanding the molecular basis of adult stem cell differentiation and phenotypic plasticity.


Liver/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cells/cytology , Animals , Animals, Genetically Modified , Animals, Newborn , Calcium/metabolism , Cell Communication , Cell Differentiation , Cell Line , Clone Cells , Coculture Techniques , Epithelial Cells/cytology , Fluorescence Recovery After Photobleaching , Fluorescent Dyes , Green Fluorescent Proteins , Immunohistochemistry , In Situ Hybridization, Fluorescence , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred Strains , Myocytes, Cardiac/ultrastructure , Myosins/metabolism , Phenotype , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retroviridae/genetics , Rhodamines , beta-Galactosidase/metabolism
...