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










Publication year range
1.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(5): 535-541, 2024 May 15.
Article in Chinese | MEDLINE | ID: mdl-38752238

ABSTRACT

Objective: To evaluate the effectiveness of using titanium alloy trabecular bone three-dimensional (3D) printed artificial vertebral body in treating cervical ossification of the posterior longitudinal ligament (OPLL). Methods: A retrospective analysis was conducted on clinical data from 45 patients with cervical OPLL admitted between September 2019 and August 2021 and meeting the selection criteria. All patients underwent anterior cervical corpectomy and decompression, interbody bone graft fusion, and titanium plate internal fixation. During operation, 21 patients in the study group received titanium alloy trabecular bone 3D printed artificial vertebral bodies, while 24 patients in the control group received titanium cages. There was no significant difference in baseline data such as gender, age, disease duration, affected segments, or preoperative pain visual analogue scale (VAS) score, Japanese Orthopaedic Association (JOA) score, Neck Disability Index (NDI), vertebral height, and C 2-7Cobb angle ( P>0.05). Operation time, intraoperative blood loss, and occurrence of complications were recorded for both groups. Preoperatively and at 3 and 12 months postoperatively, the functionality and symptom relief were assessed using JOA scores, VAS scores, and NDI evaluations. The vertebral height and C 2-7 Cobb angle were detected by imaging examinations and the implant subsidence and intervertebral fusion were observed. Results: The operation time and incidence of complications were significantly lower in the study group than in the control group ( P<0.05), while the difference in intraoperative blood loss between the two groups was not significant ( P>0.05). All patients were followed up 12-18 months, with the follow-up time of (14.28±4.34) months in the study group and (15.23±3.54) months in the control group, showing no significant difference ( t=0.809, P=0.423). The JOA score, VAS score, and NDI of the two groups improved after operation, and further improved at 12 months compared to 3 months, with significant differences ( P<0.05). At each time point, the study group exhibited significantly higher JOA scores and improvement rate compared to the control group ( P<0.05); but there was no significantly difference in VAS score and NDI between the two groups ( P>0.05). Imaging re-examination showed that the vertebral height and C 2-7Cobb angle of the two groups significantly increased at 3 and 12 months after operation ( P<0.05), and there was no significant difference between 3 and 12 months after operation ( P>0.05). At each time point, the vertebral height and C 2-7Cobb angle of the study group were significantly higher than those of the control group ( P<0.05), and the implant subsidence rate was significantly lower than that of the control group ( P<0.05). However, there was no significant difference in intervertebral fusion rate between the two groups ( P>0.05). Conclusion: Compared to traditional titanium cages, the use of titanium alloy trabecular bone 3D-printed artificial vertebral bodies for treating cervical OPLL results in shorter operative time, fewer postoperative complications, and lower implant subsidence rates, making it superior in vertebral reconstruction.


Subject(s)
Alloys , Cervical Vertebrae , Ossification of Posterior Longitudinal Ligament , Printing, Three-Dimensional , Spinal Fusion , Titanium , Humans , Ossification of Posterior Longitudinal Ligament/surgery , Cervical Vertebrae/surgery , Retrospective Studies , Spinal Fusion/methods , Spinal Fusion/instrumentation , Decompression, Surgical/methods , Cancellous Bone , Treatment Outcome , Vertebral Body/surgery , Female , Male , Bone Plates , Middle Aged
2.
Discov Med ; 34(172): 115-129, 2022.
Article in English | MEDLINE | ID: mdl-36333113

ABSTRACT

Poly (ADP-ribose) polymerase 1 (PARP1) plays an irreplaceable role in the progression of diabetic retinopathy (DR). The m6A methylation in mRNA controls gene expression under various physiological and pathological conditions. However, effects of m6A methylation on PARP1 expression and DR progression at molecular level have not been documented. This study shows that the levels of PARP1, inflammatory factors, and fibrosis markers were significantly upregulated via evaluation by real-time PCR, western blotting, and immunofluorescence in both in vivo and in vitro experiments. EdU, CCK8, and apoptosis assays demonstrate that knockdown of PARP1 not only significantly improved the vitality of hRMECs (human retinal microvascular endothelial cells) even under high glucose conditions but also prevented glucose-induced inflammation, fibrosis, and angiogenesis in vivo. Mechanistically, dot blot, RNA pull-down, and immunoblots were implemented to explore the mechanism of m6A-mediated PARP1 stability and function. PARP1 is identified as a target of YTHDF2-mediated m6A modification. Overexpression of YTHDF2 substantially suppressed PARP1 mRNA m6A modification and inhibited its mRNA expression. Collectively, it has been demonstrated that PARP1 is frequently upregulated in human retinas and contributes to DR progression, and that YTHDF2-mediated m6A modification epigenetically regulates diabetes-induced PARP1 expression. Findings from this work may engender therapeutic targets for treating diabetic retinopathy.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Hyperglycemia , Humans , Diabetic Retinopathy/genetics , Endothelial Cells , Adenosine/metabolism , RNA, Messenger/genetics , Hyperglycemia/genetics , Glucose , Fibrosis , Poly (ADP-Ribose) Polymerase-1/genetics
3.
Nat Commun ; 12(1): 17, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33397957

ABSTRACT

Methanol-to-olefins (MTO), the most important catalytic process producing ethylene and propylene from non-oil feedstocks (coal, natural gas, biomass, CO2, etc.), is hindered by rapid catalyst deactivation due to coke deposition. Common practice to recover catalyst activity, i.e. removing coke via air combustion or steam gasification, unavoidably eliminates the active hydrocarbon pool species (HCPs) favoring light olefins formation. Density functional theory calculations and structured illumination microscopy reveal that naphthalenic cations, active HCPs enhancing ethylene production, are highly stable within SAPO-34 zeolites at high temperature. Here, we demonstrate a strategy of directly transforming coke to naphthalenic species in SAPO-34 zeolites via steam cracking. Fluidized bed reactor-regenerator pilot experiments show that an unexpectedly high light olefins selectivity of 85% is achieved in MTO reaction with 88% valuable CO and H2 and negligible CO2 as byproducts from regeneration under industrial-alike continuous operations. This strategy significantly boosts the economics and sustainability of MTO process.

4.
Circulation ; 134(19): 1467-1483, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27660293

ABSTRACT

BACKGROUND: Bone marrow cell (BMC)-based treatment for critical limb ischemia in diabetic patients yielded a modest therapeutic effect resulting from cell dysfunction. Therefore, approaches that improve diabetic stem/progenitor cell functions may provide therapeutic benefits. Here, we tested the hypothesis that restoration of hydrogen sulfide (H2S) production in diabetic BMCs improves their reparative capacities. METHODS: Mouse BMCs were isolated by density-gradient centrifugation. Unilateral hind limb ischemia was conducted in 12- to 14-week-old db/+ and db/db mice by ligation of the left femoral artery. The H2S level was measured by either gas chromatography or staining with florescent dye sulfidefluor 7 AM. RESULTS: Both H2S production and cystathionine γ-lyase (CSE), an H2S enzyme, levels were significantly decreased in BMCs from diabetic db/db mice. Administration of H2S donor diallyl trisulfide (DATS) or overexpression of CSE restored H2S production and enhanced cell survival and migratory capacity in high glucose (HG)-treated BMCs. Immediately after hind limb ischemia surgery, the db/+ and db/db mice were administered DATS orally and/or given a local intramuscular injection of green fluorescent protein-labeled BMCs or red fluorescent protein-CSE-overexpressing BMCs (CSE-BMCs). Mice with hind limb ischemia were divided into 6 groups: db/+, db/db, db/db+BMCs, db/db+DATS, db/db+DATS+BMCs, and db/db+CSE-BMCs. DATS and CSE overexpression greatly enhanced diabetic BMC retention in ischemic hind limbs followed by improved blood perfusion, capillary/arteriole density, skeletal muscle architecture, and cell survival and decreased perivascular CD68+ cell infiltration in the ischemic hind limbs of diabetic mice. It is interesting to note that DATS or CSE overexpression rescued high glucose-impaired migration, tube formation, and survival of BMCs or mature human cardiac microvascular endothelial cells. Moreover, DATS restored nitric oxide production and decreased endothelial nitric oxide synthase phosphorylation at threonine 495 levels in human cardiac microvascular endothelial cells and improved BMC angiogenic activity under high glucose condition. Last, silencing CSE by siRNA significantly increased endothelial nitric oxide synthase phosphorylation at threonine 495 levels in human cardiac microvascular endothelial cells. CONCLUSIONS: Decreased CSE-mediated H2S bioavailability is an underlying source of BMC dysfunction in diabetes mellitus. Our data indicate that H2S and overexpression of CSE in diabetic BMCs may rescue their dysfunction and open novel avenues for cell-based therapeutics of critical limb ischemia in diabetic patients.


Subject(s)
Bone Marrow Transplantation , Diabetes Mellitus, Experimental , Diabetic Angiopathies , Hindlimb/blood supply , Hydrogen Sulfide/blood , Ischemia , Allografts , Animals , Bone Marrow Cells/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Diabetic Angiopathies/blood , Diabetic Angiopathies/therapy , Humans , Ischemia/blood , Ischemia/therapy , Male , Mice
6.
Circ Res ; 118(8): 1208-22, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26976650

ABSTRACT

RATIONALE: Cardiac myocyte-specific deletion of either glycogen synthase kinase (GSK)-3α and GSK-3ß leads to cardiac protection after myocardial infarction, suggesting that deletion of both isoforms may provide synergistic protection. This is an important consideration because of the fact that all GSK-3-targeted drugs, including the drugs already in clinical trial target both isoforms of GSK-3, and none are isoform specific. OBJECTIVE: To identify the consequences of combined deletion of cardiac myocyte GSK-3α and GSK-3ß in heart function. METHODS AND RESULTS: We generated tamoxifen-inducible cardiac myocyte-specific mice lacking both GSK-3 isoforms (double knockout). We unexpectedly found that cardiac myocyte GSK-3 is essential for cardiac homeostasis and overall survival. Serial echocardiographic analysis reveals that within 2 weeks of tamoxifen treatment, double-knockout hearts leads to excessive dilatative remodeling and ventricular dysfunction. Further experimentation with isolated adult cardiac myocytes and fibroblasts from double-knockout implicated cardiac myocytes intrinsic factors responsible for observed phenotype. Mechanistically, loss of GSK-3 in adult cardiac myocytes resulted in induction of mitotic catastrophe, a previously unreported event in cardiac myocytes. Double-knockout cardiac myocytes showed cell cycle progression resulting in increased DNA content and multinucleation. However, increased cell cycle activity was rivaled by marked activation of DNA damage, cell cycle checkpoint activation, and mitotic catastrophe-induced apoptotic cell death. Importantly, mitotic catastrophe was also confirmed in isolated adult cardiac myocytes. CONCLUSIONS: Together, our findings suggest that cardiac myocyte GSK-3 is required to maintain normal cardiac homeostasis, and its loss is incompatible with life because of cell cycle dysregulation that ultimately results in a severe fatal dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/mortality , Glycogen Synthase Kinase 3/deficiency , Mitosis/physiology , Myocytes, Cardiac/metabolism , Animals , Cardiomyopathy, Dilated/pathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology
7.
J Am Coll Cardiol ; 64(7): 696-706, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25125302

ABSTRACT

BACKGROUND: Injury due to myocardial infarction (MI) is largely irreversible. Once an infarct has occurred, the clinical goal becomes limiting remodeling, preserving left ventricular function, and preventing heart failure. Although traditional approaches (e.g., ß-blockers) partially preserve left ventricular function, novel strategies are needed to limit ventricular remodeling post-MI. OBJECTIVES: The aim of this study was to determine the role of glycogen synthase kinase-3α (GSK-3α) in post-MI remodeling. METHODS: Mice with cardiomyocyte-specific conditional deletion of Gsk3α and littermate controls underwent sham or MI surgery. Heart function was assessed using serial M-mode echocardiography. RESULTS: Gsk3α deletion in the heart markedly limits remodeling and preserves left ventricular function post-MI. This is due at least in part to dramatic thinning and expansion of the scar in the control hearts, which was less in the heart of knockout (KO) mice. In contrast, the border zone in the KO mice demonstrated a much thicker scar, and there were more viable cardiomyocytes within the scar/border zone. This was associated with less apoptosis and more proliferation of cardiomyocytes in the KO mice. Mechanistically, reduced apoptosis was due, at least in part, to a marked decrease in the Bax/Bcl-2 ratio, and increased cardiomyocyte proliferation was mediated through cyclin E1 and E2F-1 in the hearts of the KO mice. CONCLUSIONS: Taken together, these findings show that reducing GSK-3α expression in cardiomyocytes limits ventricular remodeling and preserves cardiac function post-MI. Specifically targeting GSK-3α could be a novel strategy to limit adverse remodeling and heart failure.


Subject(s)
Gene Deletion , Glycogen Synthase Kinase 3/genetics , Heart Failure/genetics , Myocardial Infarction/complications , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Remodeling/genetics , Animals , Apoptosis , Cell Proliferation , DNA/genetics , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Heart Failure/etiology , Heart Failure/physiopathology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology
8.
Circulation ; 130(5): 419-30, 2014 07 29.
Article in English | MEDLINE | ID: mdl-24899689

ABSTRACT

BACKGROUND: Myocardial infarction-induced remodeling includes chamber dilatation, contractile dysfunction, and fibrosis. Of these, fibrosis is the least understood. After myocardial infarction, activated cardiac fibroblasts deposit extracellular matrix. Current therapies to prevent fibrosis are inadequate, and new molecular targets are needed. METHODS AND RESULTS: Herein we report that glycogen synthase kinase-3ß (GSK-3ß) is phosphorylated (inhibited) in fibrotic tissues from ischemic human and mouse heart. Using 2 fibroblast-specific GSK-3ß knockout mouse models, we show that deletion of GSK-3ß in cardiac fibroblasts leads to fibrogenesis, left ventricular dysfunction, and excessive scarring in the ischemic heart. Deletion of GSK-3ß induces a profibrotic myofibroblast phenotype in isolated cardiac fibroblasts, in post-myocardial infarction hearts, and in mouse embryonic fibroblasts deleted for GSK-3ß. Mechanistically, GSK-3ß inhibits profibrotic transforming growth factor-ß1/SMAD-3 signaling via interactions with SMAD-3. Moreover, deletion of GSK-3ß resulted in the significant increase of SMAD-3 transcriptional activity. This pathway is central to the pathology because a small-molecule inhibitor of SMAD-3 largely prevented fibrosis and limited left ventricular remodeling. CONCLUSIONS: These studies support targeting GSK-3ß in myocardial fibrotic disorders and establish critical roles of cardiac fibroblasts in remodeling and ventricular dysfunction.


Subject(s)
Fibroblasts/enzymology , Glycogen Synthase Kinase 3/metabolism , Myocardial Ischemia/metabolism , Myocardium/enzymology , Ventricular Remodeling/physiology , Aged , Animals , Enzyme Activation/physiology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibrosis/metabolism , Fibrosis/pathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice, Knockout , Middle Aged , Myocardial Ischemia/pathology , Myocardium/cytology , Primary Cell Culture , RNA, Small Interfering/genetics , Smad3 Protein/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
10.
J Clin Invest ; 123(4): 1821-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23549082

ABSTRACT

Aging is regulated by conserved signaling pathways. The glycogen synthase kinase-3 (GSK-3) family of serine/threonine kinases regulates several of these pathways, but the role of GSK-3 in aging is unknown. Herein, we demonstrate premature death and acceleration of age-related pathologies in the Gsk3a global KO mouse. KO mice developed cardiac hypertrophy and contractile dysfunction as well as sarcomere disruption and striking sarcopenia in cardiac and skeletal muscle, a classical finding in aging. We also observed severe vacuolar degeneration of myofibers and large tubular aggregates in skeletal muscle, consistent with impaired clearance of insoluble cellular debris. Other organ systems, including gut, liver, and the skeletal system, also demonstrated age-related pathologies. Mechanistically, we found marked activation of mTORC1 and associated suppression of autophagy markers in KO mice. Loss of GSK-3α, either by pharmacologic inhibition or Gsk3a gene deletion, suppressed autophagy in fibroblasts. mTOR inhibition rescued this effect and reversed the established pathologies in the striated muscle of the KO mouse. Thus, GSK-3α is a critical regulator of mTORC1, autophagy, and aging. In its absence, aging/senescence is accelerated in multiple tissues. Strategies to maintain GSK-3α activity and/or inhibit mTOR in the elderly could retard the appearance of age-related pathologies.


Subject(s)
Cardiovascular Diseases/enzymology , Glycogen Synthase Kinase 3/genetics , Sarcopenia/enzymology , Aging , Animals , Autophagy , Bone and Bones/pathology , Cellular Senescence , Everolimus , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hepatocytes/enzymology , Hepatocytes/physiology , Indoles/pharmacology , Kaplan-Meier Estimate , Knee Joint/pathology , Liver/enzymology , Liver/pathology , Maleimides/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Myocardium/enzymology , Myocardium/pathology , Phenotype , Proteins/antagonists & inhibitors , Proteins/metabolism , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
11.
Circulation ; 125(1): 65-75, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22086876

ABSTRACT

BACKGROUND: The molecular pathways that regulate the extent of ischemic injury and post-myocardial infarction (MI) remodeling are not well understood. We recently demonstrated that glycogen synthase kinase-3α (GSK-3α) is critical to the heart's response to pressure overload. However, the role, if any, of GSK-3α in regulating ischemic injury and its consequences is not known. METHODS AND RESULTS: MI was induced in wild-type (WT) versus GSK-3α((-/-)) (KO) littermates by left anterior descending coronary artery ligation. Pre-MI, WT, and KO hearts had comparable chamber dimensions and ventricular function, but as early as 1 week post-MI, KO mice had significantly more left ventricular dilatation and dysfunction than WT mice. KO mice also had increased mortality during the first 10 days post-MI (43% versus 22%; P=0.04), and postmortem examination confirmed cardiac rupture as the cause of most of the deaths. In the mice that survived the first 10 days, left ventricular dilatation and dysfunction remained worse in the KO mice throughout the study (8 weeks). Hypertrophy, fibrosis, and heart failure were all increased in the KO mice. Given the early deaths due to rupture and the significant reduction in left ventricular function evident as early as 1 week post-MI, we examined infarct size following a 48-hour coronary artery ligation and found it to be increased in the KO mice. This was accompanied by increased apoptosis in the border zone of the MI. This increased susceptibility to ischemic injury-induced apoptosis was also seen in cardiomyocytes isolated from the KO mice that were exposed to hypoxia. Finally, Bax translocation to the mitochondria and cytochrome C release into the cytosol were increased in the KO mice. CONCLUSION: GSK-3α confers resistance to ischemic injury, at least in part, via limiting apoptosis. Loss of GSK-3α promotes ischemic injury, increases risk of cardiac rupture, accentuates post-MI remodeling and left ventricular dysfunction, and increases the progression to heart failure. These findings are in striking contrast to multiple previous reports in which deletion or inhibition of GSK-3ß is protective.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Heart Rupture/enzymology , Myocardial Infarction/enzymology , Ventricular Remodeling/physiology , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Cardiotonic Agents/metabolism , Cardiotonic Agents/therapeutic use , Cells, Cultured , Death , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Heart Rupture/genetics , Heart Rupture/mortality , Male , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/mortality , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Ventricular Remodeling/genetics
12.
J Clin Invest ; 120(7): 2280-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20516643

ABSTRACT

The glycogen synthase kinase-3 (GSK-3) family of serine/threonine kinases consists of 2 highly related isoforms, alpha and beta. Although GSK-3beta has an important role in cardiac development, much remains unknown about the function of either GSK-3 isoform in the postnatal heart. Herein, we present what we believe to be the first studies defining the role of GSK-3alpha in the mouse heart using gene targeting. Gsk3a(-/-) mice over 2 months of age developed progressive cardiomyocyte and cardiac hypertrophy and contractile dysfunction. Following thoracic aortic constriction in young mice, we observed enhanced hypertrophy that rapidly transitioned to ventricular dilatation and contractile dysfunction. Surprisingly, markedly impaired beta-adrenergic responsiveness was found at both the organ and cellular level. This phenotype was reproduced by acute treatment of WT cardiomyocytes with a small molecule GSK-3 inhibitor, confirming that the response was not due to a chronic adaptation to LV dysfunction. Thus, GSK-3alpha appears to be the central regulator of a striking range of essential processes, including acute and direct positive regulation of beta-adrenergic responsiveness. In the absence of GSK-3alpha, the heart cannot respond effectively to hemodynamic stress and rapidly fails. Our findings identify what we believe to be a new paradigm of regulation of beta-adrenergic signaling and raise concerns given the rapid expansion of drug development targeting GSK-3.


Subject(s)
Glycogen Synthase Kinase 3 , Heart/growth & development , Adrenergic Agents , Animals , Cardiomegaly/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Hemodynamics/genetics , Hypertrophy/genetics , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Norepinephrine/genetics , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics
13.
Circ Res ; 106(10): 1635-45, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20360256

ABSTRACT

RATIONALE: Numerous studies have proposed that glycogen synthase kinase (GSK)-3beta is a central regulator of the hypertrophic response of cardiomyocytes. However, all of this work has relied on overexpression of GSK-3beta, expression of constitutively active mutants, or small molecule inhibitors with documented off-target effects. Genetic loss of function approaches have not been used in the adult mouse because germ-line deletion of GSK-3beta is embryonic-lethal. OBJECTIVE: This study was designed to define the role played by GSK-3beta in pressure overload (PO)-induced hypertrophy and remodeling following myocardial infarction (MI). METHODS AND RESULTS: We used a mouse model that allows inducible, cardiomyocyte-specific deletion of GSK-3beta in the adult knockout. Surprisingly, we find that knockout mice exposed to PO induced by thoracic aortic constriction exhibit a normal hypertrophic response. Thus, in contrast to virtually all prior published studies, GSK-3beta appears to play at most a minor role in the hypertrophic response to PO stress. However, GSK-3beta does regulate post-MI remodeling because the GSK-3beta knockouts had less left ventricular dilatation and better-preserved left ventricular function at up to 8 weeks post-MI despite demonstrating significantly more hypertrophy in the remote myocardium. Deletion of GSK-3beta also led to increased cardiomyocyte proliferation following PO and MI. CONCLUSIONS: Deletion of GSK-3beta protects against post-MI remodeling and promotes stress-induced cardiomyocyte proliferation in the adult heart. These studies suggest that inhibition of GSK-3beta could be a strategy to both prevent remodeling and to promote cardiac regeneration in pathological states.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Ventricular Remodeling/physiology , Animals , Aorta, Thoracic/pathology , Cardiomegaly/pathology , Cell Division , Exons , Gene Deletion , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Promoter Regions, Genetic , Vasoconstriction
14.
Mol Cell Biol ; 29(15): 4167-76, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19451227

ABSTRACT

Oxidant stress is a ubiquitous stressor with negative impacts on multiple cell types. ASK1 is a central mediator of oxidant injury, but while mechanisms of its inhibition, such as sequestration by 14-3-3 proteins and thioredoxin, have been identified, mechanisms of activation have remained obscure and the signaling pathways regulating this are not clear. Here, we report that phosphorylation of 14-3-3zeta at serine 58 (S58) is dynamically regulated in the cell and that the phosphorylation status of S58 is a critical factor regulating oxidant stress-induced cell death. Phosphorylation of S58 releases ASK1 from 14-3-3zeta, and ASK1 then activates stress-activated protein kinases, leading to cell death. While several members of the mammalian sterile 20 (Mst) family of kinases can phosphorylate S58 when overexpressed, we identify Ste20/oxidant stress response kinase 1 (SOK-1), an Mst family member known to be activated by oxidant stress, as a central endogenous regulator of S58 phosphorylation and thereby of ASK1-mediated cell death. Our findings identify a novel pathway that regulates ASK1 activation and oxidant stress-induced cell death.


Subject(s)
14-3-3 Proteins/metabolism , Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase Kinase 5/metabolism , Serine/metabolism , 14-3-3 Proteins/genetics , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinase 5/genetics , Mice , Mice, Knockout , Mutation , Oxidants/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Serine/genetics
15.
J Clin Invest ; 118(11): 3609-18, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18830417

ABSTRACT

Based on extensive preclinical data, glycogen synthase kinase-3 (GSK-3) has been proposed to be a viable drug target for a wide variety of disease states, ranging from diabetes to bipolar disorder. Since these new drugs, which will be more powerful GSK-3 inhibitors than lithium, may potentially be given to women of childbearing potential, and since it has controversially been suggested that lithium therapy might be linked to congenital cardiac defects, we asked whether GSK-3 family members are required for normal heart development in mice. We report that terminal cardiomyocyte differentiation was substantially blunted in Gsk3b(-/-) embryoid bodies. While GSK-3alpha-deficient mice were born without a cardiac phenotype, no live-born Gsk3b(-/-) pups were recovered. The Gsk3b(-/-) embryos had a double outlet RV, ventricular septal defects, and hypertrophic myopathy, with near obliteration of the ventricular cavities. The hypertrophic myopathy was caused by cardiomyocyte hyperproliferation without hypertrophy and was associated with increased expression and nuclear localization of three regulators of proliferation - GATA4, cyclin D1, and c-Myc. These studies, which we believe are the first in mammals to examine the role of GSK-3alpha and GSK-3beta in the heart using loss-of-function approaches, implicate GSK-3beta as a central regulator of embryonic cardiomyocyte proliferation and differentiation, as well as of outflow tract development. Although controversy over the teratogenic effects of lithium remains, our studies suggest that caution should be exercised in the use of newer, more potent drugs targeting GSK-3 in women of childbearing age.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cell Proliferation , Gene Deletion , Glycogen Synthase Kinase 3/genetics , Myoblasts, Cardiac/physiology , Animals , Cardiomyopathy, Hypertrophic/embryology , Cardiomyopathy, Hypertrophic/metabolism , Cell Differentiation/genetics , Cell Size , Embryo, Mammalian , Glycogen Synthase Kinase 3 beta , Mice , Mice, Knockout
16.
J Mol Cell Cardiol ; 43(3): 319-26, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17673255

ABSTRACT

In addition to its role in cell adhesion, beta-catenin is an important signaling molecule in the Wnt/Wingless signaling pathway. Recent studies have indicated that beta-catenin is stabilized by hypertrophic stimuli and may regulate cardiac hypertrophic responses. To explore the role and requirement of beta-catenin in cardiac development and hypertrophy, we deleted the beta-catenin gene specifically in cardiac myocytes by crossing loxP-floxed beta-catenin mice with transgenic mice expressing a Cre recombinase under the control of the alpha-myosin heavy chain promoter. No homozygous beta-catenin-deleted mice were born alive and died before embryonic day 14.5, indicating significant and irreplaceable roles of beta-catenin in embryonic heart development. Heterozygous beta-catenin-deleted mice, however, demonstrated no structural and functional abnormality. The response of heterozygous beta-catenin-deleted mice to transverse aortic constriction, however, was significantly attenuated with decreased heart weight and heart weight/body weight ratio compared to controls with intact beta-catenin genes. Hemodynamic analysis revealed that there was no difference in cardiac function between wild-type and heterozygous beta-catenin-deleted mice. On the other hand, the expression of fetal genes, beta-myosin heavy chain, atrial and brain natriuretic peptides was significantly higher in heterozygous beta-catenin-deleted mice when compared to wild-type beta-catenin mice. These results suggest that the cytoplasmic level of beta-catenin modulates hypertrophic response and fetal gene reprogramming after pressure overload.


Subject(s)
Aortic Coarctation/complications , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Gene Expression , beta Catenin/metabolism , Animals , Cells, Cultured , Echocardiography , Gene Deletion , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Transgenes , beta Catenin/genetics
17.
Am J Physiol Heart Circ Physiol ; 292(1): H270-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16936006

ABSTRACT

Recent progresses in signal transduction have revealed that beta-catenin signaling controls embryonic development, tumorigenesis, cell shape, and polarity. The role of this pathway in myocyte shape regulation during cardiac hypertrophy and failure is, however, not clearly defined. Since homozygous knockout of beta-catenin is embryonically lethal, we have deleted beta-catenin genes specifically in the heart of adult mice by crossing loxP-flanked beta-catenin mice with transgenic mice expressing tamoxifen-activated MerCreMer protein (MCM) driven by the alpha-myosin heavy chain promoter. Administration of tamoxifen to homozygous loxP-flanked beta-catenin mice positive for MCM induces the deletion of beta-catenin only in cardiomyocytes. Immunolabeling with beta-catenin antibody demonstrates that 90% of cardiomyocytes completely lose their beta-catenin expression but maintain normal rod-shaped morphology. The intercalated disk of cardiomyocytes lacking beta-catenin is morphologically unremarkable with normal distribution of vinculin, N-cadherin, desmoplakin, ZO-1, connexin43, and alpha-, gamma-, and p120 catenins. The expression level of these proteins, except that of gamma-catenin, is also similar in tamoxifen-treated and control mice with both homozygous loxP-flanked beta-catenin genes and the MCM transgene. Western blot analyses reveal that gamma-catenin increases in the heart of beta-catenin knockout mice compared with controls. Confocal microscopy also demonstrates that gamma-catenin has significantly increased in the intercalated disk of cardiomyocytes lacking beta-catenin. Echocardiographic data indicate that the knockout mice maintain normal ventricular geometry and cardiac function. The results suggest that upregulation of gamma-catenin can compensate for the loss of beta-catenin in cardiomyocytes to maintain normal cardiac structure and function.


Subject(s)
Adherens Junctions/metabolism , Myocytes, Cardiac/metabolism , beta Catenin/metabolism , gamma Catenin/metabolism , Adherens Junctions/ultrastructure , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Up-Regulation , beta Catenin/genetics
18.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 23(4): 903-6, 2006 Aug.
Article in Chinese | MEDLINE | ID: mdl-17002135

ABSTRACT

Ligaments are the main parts which stabilize the knee joint. How to analog the ligaments in biomechanical model will affect the characteristics of the human knee dynamics and in the computation of the stress in ligaments between two bones. This symposium is aimed at the survey of the simplified method of the ligaments via mechanical parameters, and providing an exact method of constructing model.


Subject(s)
Knee Joint/physiology , Ligaments, Articular/physiology , Models, Biological , Biomechanical Phenomena , Humans , Knee Joint/anatomy & histology , Ligaments, Articular/anatomy & histology , Models, Anatomic
19.
Am J Physiol Heart Circ Physiol ; 290(6): H2509-15, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16373587

ABSTRACT

Focal adhesion kinase (FAK) and FAK-related non-kinase (FRNK) accumulate in the nucleus of cardiac myocytes during hypertensive hypertrophy. Nuclear FAK and FRNK are phosphorylated on different serines and form distinct bright spots. The subnuclear distribution of serine-phosphorylated FAK and FRNK was examined in this study by double labeling with fibrillarin, a component of nucleoli, and Sam68, a constituent of Sam68 nuclear bodies. We also investigated the role of protein kinase C (PKC)-mediated phosphorylation of FAK and FRNK on nuclear translocation. PKC activation by 12-O-tetradecanoylphorbol 13-acetate treatment increased serine phosphorylation of FAK and FRNK. Specifically, FAK was phosphorylated on serine 722 but not serine 910. On the other hand, FRNK was phosphorylated on serine 217, the equivalent site of FAK serine 910, but not serine 30, the homologous site of FAK serine 722. Serine-phosphorylated FAK and FRNK redistributed into the nucleus and formed distinct patterns. FAK with phosphorylation on serine 722 colocalized with Sam68 but not fibrillarin. On the contrary, FRNK phosphorylated on 217 coexisted with fibrillarin but not Sam68. Immunoprecipitation also confirmed that FAK associated with Sam68 and FRNK interacted with fibrillarin, respectively. These results suggest that FAK and FRNK target different nuclear subdomains by their association with distinct nuclear proteins.


Subject(s)
Cell Nucleus/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Myocytes, Cardiac/enzymology , Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cell Separation , Chromosomal Proteins, Non-Histone/pharmacology , DNA-Binding Proteins/metabolism , Female , Fluorescent Antibody Technique , Heart Failure/enzymology , Heart Failure/genetics , Immunoprecipitation , Microscopy, Confocal , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology
20.
Anat Rec A Discov Mol Cell Evol Biol ; 282(1): 13-23, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15584034

ABSTRACT

G-protein-coupled receptor kinases (GRKs) are involved in cardiac hypertrophy and failure. But their temporal expression and cellular localization during the development of hypertrophy and its transition to failure remains to be investigated. In this study, we determined the expression and subcellular distribution of GRK2, GRK3, GRK5, and GRK6 in cardiac myocytes of 2- to 24-month-old spontaneously hypertensive heart failure (SHHF) rats. GRK2 increased in the intercalated disks in 6-, 12-, and 24-month-old SHHF rats, although total expression remained relatively constant from 2 to 24 months in both SHHF and normotensive rats. GRK3 expression progressively increased in 6-, 12-, and 24-month-old SHHF rats and was significantly higher than in age-matched controls. Immunolabeling of GRK3 showed a typical pattern of cross-striations that colocalized with alpha-actinin and G(alphas) at Z-lines in both SHHF and control rats. GRK5 expression showed no change from 2 to 24 months in both SHHF and normotensive rats. Confocal analysis revealed nuclear translocation of GRK5 in myocytes of SHHF rats. GRK6 had a striated pattern colocalized with alpha-actinin at Z-lines in the cytoplasm and was also present in the intercalated disks of cardiac myocytes from both SHHF and control rats. GRK6 expression increased in 12- and 24-month-old SHHF rats and was significantly higher than in age-matched controls. GRK6 labeling was reduced at the intercalated disks, but increased in the cytoplasm of cardiac myocytes from SHHF rats compared to age-matched controls. The increased expression of GRK3 and GRK6 and subcellular redistribution of GRK2, GRK5, and GRK6 in SHHF rats may be involved in abnormal remodeling of cardiac myocytes in hypertensive hypertrophy and failure.


Subject(s)
Heart Failure/enzymology , Hypertrophy, Left Ventricular/enzymology , Myocardium/enzymology , Rats, Inbred SHR/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Nucleus/enzymology , Cell Nucleus/pathology , Disease Models, Animal , Female , Fluorescent Antibody Technique, Indirect , Heart Failure/genetics , Heart Failure/pathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Microscopy, Confocal , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...