Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
Add more filters

Publication year range
1.
Genes Dev ; 32(13-14): 903-908, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29950491

ABSTRACT

Loss of function of the DIS3L2 exoribonuclease is associated with Wilms tumor and the Perlman congenital overgrowth syndrome. LIN28, a Wilms tumor oncoprotein, triggers the DIS3L2-mediated degradation of the precursor of let-7, a microRNA that inhibits Wilms tumor development. These observations have led to speculation that DIS3L2-mediated tumor suppression is attributable to let-7 regulation. Here we examine new DIS3L2-deficient cell lines and mouse models, demonstrating that DIS3L2 loss has no effect on mature let-7 levels. Rather, analysis of Dis3l2-null nephron progenitor cells, a potential cell of origin of Wilms tumors, reveals up-regulation of Igf2, a growth-promoting gene strongly associated with Wilms tumorigenesis. These findings nominate a new potential mechanism underlying the pathology associated with DIS3L2 deficiency.


Subject(s)
Exoribonucleases/genetics , Fetal Macrosomia/genetics , Insulin-Like Growth Factor II/genetics , Up-Regulation , Wilms Tumor/genetics , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , Mutation , Nephrons/cytology , Nephrons/physiopathology , Stem Cells
2.
Cell ; 143(1): 35-45, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20887891

ABSTRACT

Maintenance of skeletal muscle structure and function requires innervation by motor neurons, such that denervation causes muscle atrophy. We show that myogenin, an essential regulator of muscle development, controls neurogenic atrophy. Myogenin is upregulated in skeletal muscle following denervation and regulates expression of the E3 ubiquitin ligases MuRF1 and atrogin-1, which promote muscle proteolysis and atrophy. Deletion of myogenin from adult mice diminishes expression of MuRF1 and atrogin-1 in denervated muscle and confers resistance to atrophy. Mice lacking histone deacetylases (HDACs) 4 and 5 in skeletal muscle fail to upregulate myogenin and also preserve muscle mass following denervation. Conversely, forced expression of myogenin in skeletal muscle of HDAC mutant mice restores muscle atrophy following denervation. Thus, myogenin plays a dual role as both a regulator of muscle development and an inducer of neurogenic atrophy. These findings reveal a specific pathway for muscle wasting and potential therapeutic targets for this disorder.


Subject(s)
Histone Deacetylases/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Myogenin/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Atrophy , Mice , Mice, Knockout , Tripartite Motif Proteins
3.
Genes Dev ; 28(23): 2585-90, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25395662

ABSTRACT

Down-regulation of miR-26 family members has been implicated in the pathogenesis of multiple malignancies. In some settings, including glioma, however, miR-26-mediated repression of PTEN promotes tumorigenesis. To investigate the contexts in which the tumor suppressor versus oncogenic activity of miR-26 predominates in vivo, we generated miR-26a transgenic mice. Despite measureable repression of Pten, elevated miR-26a levels were not associated with malignancy in transgenic animals. We documented reduced miR-26 expression in human colorectal cancer and, accordingly, showed that miR-26a expression potently suppressed intestinal adenoma formation in Apc(min/+) mice, a model known to be sensitive to Pten dosage. These studies reveal a tumor suppressor role for miR-26 in intestinal cancer that overrides putative oncogenic activity, highlighting the therapeutic potential of miR-26 delivery to this tumor type.


Subject(s)
Adenoma/physiopathology , Carcinogenesis/genetics , Intestinal Neoplasms/physiopathology , MicroRNAs/metabolism , Adenoma/genetics , Animals , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/physiopathology , Intestinal Neoplasms/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Tumor Cells, Cultured
4.
Surg Innov ; 29(2): 183-194, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34414835

ABSTRACT

Background and Purpose. This study established a suitable animal model of ovariohysterectomy; characterized the course and pattern of vaginal healing after ovariohysterectomy; and compared healing obtained after closure of the vaginal cuff with a novel cuff-closure device (Zip-stitch® clips) and VICRYL® sutures. Research Design and Study Sample. This prospective, randomized, controlled, blinded animal study was conducted in 27 mongrel hounds according to an IACUC-approved protocol. Each animal underwent ovariohysterectomy followed by vaginal cuff closure with Zip-stitch or VICRYL. At two or six weeks, animals were sacrificed for gross and histological analysis. Data Collection. The primary endpoint was the difference in the fraction of vaginal cuff healed six weeks after application of the closure device. Secondary endpoints included histopathologic cellular and tissue responses, including inflammation, necrosis, infection, and vascular and muscle changes. Results. In the test group, there were two distinct locations where fibrotic or granular tissue fusion between the anterior and posterior vaginal walls was observed: in tissue "captured" by a clip or in tissue around the clip. The fraction of the vaginal cuff healed was similar in animals treated with Zip-stitch clips and those treated with sutures at six weeks (68±10% vs 67±18%; P=.148, test for non-inferiority) after surgery. The test article performed similarly or better than the control article in terms of the intensity or extent of the secondary endpoints. Conclusions. Subject to further confirmation, this study supports Zip-stitch clips as a method to maintain immediate post-operative approximation of the vaginal cuff leading to healing but did not achieve statistical significance in its primary endpoint.


Subject(s)
Laparoscopy , Polyglactin 910 , Animals , Dogs , Female , Humans , Hysterectomy/methods , Laparoscopy/methods , Prospective Studies , Suture Techniques , Treatment Outcome , Vagina/surgery
5.
Development ; 145(1)2018 01 08.
Article in English | MEDLINE | ID: mdl-29222391

ABSTRACT

The role of basal suppression of the sonic hedgehog (Shh) pathway and its interaction with Indian hedgehog (Ihh) signaling during limb/skeletal morphogenesis is not well understood. The orphan G protein-coupled receptor Gpr161 localizes to primary cilia and functions as a negative regulator of Shh signaling by promoting Gli transcriptional repressor versus activator formation. Here, we show that forelimb buds are not formed in Gpr161 knockout mouse embryos despite establishment of prospective limb fields. Limb-specific deletion of Gpr161 resulted in prematurely expanded Shh signaling and ectopic Shh-dependent patterning defects resulting in polysyndactyly. In addition, endochondral bone formation in forearms, including formation of both trabecular bone and bone collar was prevented. Endochondral bone formation defects resulted from accumulation of proliferating round/periarticular-like chondrocytes, lack of differentiation into columnar chondrocytes, and corresponding absence of Ihh signaling. Gpr161 deficiency in craniofacial mesenchyme also prevented intramembranous bone formation in calvarium. Defects in limb patterning, endochondral and intramembranous skeletal morphogenesis were suppressed in the absence of cilia. Overall, Gpr161 promotes forelimb formation, regulates limb patterning, prevents periarticular chondrocyte proliferation and drives osteoblastogenesis in intramembranous bones in a cilium-dependent manner.


Subject(s)
Body Patterning/physiology , Forelimb/embryology , Osteogenesis/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Cilia/genetics , Cilia/metabolism , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Skull/embryology
6.
Nature ; 523(7559): 226-30, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26098368

ABSTRACT

Although the adult mammalian heart is incapable of meaningful functional recovery following substantial cardiomyocyte loss, it is now clear that modest cardiomyocyte turnover occurs in adult mouse and human hearts, mediated primarily by proliferation of pre-existing cardiomyocytes. However, fate mapping of these cycling cardiomyocytes has not been possible thus far owing to the lack of identifiable genetic markers. In several organs, stem or progenitor cells reside in relatively hypoxic microenvironments where the stabilization of the hypoxia-inducible factor 1 alpha (Hif-1α) subunit is critical for their maintenance and function. Here we report fate mapping of hypoxic cells and their progenies by generating a transgenic mouse expressing a chimaeric protein in which the oxygen-dependent degradation (ODD) domain of Hif-1α is fused to the tamoxifen-inducible CreERT2 recombinase. In mice bearing the creERT2-ODD transgene driven by either the ubiquitous CAG promoter or the cardiomyocyte-specific α myosin heavy chain promoter, we identify a rare population of hypoxic cardiomyocytes that display characteristics of proliferative neonatal cardiomyocytes, such as smaller size, mononucleation and lower oxidative DNA damage. Notably, these hypoxic cardiomyocytes contributed widely to new cardiomyocyte formation in the adult heart. These results indicate that hypoxia signalling is an important hallmark of cycling cardiomyocytes, and suggest that hypoxia fate mapping can be a powerful tool for identifying cycling cells in adult mammals.


Subject(s)
Myocardium/cytology , Myocytes, Cardiac/cytology , Recombinant Fusion Proteins/metabolism , Animals , Cell Hypoxia , Cell Proliferation/genetics , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinases/genetics , Recombinases/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
7.
Magn Reson Med ; 81(6): 3787-3797, 2019 06.
Article in English | MEDLINE | ID: mdl-30697815

ABSTRACT

PURPOSE: Blood oxygen level dependent (BOLD) MRI based on R2* measurements can provide insights into tumor vascular oxygenation. However, measurements are susceptible to blood flow, which may vary accompanying a hyperoxic gas challenge. We investigated flow sensitivity by comparing R2* measurements with and without flow suppression (fs) in 2 orthotopic lung xenograft tumor models. METHODS: H460 (n = 20) and A549 (n = 20) human lung tumor xenografts were induced by surgical implantation of cancer cells in the right lung of nude rats. MRI was performed at 4.7T after tumors reached 5 to 8 mm in diameter. A multiecho gradient echo MRI sequence was acquired with and without spatial saturation bands on each side of the imaging plane to evaluate the effect of flow on R2* . fs and non-fs R2* MRI measurements were interleaved during an oxygen breathing challenge (from air to 100% O2 ). T2* -weighted signal intensity changes (ΔSI(%)) and R2* measurements were obtained for regions of interest and on a voxel-by-voxel basis and discrepancies quantified with Bland-Altman analysis. RESULTS: Flow suppression affected ΔSI(%) and R2* measurements in each tumor model. Average discrepancy and limits of agreement from Bland-Altman analyses revealed greater flow-related bias in A549 than H460. CONCLUSION: The effect of flow on R2* , and hence BOLD, was tumor model dependent with measurements being more sensitive in well-perfused A549 tumors.


Subject(s)
Lung Neoplasms , Lung , Magnetic Resonance Imaging , Oxygen , A549 Cells , Animals , Female , Heterografts , Humans , Lung/diagnostic imaging , Lung/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oximetry/methods , Oxygen/blood , Oxygen/metabolism , Rats , Rats, Nude
8.
Hum Mol Genet ; 24(2): 424-35, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25209980

ABSTRACT

T-box transcription factor, TBX1, is the major candidate gene for 22q11.2 deletion syndrome (DiGeorge/ Velo-cardio-facial syndrome) characterized by facial defects, thymus hypoplasia, cardiovascular anomalies and cleft palates. Here, we report that the loss of Tbx1 in mouse (Tbx1(-/-)) results in skeletal abnormalities similar to those of cleidocranial dysplasia (CCD) in humans, which is an autosomal-dominant skeletal disease caused by mutations in RUNX2. Tbx1(-/-) mice display short stature, absence of hyoid bone, failed closure of fontanelle, bifid xiphoid process and hypoplasia of clavicle and zygomatic arch. A cell-type-specific deletion of Tbx1 in osteochondro-progenitor (Tbx1(OPKO)) or mesodermal (Tbx1(MKO)) lineage partially recapitulates the Tbx1(-/-) bone phenotypes. Although Tbx1 expression has not been previously reported in neural crest, inactivation of Tbx1 in the neural crest lineage (Tbx1(NCKO)) leads to an absence of the body of hyoid bone and postnatal lethality, indicating an unanticipated role of Tbx1 in neural crest development. Indeed, Tbx1 is expressed in the neural crest-derived hyoid bone primordium, in addition to mesoderm-derived osteochondral progenitors. Ablation of Tbx1 affected Runx2 expression in calvarial bones and overexpression of Tbx1 induced Runx2 expression in vitro. Taken together, our current studies reveal that Tbx1 is required for mesoderm- and neural crest-derived osteoblast differentiation and normal skeletal development. TBX1 mutation could lead to CCD-like bone phenotypes in human.


Subject(s)
Bone and Bones/abnormalities , Cleidocranial Dysplasia/metabolism , T-Box Domain Proteins/deficiency , Animals , Bone and Bones/metabolism , Cell Differentiation , Cleidocranial Dysplasia/embryology , Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Neural Crest/abnormalities , Neural Crest/embryology , Neural Crest/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Phenotype , T-Box Domain Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 111(11): 4109-14, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591619

ABSTRACT

Regeneration of adult skeletal muscle following injury occurs through the activation of satellite cells, an injury-sensitive muscle stem cell population that proliferates, differentiates, and fuses with injured myofibers. Members of the myocyte enhancer factor 2 (MEF2) family of transcription factors play essential roles in muscle differentiation during embryogenesis, but their potential contributions to adult muscle regeneration have not been systematically explored. To investigate the potential involvement of MEF2 factors in muscle regeneration, we conditionally deleted the Mef2a, c, and d genes, singly and in combination, within satellite cells in mice, using tamoxifen-inducible Cre recombinase under control of the satellite cell-specific Pax7 promoter. We show that deletion of individual Mef2 genes has no effect on muscle regeneration in response to cardiotoxin injury. However, combined deletion of the Mef2a, c, and d genes results in a blockade to regeneration. Satellite cell-derived myoblasts lacking MEF2A, C, and D proliferate normally in culture, but cannot differentiate. The absence of MEF2A, C, and D in satellite cells is associated with aberrant expression of a broad collection of known and unique protein-coding and long noncoding RNA genes. These findings reveal essential and redundant roles of MEF2A, C, and D in satellite cell differentiation and identify a MEF2-dependent transcriptome associated with skeletal muscle regeneration.


Subject(s)
Gene Expression Regulation/genetics , Muscle, Skeletal/growth & development , Regeneration/physiology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation/genetics , Flow Cytometry , Immunohistochemistry , MEF2 Transcription Factors/deficiency , MEF2 Transcription Factors/metabolism , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
Proc Natl Acad Sci U S A ; 111(1): E129-38, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24367119

ABSTRACT

Mammalian skeletal muscle can remodel, repair, and regenerate itself by mobilizing satellite cells, a resident population of myogenic progenitor cells. Muscle injury and subsequent activation of myogenic progenitor cells is associated with oxidative stress. Cytoglobin is a hemoprotein expressed in response to oxidative stress in a variety of tissues, including striated muscle. In this study, we demonstrate that cytoglobin is up-regulated in activated myogenic progenitor cells, where it localizes to the nucleus and contributes to cell viability. siRNA-mediated depletion of cytoglobin from C2C12 myoblasts increased levels of reactive oxygen species and apoptotic cell death both at baseline and in response to stress stimuli. Conversely, overexpression of cytoglobin reduced reactive oxygen species levels, caspase activity, and cell death. Mice in which cytoglobin was knocked out specifically in skeletal muscle were generated to examine the role of cytoglobin in vivo. Myogenic progenitor cells isolated from these mice were severely deficient in their ability to form myotubes as compared with myogenic progenitor cells from wild-type littermates. Consistent with this finding, the capacity for muscle regeneration was severely impaired in mice deficient for skeletal-muscle cytoglobin. Collectively, these data demonstrate that cytoglobin serves an important role in muscle repair and regeneration.


Subject(s)
Gene Expression Regulation , Globins/metabolism , Muscles/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Animals , Apoptosis , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Cytoglobin , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/metabolism , Reactive Oxygen Species , Satellite Cells, Skeletal Muscle/pathology , Stem Cells/cytology , Time Factors
12.
Proc Natl Acad Sci U S A ; 111(31): 11521-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049392

ABSTRACT

Members of the calmodulin-binding transcription activator (CAMTA) family of proteins function as calcium-sensitive regulators of gene expression in multicellular organisms ranging from plants to humans. Here, we show that global or nervous system deletion of CAMTA1 in mice causes severe ataxia with Purkinje cell degeneration and cerebellar atrophy, partially resembling the consequences of haploinsufficiency of the human CAMTA1 locus. Gene-expression analysis identified a large collection of neuronal genes that were dysregulated in the brains of CAMTA1-mutant mice, and elucidation of a consensus sequence for binding of CAMTA proteins to DNA revealed the association of CAMTA-binding sites with many of these genes. We conclude that CAMTA1 plays an essential role in the control of Purkinje cell function and survival. CAMTA1-mutant mice provide a model to study the molecular mechanisms of neurodegenerative diseases and for screening potential therapeutic interventions for such disorders.


Subject(s)
Ataxia/metabolism , Ataxia/pathology , Calcium-Binding Proteins/deficiency , Purkinje Cells/metabolism , Purkinje Cells/pathology , Trans-Activators/deficiency , Transcription Factors/deficiency , AT Rich Sequence , Animals , Ataxia/physiopathology , Base Sequence , Binding Sites , Calcium-Binding Proteins/metabolism , Gene Expression Regulation , Integrases/metabolism , Inverted Repeat Sequences/genetics , Male , Mice , Mice, Knockout , Molecular Sequence Data , Motor Activity , Nestin/metabolism , Nucleotide Motifs/genetics , Protein Multimerization , Trans-Activators/metabolism , Transcription Factors/metabolism
13.
Genes Dev ; 23(18): 2166-78, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19720868

ABSTRACT

Vascular injury triggers dedifferentiation and cytoskeletal remodeling of smooth muscle cells (SMCs), culminating in vessel occlusion. Serum response factor (SRF) and its coactivator, myocardin, play a central role in the control of smooth muscle phenotypes by regulating the expression of cytoskeletal genes. We show that SRF and myocardin regulate a cardiovascular-specific microRNA (miRNA) cluster encoding miR-143 and miR-145. To assess the functions of these miRNAs in vivo, we systematically deleted them singly and in combination in mice. Mice lacking both miR-143 and miR-145 are viable and do not display overt abnormalities in smooth muscle differentiation, although they show a significant reduction in blood pressure due to reduced vascular tone. Remarkably, however, neointima formation in response to vascular injury is profoundly impeded in mice lacking these miRNAs, due to disarray of actin stress fibers and diminished migratory activity of SMCs. These abnormalities reflect the regulation of a cadre of modulators of SRF activity and actin dynamics by miR-143 and miR-145. Thus, miR-143 and miR-145 act as integral components of the regulatory network whereby SRF controls cytoskeletal remodeling and phenotypic switching of SMCs during vascular disease.


Subject(s)
Cytoskeleton/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism , Actins/metabolism , Animals , Base Sequence , Carotid Artery Injuries/metabolism , Cells, Cultured , Enhancer Elements, Genetic/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Rats , Sequence Alignment , Trans-Activators/metabolism
14.
Catheter Cardiovasc Interv ; 88(6): 902-911, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27471092

ABSTRACT

BACKGROUND: Metallic endovascular stents are utilized off-label in congenital heart disease. Biodegradable stents (BDS) offer potential advantages in a growing child. We have previously reported double opposed helical (DH) BDS up to 6 mm diameter (DH-6). The objectives are to investigate the bench characteristics of larger 8 mm diameter BDS (DH-8) manufactured with increasing strut thicknesses and the inflammatory profile in a porcine model. METHODS: DH-8 were manufactured with strut thicknesses 0.10, 0.12, and 0.18 mm and mechanical testing performed. Stents were deployed into the infrarenal descending aorta (DAO) of nine minipigs. At insertion (nonsurvival = 2), 1 week (n = 2), 1 month (n = 2), and 9 months (n = 3) follow-up angiography, intravascular ultrasound and histopathology were performed. RESULTS: There was superior recoil and collapse pressure with increasing strut thickness, with 0.18 mm having 1.0% elastic recoil and collapse pressure 0.75 Atmospheres. There was good wall apposition at insertion with 5 BDS (4 DH-8 and 1 DH-6) but suboptimal in 4 as the minipigs infrarenal DAO were >8 mm (deployed at iliac bifurcation). Structural integrity was maintained in 8 BDS with 1 DH-8 collapsed at 9 months, secondary to strut damage at insertion. No thrombosis was seen. There was mild inflammation and neointimal proliferation at 1 week and 1 month, but a moderate inflammatory response at 9 months. CONCLUSIONS: DH-8 with increased strut thickness had acceptable mechanical properties at the cost of an increased inflammatory response. Miniaturization to improve delivery and further investigation on the long-term inflammatory profile of thicker struts, including through degradation, is needed. © 2016 Wiley Periodicals, Inc.


Subject(s)
Aorta, Abdominal/surgery , Aortic Diseases/surgery , Drug-Eluting Stents , Endovascular Procedures/methods , Angiography , Animals , Aorta, Abdominal/diagnostic imaging , Aortic Diseases/diagnosis , Disease Models, Animal , Female , Follow-Up Studies , Prosthesis Design , Swine , Swine, Miniature , Tomography, Optical Coherence , Ultrasonography, Interventional
15.
Proc Natl Acad Sci U S A ; 110(34): 13839-44, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918388

ABSTRACT

The adult mammalian heart has limited potential for regeneration. Thus, after injury, cardiomyocytes are permanently lost, and contractility is diminished. In contrast, the neonatal heart can regenerate owing to sustained cardiomyocyte proliferation. Identification of critical regulators of cardiomyocyte proliferation and quiescence represents an important step toward potential regenerative therapies. Yes-associated protein (Yap), a transcriptional cofactor in the Hippo signaling pathway, promotes proliferation of embryonic cardiomyocytes by activating the insulin-like growth factor and Wnt signaling pathways. Here we report that mice bearing mutant alleles of Yap and its paralog WW domain containing transcription regulator 1 (Taz) exhibit gene dosage-dependent cardiac phenotypes, suggesting redundant roles of these Hippo pathway effectors in establishing proper myocyte number and maintaining cardiac function. Cardiac-specific deletion of Yap impedes neonatal heart regeneration, resulting in a default fibrotic response. Conversely, forced expression of a constitutively active form of Yap in the adult heart stimulates cardiac regeneration and improves contractility after myocardial infarction. The regenerative activity of Yap is correlated with its activation of embryonic and proliferative gene programs in cardiomyocytes. These findings identify Yap as an important regulator of cardiac regeneration and provide an experimental entry point to enhance this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Heart/physiology , Myocytes, Cardiac/physiology , Phosphoproteins/metabolism , Regeneration/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Cell Cycle Proteins , DNA Primers/genetics , Echocardiography , Hippo Signaling Pathway , Histological Techniques , Mice , Mice, Transgenic , Mutation, Missense/genetics , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Tetrazolium Salts , Transcription Factors/genetics , YAP-Signaling Proteins
16.
Catheter Cardiovasc Interv ; 85(4): 629-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25157439

ABSTRACT

OBJECTIVES: This study evaluates the feasibility of delivery and deployment of low and medium molecular weight (LMW and MMW, respectively) double-opposing helical (DH) poly-l-lactic acid biodegradable stent (BDS) in rabbit descending aorta (DAO). Secondary objectives were to assess patency and inflammation of stented vessels at 9 months and to investigate safety following intentional embolization of stent fragments in DAO. BACKGROUND: A BDS that will relieve aortic obstruction and disappears as the child grows older allowing for preservation of aortic wall elasticity and natural growth of aorta will be ideal to treat Coarctation (CoA). BDS have never been evaluated in the DAO. METHODS: Seven New Zealand white rabbits underwent implantation of DH-LMW (n = 7), DH-MMW (n = 3), and metal stents (n = 7) in DAO. BDS fragments were intentionally embolized into DAO in two rabbits. RESULTS: All stents were deployed via a 6-French sheath. Five BDS covered the origin of major DAO side branches. Angiography and intravascular ultrasound showed good stent apposition to the wall of DAO with minimal luminal loss at 9 months follow-up. All stents had minimal neointimal hyperplasia on histopathology. Adverse events included 1 death, 1 aortic aneurysm, and lower extremity ulceration due to self-mutilation in an embolization rabbit. CONCLUSIONS: Pilot study confirms the feasibility of delivery and deployment of up to 6-millimeter diameter DH BDS in rabbit DAO. Stent integrity with DH design was maintained at 9 months with minimal vessel inflammation. Potential morbidity due to embolized BD fragments cannot be ruled out and needs further evaluation.


Subject(s)
Absorbable Implants , Aorta, Thoracic , Aortic Diseases/therapy , Arterial Occlusive Diseases/therapy , Endovascular Procedures/instrumentation , Heart Defects, Congenital/therapy , Stents , Animals , Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/pathology , Aortography , Arterial Occlusive Diseases/diagnosis , Constriction, Pathologic , Disease Models, Animal , Embolism/etiology , Feasibility Studies , Female , Foreign-Body Migration/etiology , Lactic Acid/chemistry , Molecular Weight , Pilot Projects , Polyesters , Polymers/chemistry , Prosthesis Design , Prosthesis Failure , Rabbits , Time Factors , Ultrasonography, Interventional
17.
Mol Cell Probes ; 29(4): 228-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25981184

ABSTRACT

Analysis of nucleic acid amplification products has become the gold standard for applications such as pathogen detection and characterisation of single nucleotide polymorphisms and short tandem repeat sequences. The development of real-time PCR and melting curve analysis using fluorescent probes has simplified nucleic acid analyses. However, the cost of probe synthesis can be prohibitive when developing large panels of tests. We describe an economic two-stage method for probe synthesis, and a new method for nucleic acid sequence analysis which together considerably reduce costs. The analysis method utilises three-strand and four-strand hybridisation complexes for the detection and identification of nucleic acid target sequences by real-time PCR and fluorescence melting.


Subject(s)
DNA Primers/chemical synthesis , Fluorescent Dyes/chemistry , Nucleic Acid Hybridization , Chlamydia trachomatis/isolation & purification , DNA Primers/chemistry , DNA Primers/economics , Nucleic Acid Amplification Techniques
18.
Mol Cell Probes ; 29(2): 92-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25542839

ABSTRACT

Isothermal amplification is a rapid, simple alternative to PCR, with amplification commonly detected using fluorescently labelled oligonucleotide probes, intercalating dyes or increased turbidity as a result of magnesium pyrophosphate generation. SNP identification is possible but requires either allele-specific primers or multiple dye-labelled probes, but further downstream processing is often required for allelic identification. Here we demonstrate that modification of common isothermal amplification methods by the addition of HyBeacon probes permits homogeneous sequence detection and discrimination by melting or annealing curve analysis. Furthermore, we demonstrate that isothermal amplification and sequence discrimination is possible directly from a crude sample such as an expressed buccal swab.


Subject(s)
Molecular Probes/genetics , Nucleic Acid Amplification Techniques/methods , Chlamydia trachomatis/genetics , Chlamydia trachomatis/isolation & purification , Humans , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods , Vitamin K Epoxide Reductases/genetics
19.
Mol Cell Proteomics ; 12(11): 3221-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23938467

ABSTRACT

Spermiogenesis is a postmeiotic process that drives development of round spermatids into fully elongated spermatozoa. Spermatid elongation is largely controlled post-transcriptionally after global silencing of mRNA synthesis from the haploid genome. Here, rats that differentially express EGFP from a lentiviral transgene during early and late steps of spermiogenesis were used to flow sort fractions of round and elongating spermatids. Mass-spectral analysis of 2D gel protein spots enriched >3-fold in each fraction revealed a heterogeneous RNA binding proteome (hnRNPA2/b1, hnRNPA3, hnRPDL, hnRNPK, hnRNPL, hnRNPM, PABPC1, PABPC4, PCBP1, PCBP3, PTBP2, PSIP1, RGSL1, RUVBL2, SARNP2, TDRD6, TDRD7) abundantly expressed in round spermatids prior to their elongation. Notably, each protein within this ontology cluster regulates alternative splicing, sub-cellular transport, degradation and/or translational repression of mRNAs. In contrast, elongating spermatid fractions were enriched with glycolytic enzymes, redox enzymes and protein synthesis factors. Retrogene-encoded proteins were over-represented among the most abundant elongating spermatid factors identified. Consistent with these biochemical activities, plus corresponding histological profiles, the identified RNA processing factors are predicted to collectively drive post-transcriptional expression of an alternative exome that fuels finishing steps of sperm maturation and fitness.


Subject(s)
RNA-Binding Proteins/metabolism , Spermatids/metabolism , Animals , Cell Shape , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Models, Biological , Proteome/genetics , Proteome/metabolism , Proteomics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sperm Maturation/genetics , Sperm Maturation/physiology , Spermatids/cytology , Spermatogenesis/genetics , Spermatogenesis/physiology
20.
Proc Natl Acad Sci U S A ; 109(5): 1649-54, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307625

ABSTRACT

Maintenance of skeletal muscle structure and function requires efficient and precise metabolic control. Autophagy plays a key role in metabolic homeostasis of diverse tissues by recycling cellular constituents, particularly under conditions of caloric restriction, thereby normalizing cellular metabolism. Here we show that histone deacetylases (HDACs) 1 and 2 control skeletal muscle homeostasis and autophagy flux in mice. Skeletal muscle-specific deletion of both HDAC1 and HDAC2 results in perinatal lethality of a subset of mice, accompanied by mitochondrial abnormalities and sarcomere degeneration. Mutant mice that survive the first day of life develop a progressive myopathy characterized by muscle degeneration and regeneration, and abnormal metabolism resulting from a blockade to autophagy. HDAC1 and HDAC2 regulate skeletal muscle autophagy by mediating the induction of autophagic gene expression and the formation of autophagosomes, such that myofibers of mice lacking these HDACs accumulate toxic autophagic intermediates. Strikingly, feeding HDAC1/2 mutant mice a high-fat diet from the weaning age releases the block in autophagy and prevents myopathy in adult mice. These findings reveal an unprecedented and essential role for HDAC1 and HDAC2 in maintenance of skeletal muscle structure and function and show that, at least in some pathological conditions, myopathy may be mitigated by dietary modifications.


Subject(s)
Autophagy , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Homeostasis , Muscle, Skeletal/metabolism , Animals , Electroporation , Mice , Mice, Mutant Strains , Muscle, Skeletal/enzymology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL