Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
PLoS Genet ; 14(9): e1007679, 2018 09.
Article in English | MEDLINE | ID: mdl-30256787

ABSTRACT

The mechanistic target of rapamycin (mTOR) is an established therapeutic target in renal cell carcinoma (RCC). Mechanisms of secondary resistance to rapalog therapy in RCC have not been studied previously. We identified six patients with metastatic RCC who initially responded to mTOR inhibitor therapy and then progressed, and had pre-treatment and post-treatment tumor samples available for analysis. We performed deep whole exome sequencing on the paired tumor samples and a blood sample. Sequence data was analyzed using Mutect, CapSeg, Absolute, and Phylogic to identify mutations, copy number changes, and their changes over time. We also performed in vitro functional assays on PBRM1 in RCC cell lines. Five patients had clear cell and one had chromophobe RCC. 434 somatic mutations in 416 genes were identified in the 12 tumor samples. 201 (46%) of mutations were clonal in both samples while 129 (30%) were acquired in the post-treatment samples. Tumor heterogeneity or sampling issues are likely to account for some mutations that were acquired in the post-treatment samples. Three samples had mutations in TSC1; one in PTEN; and none in MTOR. PBRM1 was the only gene in which mutations were acquired in more than one post-treatment sample. We examined the effect of PBRM1 loss in multiple RCC cell lines, and could not identify any effect on rapalog sensitivity in in vitro culture assays. We conclude that mTOR pathway gene mutations did not contribute to rapalog resistance development in these six patients with advanced RCC. Furthermore, mechanisms of resistance to rapalogs in RCC remain unclear and our results suggest that PBRM1 loss may contribute to sensitivity through complex transcriptional effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm/genetics , Kidney Neoplasms/drug therapy , Nuclear Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Transcription Factors/genetics , Adult , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins , Disease Progression , Epigenesis, Genetic , Everolimus/pharmacology , Everolimus/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/genetics , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Exome Sequencing
2.
Proc Natl Acad Sci U S A ; 115(27): E6274-E6282, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29891694

ABSTRACT

Chromophobe renal cell carcinoma (ChRCC) accounts for 5% of all sporadic renal cancers and can also occur in genetic syndromes including Birt-Hogg-Dube (BHD) and tuberous sclerosis complex (TSC). ChRCC has a distinct accumulation of abnormal mitochondria, accompanied by characteristic chromosomal imbalances and relatively few "driver" mutations. Metabolomic profiling of ChRCC and oncocytomas (benign renal tumors that share pathological features with ChRCC) revealed both similarities and differences between these tumor types, with principal component analysis (PCA) showing a distinct separation. ChRCC have a striking decrease in intermediates of the glutathione salvage pathway (also known as the gamma-glutamyl cycle) compared with adjacent normal kidney, as well as significant changes in glycolytic and pentose phosphate pathway intermediates. We also found that gamma glutamyl transferase 1 (GGT1), the key enzyme of the gamma-glutamyl cycle, is expressed at ∼100-fold lower levels in ChRCC compared with normal kidney, while no change in GGT1 expression was found in clear cell RCC (ccRCC). Significant differences in specific metabolite abundance were found in ChRCC vs. ccRCC, including the oxidative stress marker ophthalmate. Down-regulation of GGT1 enhanced the sensitivity to oxidative stress and treatment with buthionine sulfoximine (BSO), which was associated with changes in glutathione-pathway metabolites. These data indicate that impairment of the glutathione salvage pathway, associated with enhanced oxidative stress, may have key therapeutic implications for this rare tumor type for which there are currently no specific targeted therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Neoplasm Proteins/metabolism , Oligopeptides/metabolism , gamma-Glutamyltransferase/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Neoplasm Proteins/genetics , Oligopeptides/genetics , Oxidative Stress/genetics , Signal Transduction/genetics , gamma-Glutamyltransferase/genetics
3.
Hum Mol Genet ; 27(9): 1654-1663, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29509898

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disease caused by germline inactivating mutations of TSC1 or TSC2. In TSC-associated tumors of the brain, heart, skin, kidney and lung, inactivation of both alleles of TSC1 or TSC2 leads to hyperactivation of the mTORC1 pathway. The TSC/mTORC1 pathway is a key regulator of cellular processes related to growth, proliferation and autophagy. We and others have previously found that mTORC1 regulates microRNA biogenesis, but the mechanisms are not fully understood. Microprocessor, a multi-protein complex including the nuclease Drosha, processes the primary miR transcript. Using a dual-luciferase reporter, we found that inhibition of mTORC1 or downregulation of Raptor decreased Microprocessor activity, while loss of TSC2 led to a striking increase (∼5-fold) in Microprocessor activity. To determine the global impact of TSC2 on microRNAs we quantitatively analyzed 752 microRNAs in Tsc2-expressing and Tsc2-deficient cells. Out of 259 microRNAs expressed in both cell lines, 137 were significantly upregulated and 24 were significantly downregulated in Tsc2-deficient cells, consistent with the increased Microprocessor activity. Microprocessor activity is known to be regulated in part by GSK3ß. We found that total GSK3ß levels were higher in Tsc2-deficient cells, and the increase in Microprocessor activity associated with Tsc2 loss was reversed by three different GSK3ß inhibitors. Furthermore, mTOR inhibition increased the levels of phospho-GSK3ß (S9), which negatively affects Microprocessor activity. Taken together these data reveal that TSC2 regulates microRNA biogenesis and Microprocessor activity via GSK3ß.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , MicroRNAs/genetics , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Glycogen Synthase Kinase 3 beta/genetics , HeLa Cells , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , RNA, Small Interfering/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
4.
J Biomol NMR ; 74(8-9): 365-379, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32651751

ABSTRACT

The fluorine-19 nucleus was recognized early to harbor exceptional properties for NMR spectroscopy. With 100% natural abundance, a high gyromagnetic ratio (83% sensitivity compared to 1H), a chemical shift that is extremely sensitive to its surroundings and near total absence in biological systems, it was destined to become a favored NMR probe, decorating small and large molecules. However, after early excitement, where uptake of fluorinated aromatic amino acids was explored in a series of animal studies, 19F-NMR lost popularity, especially in large molecular weight systems, due to chemical shift anisotropy (CSA) induced line broadening at high magnetic fields. Recently, two orthogonal approaches, (i) CF3 labeling and (ii) aromatic 19F-13C labeling leveraging the TROSY (Transverse Relaxation Optimized Spectroscopy) effect have been successfully applied to study large biomolecular systems. In this perspective, we will discuss the fascinating early work with fluorinated aromatic amino acids, which reveals the enormous potential of these non-natural amino acids in biological NMR and the potential of 19F-NMR to characterize protein and nucleic acid structure, function and dynamics in the light of recent developments. Finally, we explore how fluorine NMR might be exploited to implement small molecule or fragment screens that resemble physiological conditions and discuss the opportunity to follow the fate of small molecules in living cells.


Subject(s)
Fluorine/chemistry , Magnetic Resonance Spectroscopy , Amino Acids, Aromatic/chemistry , Halogenation , Models, Molecular , Molecular Conformation , Molecular Structure , Molecular Weight , Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry
5.
Genet Med ; 22(8): 1366-1373, 2020 08.
Article in English | MEDLINE | ID: mdl-32341571

ABSTRACT

PURPOSE: Plasma cell-free DNA (cfDNA) variant analysis is commonly used in many cancer subtypes. Cell-free methylated DNA immunoprecipitation sequencing (cfMeDIP-seq) has shown high sensitivity for cancer detection. To date, studies have not compared the sensitivity of both methods in a single cancer subtype. METHODS: cfDNA from 40 metastatic RCC (mRCC) patients was subjected to targeted panel variant analysis. For 34 of 40, cfMeDIP-seq was also performed. A separate cohort of 38 mRCC patients were used in cfMeDIP-seq analysis to train an RCC classifier. RESULTS: cfDNA variant analysis detected 21 candidate variants in 11 of 40 mRCC patients (28%), after exclusion of 2 germline variants and 6 variants reflecting clonal hematopoiesis. Among 23 patients with parallel tumor sequencing, cfDNA analysis alone identified variants in 9 patients (39%), while cfDNA analysis focused on tumor sequencing variant findings improved the sensitivity to 52%. In 34 mRCC patients undergoing cfMeDIP-seq, cfDNA variant analysis identified variants in 7 (21%), while cfMeDIP-seq detected all mRCC cases (100% sensitivity) with 88% specificity in 34 control subjects. In 5 patients with cfDNA variants and serial samples, variant frequency correlated with response to therapy. CONCLUSION: cfMeDIP-seq is significantly more sensitive for mRCC detection than cfDNA variant analysis. However, cfDNA variant analysis may be useful for monitoring response to therapy.


Subject(s)
Carcinoma, Renal Cell , Cell-Free Nucleic Acids , Kidney Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cell-Free Nucleic Acids/genetics , DNA , High-Throughput Nucleotide Sequencing , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Plasma
6.
Genet Med ; 22(9): 1489-1497, 2020 09.
Article in English | MEDLINE | ID: mdl-32461669

ABSTRACT

PURPOSE: To perform comprehensive genotyping of TSC1 and TSC2 in a cohort of 94 infants with tuberous sclerosis complex (TSC) and correlate with clinical manifestations. METHODS: Infants were enrolled at age <4 months, and subject to intensive clinical monitoring including electroencephalography (EEG), brain magnetic resonance imaging (MRI), and neuropsychological assessment. Targeted massively parallel sequencing (MPS), genome sequencing, and multiplex ligation-dependent probe amplification (MLPA) were used for variant detection in TSC1/TSC2. RESULTS: Pathogenic variants in TSC1 or TSC2 were identified in 93 of 94 (99%) subjects, with 23 in TSC1 and 70 in TSC2. Nine (10%) subjects had mosaicism. Eight of 24 clinical features assessed at age 2 years were significantly less frequent in those with TSC1 versus TSC2 variants including cortical tubers, hypomelanotic macules, facial angiofibroma, renal cysts, drug-resistant epilepsy, developmental delay, subependymal giant cell astrocytoma, and median seizure-free survival. Additionally, quantitative brain MRI analysis showed a marked difference in tuber and subependymal nodule/giant cell astrocytoma volume for TSC1 versus TSC2. CONCLUSION: TSC2 pathogenic variants are associated with a more severe clinical phenotype than mosaic TSC2 or TSC1 variants in TSC infants. Early assessment of gene variant status and mosaicism might have benefit for clinical management in infants and young children with TSC.


Subject(s)
Tuberous Sclerosis , Child, Preschool , Humans , Infant , Mosaicism , Mutation , Phenotype , Tuberous Sclerosis/diagnostic imaging , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics
7.
Circ Res ; 114(1): 41-55, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24170267

ABSTRACT

RATIONALE: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. OBJECTIVE: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. METHODS AND RESULTS: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. CONCLUSIONS: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.


Subject(s)
Aging/drug effects , Cardiomyopathies/metabolism , Hypoxia/metabolism , Myoblasts, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Stem Cell Niche , Aging/metabolism , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Cell Cycle , Cell Lineage , Cell Proliferation , Cellular Senescence/drug effects , Hypoxia/pathology , Mice , Mice, Inbred C57BL , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/physiology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Stem Cell Factor/therapeutic use , Telomere Homeostasis
8.
Circulation ; 128(12): 1286-97, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23983250

ABSTRACT

BACKGROUND: Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca(2+) release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein-coupled receptor stimulation, affecting Ca(2+) cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. METHODS AND RESULTS: IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein-coupled receptor activation, Ca(2+) mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca(2+) transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca(2+) elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein-coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca(2+) load by IP3Rs promote Ca(2+) extrusion by forward-mode Na(+)/Ca(2+) exchange, an important mechanism of arrhythmic events. CONCLUSIONS: The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Heart Failure/physiopathology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Myocytes, Cardiac/physiology , Adult , Animals , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Female , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Heart Failure/genetics , Heart Ventricles/cytology , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology , Signal Transduction/physiology
9.
N Engl J Med ; 364(19): 1795-806, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21561345

ABSTRACT

BACKGROUND: Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS: Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS: Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS: Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).


Subject(s)
Lung/cytology , Stem Cells/physiology , Adult , Animals , Clone Cells , Female , Humans , Lung/embryology , Lung/physiology , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells , Proto-Oncogene Proteins c-kit/analysis , Regeneration , Stem Cell Transplantation , Stem Cells/chemistry
11.
Circ Res ; 110(5): 701-15, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22275487

ABSTRACT

RATIONALE: Embryonic and fetal myocardial growth is characterized by a dramatic increase in myocyte number, but whether the expansion of the myocyte compartment is dictated by activation and commitment of resident cardiac stem cells (CSCs), division of immature myocytes or both is currently unknown. OBJECTIVE: In this study, we tested whether prenatal cardiac development is controlled by activation and differentiation of CSCs and whether division of c-kit-positive CSCs in the mouse heart is triggered by spontaneous Ca(2+) oscillations. METHODS AND RESULTS: We report that embryonic-fetal c-kit-positive CSCs are self-renewing, clonogenic and multipotent in vitro and in vivo. The growth and commitment of c-kit-positive CSCs is responsible for the generation of the myocyte progeny of the developing heart. The close correspondence between values computed by mathematical modeling and direct measurements of myocyte number at E9, E14, E19 and 1 day after birth strongly suggests that the organogenesis of the embryonic heart is dependent on a hierarchical model of cell differentiation regulated by resident CSCs. The growth promoting effects of c-kit-positive CSCs are triggered by spontaneous oscillations in intracellular Ca(2+), mediated by IP3 receptor activation, which condition asymmetrical stem cell division and myocyte lineage specification. CONCLUSIONS: Myocyte formation derived from CSC differentiation is the major determinant of cardiac growth during development. Division of c-kit-positive CSCs in the mouse is promoted by spontaneous Ca(2+) spikes, which dictate the pattern of stem cell replication and the generation of a myocyte progeny at all phases of prenatal life and up to one day after birth.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Heart/embryology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , Embryo Culture Techniques , Inositol 1,4,5-Trisphosphate Receptors/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Models, Theoretical , Organogenesis/physiology , Proto-Oncogene Proteins c-kit/genetics
12.
Circulation ; 126(15): 1869-81, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22955965

ABSTRACT

BACKGROUND: Two opposite views of cardiac growth are currently held; one views the heart as a static organ characterized by a large number of cardiomyocytes that are present at birth and live as long as the organism, and the other views the heart a highly plastic organ in which the myocyte compartment is restored several times during the course of life. METHODS AND RESULTS: The average age of cardiomyocytes, vascular endothelial cells (ECs), and fibroblasts and their turnover rates were measured by retrospective (14)C birth dating of cells in 19 normal hearts 2 to 78 years of age and in 17 explanted failing hearts 22 to 70 years of age. We report that the human heart is characterized by a significant turnover of ventricular myocytes, ECs, and fibroblasts, physiologically and pathologically. Myocyte, EC, and fibroblast renewal is very high shortly after birth, decreases during postnatal maturation, remains relatively constant in the adult organ, and increases dramatically with age. From 20 to 78 years of age, the adult human heart entirely replaces its myocyte, EC, and fibroblast compartment ≈8, ≈6, and ≈8 times, respectively. Myocyte, EC, and fibroblast regeneration is further enhanced with chronic heart failure. CONCLUSIONS: The human heart is a highly dynamic organ that retains a remarkable degree of plasticity throughout life and in the presence of chronic heart failure. However, the ability to regenerate cardiomyocytes, vascular ECs, and fibroblasts cannot prevent the manifestations of myocardial aging or oppose the negative effects of ischemic and idiopathic dilated cardiomyopathy.


Subject(s)
Heart Failure/physiopathology , Muscle Development/physiology , Myocytes, Cardiac/physiology , Adolescent , Adult , Aged , Aging , Child , Child, Preschool , Endothelial Cells/physiology , Fibroblasts/physiology , Heart/physiology , Humans , Middle Aged , Myocytes, Cardiac/cytology , Regeneration , Tissue Donors , Young Adult
13.
Circ Res ; 108(10): 1226-37, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454756

ABSTRACT

RATIONALE: The ability of the adult heart to generate new myocytes after injury is not established. OBJECTIVE: Our purpose was to determine whether the adult heart has the capacity to generate new myocytes after injury, and to gain insight into their source. METHODS AND RESULTS: Cardiac injury was induced in the adult feline heart by infusing isoproterenol (ISO) for 10 days via minipumps, and then animals were allowed to recover for 7 or 28 days. Cardiac function was measured with echocardiography, and proliferative cells were identified by nuclear incorporation of 5-bromodeoxyuridine (BrdU; 7-day minipump infusion). BrdU was infused for 7 days before euthanasia at days 10, 17, and 38 or during injury and animals euthanized at day 38. ISO caused reduction in cardiac function with evidence of myocyte loss from necrosis. During this injury phase there was a significant increase in the number of proliferative cells in the atria and ventricle, but there was no increase in BrdU+ myocytes. cKit+ cardiac progenitor cells were BrdU labeled during injury. During the first 7 days of recovery there was a significant reduction in cellular proliferation (BrdU incorporation) but a significant increase in BrdU+ myocytes. There was modest improvement in cardiac structure and function during recovery. At day 38, overall cell proliferation was not different than control, but increased numbers of BrdU+ myocytes were found when BrdU was infused during injury. CONCLUSIONS: These studies suggest that ISO injury activates cardiac progenitor cells that can differentiate into new myocytes during cardiac repair.


Subject(s)
Cardiomyopathies/pathology , Cardiomyopathies/therapy , Myocytes, Cardiac/cytology , Stem Cells/cytology , Age Factors , Animals , Cardiomyopathies/physiopathology , Cats , Cell Differentiation/drug effects , Cell Differentiation/physiology , Isoproterenol/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Recovery of Function/drug effects , Recovery of Function/physiology , Stem Cells/drug effects , Stem Cells/pathology
14.
Circulation ; 123(12): 1287-96, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21403094

ABSTRACT

BACKGROUND: Cardiac stem cells (CSCs) delivered to the infarcted heart generate a large number of small fetal-neonatal cardiomyocytes that fail to acquire the differentiated phenotype. However, the interaction of CSCs with postmitotic myocytes results in the formation of cells with adult characteristics. METHODS AND RESULTS: On the basis of results of in vitro and in vivo assays, we report that the commitment of human CSCs (hCSCs) to the myocyte lineage and the generation of mature working cardiomyocytes are influenced by microRNA-499 (miR-499), which is barely detectable in hCSCs but is highly expressed in postmitotic human cardiomyocytes. miR-499 traverses gap junction channels and translocates to structurally coupled hCSCs favoring their differentiation into functionally competent cells. Expression of miR-499 in hCSCs represses the miR-499 target genes Sox6 and Rod1, enhancing cardiomyogenesis in vitro and after infarction in vivo. Although cardiac repair was detected in all cell-treated infarcted hearts, the aggregate volume of the regenerated myocyte mass and myocyte cell volume were greater in animals injected with hCSCs overexpressing miR-499. Treatment with hCSCs resulted in an improvement in ventricular function, consisting of a better preservation of developed pressure and positive and negative dP/dt after infarction. An additional positive effect on cardiac performance occurred with miR-499, pointing to enhanced myocyte differentiation/hypertrophy as the mechanism by which miR-499 potentiated the restoration of myocardial mass and function in the infarcted heart. CONCLUSIONS: The recognition that miR-499 promotes the differentiation of hCSCs into mechanically integrated cardiomyocytes has important clinical implications for the treatment of human heart failure.


Subject(s)
Adult Stem Cells/cytology , MicroRNAs/physiology , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Stem Cell Transplantation , Adult Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gap Junctions/physiology , Gene Expression/physiology , Humans , Myocardial Infarction/pathology , Myocytes, Cardiac/physiology , Polypyrimidine Tract-Binding Protein , RNA-Binding Proteins/genetics , Rats , Regeneration/physiology , SOXD Transcription Factors/genetics
15.
Circ Res ; 107(11): 1374-86, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-21088285

ABSTRACT

RATIONALE: The turnover of cardiomyocytes in the aging female and male heart is currently unknown, emphasizing the need to define human myocardial biology. OBJECTIVE: The effects of age and gender on the magnitude of myocyte regeneration and the origin of newly formed cardiomyocytes were determined. METHODS AND RESULTS: The interaction of myocyte replacement, cellular senescence, growth inhibition, and apoptosis was measured in normal female (n=32) and male (n=42) human hearts collected from patients 19 to 104 years of age who died from causes other than cardiovascular diseases. A progressive loss of telomeric DNA in human cardiac stem cells (hCSCs) occurs with aging and the newly formed cardiomyocytes inherit short telomeres and rapidly reach the senescent phenotype. Our data provide novel information on the superior ability of the female heart to sustain the multiple variables associated with the development of the senescent myopathy. At all ages, the female heart is equipped with a larger pool of functionally competent hCSCs and younger myocytes than the male myocardium. The replicative potential is higher and telomeres are longer in female hCSCs than in male hCSCs. In the female heart, myocyte turnover occurs at a rate of 10%, 14%, and 40% per year at 20, 60, and 100 years of age, respectively. Corresponding values in the male heart are 7%, 12%, and 32% per year, documenting that cardiomyogenesis involves a large and progressively increasing number of parenchymal cells with aging. From 20 to 100 years of age, the myocyte compartment is replaced 15 times in women and 11 times in men. CONCLUSIONS: The human heart is a highly dynamic organ regulated by a pool of resident hCSCs that modulate cardiac homeostasis and condition organ aging.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Differentiation/physiology , Cellular Senescence/physiology , Heart/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Cell Death/physiology , Cells, Cultured , Female , Heart/anatomy & histology , Humans , Male , Middle Aged , Sex Characteristics , Young Adult
16.
Circ Res ; 107(2): 305-15, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20522802

ABSTRACT

RATIONALE: The ability of the human heart to regenerate large quantities of myocytes remains controversial, and the extent of myocyte renewal claimed by different laboratories varies from none to nearly 20% per year. OBJECTIVE: To address this issue, we examined the percentage of myocytes, endothelial cells, and fibroblasts labeled by iododeoxyuridine in postmortem samples obtained from cancer patients who received the thymidine analog for therapeutic purposes. Additionally, the potential contribution of DNA repair, polyploidy, and cell fusion to the measurement of myocyte regeneration was determined. METHODS AND RESULTS: The fraction of myocytes labeled by iododeoxyuridine ranged from 2.5% to 46%, and similar values were found in fibroblasts and endothelial cells. An average 22%, 20%, and 13% new myocytes, fibroblasts, and endothelial cells were generated per year, suggesting that the lifespan of these cells was approximately 4.5, 5, and 8 years, respectively. The newly formed cardiac cells showed a fully differentiated adult phenotype and did not express the senescence-associated protein p16(INK4a). Moreover, measurements by confocal microscopy and flow cytometry documented that the human heart is composed predominantly of myocytes with 2n diploid DNA content and that tetraploid and octaploid nuclei constitute only a small fraction of the parenchymal cell pool. Importantly, DNA repair, ploidy formation, and cell fusion were not implicated in the assessment of myocyte regeneration. CONCLUSIONS: Our findings indicate that the human heart has a significant growth reserve and replaces its myocyte and nonmyocyte compartment several times during the course of life.


Subject(s)
Cell Proliferation , Endothelial Cells/pathology , Fibroblasts/pathology , Muscle Development , Myocardium/pathology , Myocytes, Cardiac/pathology , Neoplasms/pathology , Adult , Age Factors , Aged , Animals , Autopsy , Cell Death , Cell Fusion , Cell Nucleus/pathology , Cell Proliferation/drug effects , DNA Repair , Endothelial Cells/drug effects , Female , Fibroblasts/drug effects , Flow Cytometry , Humans , Idoxuridine/therapeutic use , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Muscle Development/drug effects , Myocytes, Cardiac/drug effects , Neoplasms/drug therapy , Phenotype , Polyploidy , Radiation-Sensitizing Agents/therapeutic use , Rats , Rats, Inbred F344 , Regeneration , Time Factors , Young Adult
17.
Circ Res ; 107(3): 429-41, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20558824

ABSTRACT

RATIONALE: Physiological hypertrophy in the developing heart has been considered the product of an increase in volume of preexisting fetal cardiomyocytes in the absence of myocyte formation. OBJECTIVE: In this study, we tested whether the mouse heart at birth has a pool of cardiac stem cells (CSCs) that differentiate into myocytes contributing to the postnatal expansion of the parenchymal cell compartment. METHODS AND RESULTS: We have found that the newborn heart contains a population of c-kit-positive CSCs that are lineage negative, self-renewing, and multipotent. CSCs express the Notch1 receptor and show the nuclear localization of its active fragment, N1ICD. In 60% of cases, N1ICD was coupled with the presence of Nkx2.5, indicating that the commitment of CSCs to the myocyte lineage is regulated by Notch1. Importantly, overexpression of N1ICD in neonatal CSCs significantly expanded the proportion of transit-amplifying myocytes. To establish whether these in vitro findings had a functional counterpart in vivo, the Notch pathway was blocked in newborn mice by administration of a gamma-secretase inhibitor. This intervention resulted in the development of a dilated myopathy and high mortality rates. Ventricular decompensation was characterized by a 62% reduction in amplifying myocytes, which resulted in a 54% decrease in myocyte number. After cessation of Notch blockade and recovery of myocyte regeneration, cardiac anatomy and function were largely restored. CONCLUSIONS: Notch1 signaling is a critical determinant of CSC growth and differentiation; when this cascade of events is altered, cardiomyogenesis is impaired, physiological cardiac hypertrophy is prevented, and a life-threatening myopathy supervenes.


Subject(s)
Cardiomyopathy, Dilated/etiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Receptor, Notch1/antagonists & inhibitors , Actinin/metabolism , Actins/metabolism , Animals , Animals, Newborn , Capillaries/cytology , Capillaries/physiology , Cardiomyopathy, Dilated/physiopathology , Cell Differentiation , Cell Division , Heart/growth & development , Humans , Infant, Newborn , Mice , Receptor, Notch1/physiology , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/physiology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 106(40): 17169-74, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19805158

ABSTRACT

An analysis of the clonality of cardiac progenitor cells (CPCs) and myocyte turnover in vivo requires genetic tagging of the undifferentiated cells so that the clonal marker of individual mother cells is traced in the specialized progeny. CPC niches in the atria and apex of the mouse heart were infected with a lentivirus carrying EGFP, and the destiny of the tagged cells was determined 1-5 months later. A common integration site was identified in isolated CPCs, cardiomyocytes, endothelial cells (ECs), and fibroblasts, documenting CPC self-renewal and multipotentiality and the clonal origin of the differentiated cell populations. Subsequently, the degree of EGFP-lentiviral infection of CPCs was evaluated 2-4 days after injection, and the number of myocytes expressing the reporter gene was measured 6 months later. A BrdU pulse-chasing protocol was also introduced as an additional assay for the analysis of myocyte turnover. Over a period of 6 months, each EGFP-positive CPC divided approximately eight times generating 230 cardiomyocytes; this value was consistent with the number of newly formed cells labeled by BrdU. To determine whether, human CPCs (hCPCs) are self-renewing and multipotent, these cells were transduced with the EGFP-lentivirus and injected after acute myocardial infarction in immunosuppressed rats. hCPCs, myocytes, ECs, and fibroblasts collected from the regenerated myocardium showed common viral integration sites in the human genome. Thus, our results indicate that the adult heart contains a pool of resident stem cells that regulate cardiac homeostasis and repair.


Subject(s)
Cell Differentiation , Cell Proliferation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Lineage , Clone Cells/cytology , Clone Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Lentivirus/genetics , Mice , Molecular Sequence Data , Myocardium/cytology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Rats , Rats, Inbred F344 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
19.
J Clin Invest ; 132(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35358092

ABSTRACT

BackgroundTuberous sclerosis complex (TSC) is a neurogenetic syndrome due to loss-of-function mutations in TSC2 or TSC1, characterized by tumors at multiple body sites, including facial angiofibroma (FAF). Here, an ultrasensitive assessment of the extent and range of UV-induced mutations in TSC facial skin was performed.MethodsA multiplex high-sensitivity PCR assay (MHPA) was developed, enabling mutation detection at extremely low (<0.1%) variant allele frequencies (VAFs).ResultsMHPA assays were developed for both TSC2 and TP53, and applied to 81 samples, including 66 skin biopsies. UV-induced second-hit mutation causing inactivation of TSC2 was pervasive in TSC facial skin with an average of 4.8 mutations per 2-mm biopsy at median VAF 0.08%, generating more than 150,000 incipient facial tumors (subclinical "micro-FAFs") in the average TSC subject. The MHPA analysis also led to the identification of a refined UV-related indel signature and a recurrent complex mutation pattern, consisting of both a single-nucleotide or dinucleotide variant and a 1- to 9-nucleotide deletion, in cis.ConclusionTSC facial skin can be viewed as harboring a patchwork of clonal fibroblast proliferations (micro-FAFs) with indolent growth, a small proportion of which develop into clinically observable FAF. Our observations also expand the spectrum of UV-related mutation signatures.FundingThis work was supported by the TSC Alliance; the Engles Family Fund for Research in TSC and LAM; and the NIH, National Heart, Lung, and Blood Institute (U01HL131022-04 and Intramural Research Program).


Subject(s)
Facial Neoplasms , Tuberous Sclerosis , Facial Neoplasms/genetics , Humans , Mutation , Nucleotides , Tuberous Sclerosis/diagnosis , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL