Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38949658

ABSTRACT

Contact sites between lipid droplets and other organelles are essential for cellular lipid and energy homeostasis upon metabolic demands. Detection of these contact sites at the nanometer scale over time in living cells is challenging. We developed a tool kit for detecting contact sites based on fluorogen-activated bimolecular complementation at CONtact sites, FABCON, using a reversible, low-affinity split fluorescent protein, splitFAST. FABCON labels contact sites with minimal perturbation to organelle interaction. Via FABCON, we quantitatively demonstrated that endoplasmic reticulum (ER)- and mitochondria (mito)-lipid droplet contact sites are dynamic foci in distinct metabolic conditions, such as during lipid droplet biogenesis and consumption. An automated analysis pipeline further classified individual contact sites into distinct subgroups based on size, likely reflecting differential regulation and function. Moreover, FABCON is generalizable to visualize a repertoire of organelle contact sites including ER-mito. Altogether, FABCON reveals insights into the dynamic regulation of lipid droplet-organelle contact sites and generates new hypotheses for further mechanistical interrogation during metabolic regulation.


Subject(s)
Endoplasmic Reticulum , Lipid Droplets , Mitochondria , Lipid Droplets/metabolism , Humans , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Lipid Metabolism , HeLa Cells , HEK293 Cells , Luminescent Proteins/metabolism , Luminescent Proteins/genetics
2.
Drug Resist Updat ; 72: 101017, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37988981

ABSTRACT

The role of ABCC4, an ATP-binding cassette transporter, in the process of platelet formation, megakaryopoiesis, is unknown. Here, we show that ABCC4 is highly expressed in megakaryocytes (MKs). Mining of public genomic data (ATAC-seq and genome wide chromatin interactions, Hi-C) revealed that key megakaryopoiesis transcription factors (TFs) interacted with ABCC4 regulatory elements and likely accounted for high ABCC4 expression in MKs. Importantly these genomic interactions for ABCC4 ranked higher than for genes with known roles in megakaryopoiesis suggesting a role for ABCC4 in megakaryopoiesis. We then demonstrate that ABCC4 is required for optimal platelet formation as in vitro differentiation of fetal liver derived MKs from Abcc4-/- mice exhibited impaired proplatelet formation and polyploidization, features required for optimal megakaryopoiesis. Likewise, a human megakaryoblastic cell line, MEG-01 showed that acute ABCC4 inhibition markedly suppressed key processes in megakaryopoiesis and that these effects were related to reduced cAMP export and enhanced dissociation of a negative regulator of megakaryopoiesis, protein kinase A (PKA) from ABCC4. PKA activity concomitantly increased after ABCC4 inhibition which was coupled with significantly reduced GATA-1 expression, a TF needed for optimal megakaryopoiesis. Further, ABCC4 protected MKs from 6-mercaptopurine (6-MP) as Abcc4-/- mice show a profound reduction in MKs after 6-MP treatment. In total, our studies show that ABCC4 not only protects the MKs but is also required for maximal platelet production from MKs, suggesting modulation of ABCC4 function might be a potential therapeutic strategy to regulate platelet production.


Subject(s)
Blood Platelets , Megakaryocytes , Animals , Humans , Mice , ATP-Binding Cassette Transporters/metabolism , Blood Platelets/metabolism , Cell Differentiation , Megakaryocytes/metabolism , Mercaptopurine/pharmacology , Mercaptopurine/metabolism , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism
3.
bioRxiv ; 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38076863

ABSTRACT

Contact sites between lipid droplets and other organelles are essential for cellular lipid and energy homeostasis. Detection of these contact sites at nanometer scale over time in living cells is challenging. Here, we developed a tool kit for detecting contact sites based on Fluorogen-Activated Bimolecular complementation at CONtact sites, FABCON, using a reversible, low affinity split fluorescent protein, splitFAST. FABCON labels contact sites with minimal perturbation to organelle interaction. Via FABCON, we quantitatively demonstrated that endoplasmic reticulum (ER)- and mitochondria (mito)-lipid droplet contact sites are dynamic foci in distinct metabolic conditions, such as during lipid droplet biogenesis and consumption. An automated analysis pipeline further classified individual contact sites into distinct subgroups based on size, likely reflecting differential regulation and function. Moreover, FABCON is generalizable to visualize a repertoire of organelle contact sites including ER-mito. Altogether, FABCON reveals insights into the dynamic regulation of lipid droplet-organelle contact sites and generates new hypotheses for further mechanistical interrogation during metabolic switch.

4.
J Cell Biol ; 222(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37093123

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the etiologic agent for the global COVID-19 pandemic, triggers the formation of endoplasmic reticulum (ER)-derived replication organelles, including double-membrane vesicles (DMVs), in the host cell to support viral replication. Here, we clarify how SARS-CoV-2 hijacks host factors to construct the DMVs. We show that the ER morphogenic proteins reticulon-3 (RTN3) and RTN4 help drive DMV formation, enabling viral replication, which leads to productive infection. Different SARS-CoV-2 variants, including the delta variant, use the RTN-dependent pathway to promote infection. Mechanistically, our results reveal that the membrane-embedded reticulon homology domain (RHD) of the RTNs is sufficient to functionally support viral replication and physically engage NSP3 and NSP4, two viral non-structural membrane proteins known to induce DMV formation. Our findings thus identify the ER morphogenic RTN3 and RTN4 membrane proteins as host factors that help promote the biogenesis of SARS-CoV-2-induced DMVs, which can act as viral replication platforms.


Subject(s)
Endoplasmic Reticulum , Membrane Proteins , Organelles , SARS-CoV-2 , Humans , COVID-19/virology , Endoplasmic Reticulum/virology , Membrane Proteins/metabolism , Pandemics , SARS-CoV-2/physiology , Virus Replication , Organelles/virology , Viral Nonstructural Proteins/metabolism
5.
Nat Commun ; 13(1): 5351, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36096887

ABSTRACT

The mannose-6-phosphate (M6P) biosynthetic pathway for lysosome biogenesis has been studied for decades and is considered a well-understood topic. However, whether this pathway is regulated remains an open question. In a genome-wide CRISPR/Cas9 knockout screen, we discover TMEM251 as the first regulator of the M6P modification. Deleting TMEM251 causes mistargeting of most lysosomal enzymes due to their loss of M6P modification and accumulation of numerous undigested materials. We further demonstrate that TMEM251 localizes to the Golgi and is required for the cleavage and activity of GNPT, the enzyme that catalyzes M6P modification. In zebrafish, TMEM251 deletion leads to severe developmental defects including heart edema and skeletal dysplasia, which phenocopies Mucolipidosis Type II. Our discovery provides a mechanism for the newly discovered human disease caused by TMEM251 mutations. We name TMEM251 as GNPTAB cleavage and activity factor (GCAF) and its related disease as Mucolipidosis Type V.


Subject(s)
Membrane Proteins , Mucolipidoses , Zebrafish , Animals , Humans , Lysosomes/metabolism , Mannosephosphates/metabolism , Membrane Proteins/metabolism , Mucolipidoses/genetics , Mucolipidoses/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism , Zebrafish/metabolism
6.
Cell Rep ; 37(10): 110077, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879280

ABSTRACT

Viruses rearrange host membranes to support different entry steps. Polyomavirus simian virus 40 (SV40) reorganizes the endoplasmic reticulum (ER) membrane to generate focus structures that enable virus ER-to-cytosol escape, a decisive infection step. The molecular architecture of the ER exit site that might illuminate why it is ideally suited for membrane penetration is unknown. Here 3D focused ion beam scanning electron microscopy (FIB-SEM) reconstruction reveals that the ER focus structure consists of multi-tubular ER junctions where SV40 preferentially localizes, suggesting that tubular branch points are virus ER-to-cytosol penetration sites. Functional analysis demonstrates that lunapark-an ER membrane protein that typically stabilizes three-way ER junctions-relocates to the ER foci, where it supports focus formation, leading to SV40 ER escape and infection. Our results reveal how a virus repurposes the activity of an ER membrane protein to form a virus-induced ER substructure required for membrane escape and suggest that ER tubular junctions are vulnerable sites exploited by viruses for membrane penetration.


Subject(s)
Cytosol/virology , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Simian virus 40/metabolism , Virus Internalization , Animals , Cell Line , Chlorocebus aethiops , Cytosol/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Host-Pathogen Interactions , Intracellular Membranes/ultrastructure , Intracellular Membranes/virology , Male , Membrane Proteins/genetics , Simian virus 40/pathogenicity , Simian virus 40/ultrastructure
7.
Cardiovasc Res ; 117(1): 188-200, 2021 01 01.
Article in English | MEDLINE | ID: mdl-31995179

ABSTRACT

AIMS: Heart failure is a major complication in cancer treatment due to the cardiotoxic effects of anticancer drugs, especially from the anthracyclines such as doxorubicin (DXR). DXR enhances oxidative stress and stimulates matrix metalloproteinase-2 (MMP-2) in cardiomyocytes. We investigated whether MMP inhibitors protect against DXR cardiotoxicity given the role of MMP-2 in proteolyzing sarcomeric proteins in the heart and remodelling the extracellular matrix. METHODS AND RESULTS: Eight-week-old male C57BL/6J mice were treated with DXR weekly with or without MMP inhibitors doxycycline or ONO-4817 by daily oral gavage for 4 weeks. Echocardiography was used to determine cardiac function and left ventricular remodelling before and after treatment. MMP inhibitors ameliorated DXR-induced systolic and diastolic dysfunction by reducing the loss in left ventricular ejection fraction, fractional shortening, and E'/A'. MMP inhibitors attenuated adverse left ventricular remodelling, reduced cardiomyocyte dropout, and prevented myocardial fibrosis. DXR increased myocardial MMP-2 activity in part also by upregulating N-terminal truncated MMP-2. Immunogold transmission electron microscopy showed that DXR elevated MMP-2 levels within the sarcomere and mitochondria which were associated with myofilament lysis, mitochondrial degeneration, and T-tubule distention. DXR-induced myofilament lysis was associated with increased titin proteolysis in the heart which was prevented by ONO-4817. DXR also increased the level and activity of MMP-2 in human embryonic stem cell-derived cardiomyocytes, which was reduced by ONO-4817. CONCLUSIONS: MMP-2 activation is an early event in DXR cardiotoxicity and contributes to myofilament lysis by proteolyzing cardiac titin. Two orally available MMP inhibitors ameliorated DXR cardiotoxicity by attenuating intracellular and extracellular matrix remodelling, suggesting their use may be a potential prophylactic strategy to prevent heart injury during chemotherapy.


Subject(s)
Doxycycline/pharmacology , Extracellular Matrix/drug effects , Heart Diseases/prevention & control , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Phenyl Ethers/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiotoxicity , Cell Line , Disease Models, Animal , Doxorubicin , Extracellular Matrix/enzymology , Extracellular Matrix/pathology , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/physiopathology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/enzymology , Humans , Male , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Protein Kinases/metabolism , Proteolysis
8.
Basic Res Cardiol ; 114(6): 42, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506724

ABSTRACT

Junctophilin-2 is a structural membrane protein that tethers T-tubules to the sarcoplasmic reticulum to allow for coordinated calcium-induced calcium release in cardiomyocytes. Defective excitation-contraction coupling in myocardial ischemia-reperfusion (IR) injury is associated with junctophilin-2 proteolysis. However, it remains unclear whether preventing junctophilin-2 proteolysis improves the recovery of cardiac contractile dysfunction in IR injury. Matrix metalloproteinase-2 (MMP-2) is a zinc and calcium-dependent protease that is activated by oxidative stress in myocardial IR injury and cleaves both intracellular and extracellular substrates. To determine whether junctophilin-2 is targeted by MMP-2, isolated rat hearts were perfused in working mode aerobically or subjected to IR injury with the selective MMP inhibitor ARP-100. IR injury impaired the recovery of cardiac contractile function which was associated with increased degradation of junctophilin-2 and damaged cardiac dyads. In IR hearts, ARP-100 improved the recovery of cardiac contractile function, attenuated junctophilin-2 proteolysis, and prevented ultrastructural damage to the dyad. MMP-2 was co-localized with junctophilin-2 in aerobic and IR hearts by immunoprecipitation and immunohistochemistry. In situ zymography showed that MMP activity was localized to the Z-disc and sarcomere in aerobic hearts and accumulated at sites where the striated JPH-2 staining was disrupted in IR hearts. In vitro proteolysis assays determined that junctophilin-2 is susceptible to proteolysis by MMP-2 and in silico analysis predicted multiple MMP-2 cleavage sites between the membrane occupation and recognition nexus repeats and within the divergent region of junctophilin-2. Degradation of junctophilin-2 by MMP-2 is an early consequence of myocardial IR injury which may initiate a cascade of sequelae leading to impaired contractile function.


Subject(s)
Hydroxamic Acids/therapeutic use , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/therapeutic use , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Sulfones/therapeutic use , Animals , Computer Simulation , Drug Evaluation, Preclinical , Hydroxamic Acids/pharmacology , Male , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/ultrastructure , Rats, Sprague-Dawley , Sulfones/pharmacology
9.
Invest Ophthalmol Vis Sci ; 59(11): 4464-4470, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30193321

ABSTRACT

Purpose: To confirm whether choroideremia (CHM) is a systemic disease characterized by blood lipid abnormalities and crystals found in, or associated with, circulating peripheral blood cells of patients. Methods: Peripheral blood samples obtained from three subjects with confirmed mutations in the CHM gene and three age-matched normal controls were processed for transmission electron microscopy (TEM) and scanning electron microscopy (SEM). Fatty acids from plasma of nine male CHM subjects were analyzed and compared to reference values for a sample from a Canadian population. Results: Intracellular crystals were not observed in the cells from choroideremia-affected males. No crystals were found adherent to the external plasma membrane of red blood cells. Fatty acid profiles of patients were similar to reference values, with the exception of lower levels of nervonic acid. Conclusions: This investigation failed to observe crystals previously reported in peripheral circulating blood cells derived from CHM subjects, and showed no significant fatty acid abnormalities, not supporting the view of CHM as a systemic disease.


Subject(s)
Choroideremia/blood , Erythrocyte Inclusions/ultrastructure , Erythrocyte Membrane/ultrastructure , Phospholipids/blood , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Child , Choroideremia/genetics , Choroideremia/pathology , Crystallization , Humans , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Middle Aged , Mutation , Young Adult
10.
Sci Rep ; 8(1): 11685, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30076349

ABSTRACT

Tissue culture medium routinely contains fetal bovine serum (FBS). Here we show that culturing human hepatoma cells in their native, adult serum (human serum, HS) results in the restoration of key morphological and metabolic features of normal liver cells. When moved to HS, these cells show differential transcription of 22-32% of the genes, stop proliferating, and assume a hepatocyte-like morphology. Metabolic analysis shows that the Warburg-like metabolic profile, typical for FBS-cultured cells, is replaced by a diverse metabolic profile consistent with in vivo hepatocytes, including the formation of large lipid and glycogen stores, increased glycogenesis, increased beta-oxidation and ketogenesis, and decreased glycolysis. Finally, organ-specific functions are restored, including xenobiotics degradation and secretion of bile, VLDL and albumin. Thus, organ-specific functions are not necessarily lost in cell cultures, but might be merely suppressed in FBS. The effect of serum is often overseen in cell culture and we provide a detailed study in the changes that occur and provide insight in some of the serum components that may play a role in the establishment of the differentiated phenotype.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Serum/metabolism , Adult , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/ultrastructure , Cell Shape , Cytochrome P-450 Enzyme System/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Liver Neoplasms/genetics , Liver Neoplasms/ultrastructure , Metabolic Networks and Pathways , Principal Component Analysis , Tumor Cells, Cultured , Xenobiotics/metabolism
11.
Article in English | MEDLINE | ID: mdl-28104457

ABSTRACT

BACKGROUND: Pathophysiological responses, including cardiovascular complications, often alter with age. Cardioprotective effects of epoxyeicosatrienoic acids (EETs) toward acute myocardial ischemia-reperfusion injury have been well documented. However, biological relevance of EET-evoked cardioprotection in the ageing myocardium remains unknown. EETs are metabolized to less active metabolites by the enzyme soluble epoxide hydrolase (sEH). This study uses permanent occlusion of the left anterior descending artery (LAD) in young and aged sEH null and WT mice to compare cardiac and mitochondrial function following ischemic injury. METHODS: Age-matched 16 month old (aged) and 3 month old (young) sEH null and littermate wild-type (WT) mice were subjected to permanent occlusion of the left anterior descending coronary artery. Echocardiography was used to assess cardiac structure and function prior-to and 7days post-myocardial infarction with tetrazolium chloride staining to determine infarct size. Mitochondrial ultrastructure was obtained using electron microscopy. Caspase-3, 20S proteasome, aconitase and mitochondrial ETC enzymatic activities were ascertained using established protocols. Mitochondrial respiration was assessed using a Clark electrode in permeabilized cardiac fibers to obtain respiratory control ratios. RESULTS: Markers of cell injury, mitochondrial efficiency and overall cardiac function were preserved in aged sEH null mice, although less robustly than in their young counterparts. While aged animals of both genotypes demonstrated a similar overall age-related decline, sEH deletion consistently demonstrated protection from myocardial ischemic injury regardless of age. CONCLUSION: Our data demonstrates the protection originating from sEH deletion in aged mice was markedly reduced compared to young animals, signifying unavoidable detrimental consequences of biological ageing on cardiac function.


Subject(s)
Aging/genetics , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , Gene Deletion , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardium/metabolism , Animals , Epoxide Hydrolases/chemistry , Heart/physiopathology , Mice , Mitochondria/enzymology , Mitochondria/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Solubility
12.
Front Pharmacol ; 7: 133, 2016.
Article in English | MEDLINE | ID: mdl-27375480

ABSTRACT

AIMS: Myocardial ischemia can result in marked mitochondrial damage leading to cardiac dysfunction, as such identifying novel mechanisms to limit mitochondrial injury is important. This study investigated the hypothesis that inhibiting soluble epoxide hydrolase (sEH), responsible for converting epoxyeicosatrienoic acids to dihydroxyeicosatrienoic acids protects mitochondrial from injury caused by myocardial infarction. METHODS: sEH null and WT littermate mice were subjected to surgical occlusion of the left anterior descending (LAD) artery or sham operation. A parallel group of WT mice received an sEH inhibitor, trans-4-[4-(3-adamantan-1-y1-ureido)-cyclohexyloxy]-benzoic acid (tAUCB; 10 mg/L) or vehicle in the drinking water 4 days prior and 7 days post-MI. Cardiac function was assessed by echocardiography prior- and 7-days post-surgery. Heart tissues were dissected into infarct, peri-, and non-infarct regions to assess ultrastructure by electron microscopy. Complexes I, II, IV, citrate synthase, PI3K activities, and mitochondrial respiration were assessed in non-infarct regions. Isolated working hearts were used to measure the rates of glucose and palmitate oxidation. RESULTS: Echocardiography revealed that tAUCB treatment or sEH deficiency significantly improved systolic and diastolic function post-MI compared to controls. Reduced infarct expansion and less adverse cardiac remodeling were observed in tAUCB-treated and sEH null groups. EM data demonstrated mitochondrial ultrastructure damage occurred in infarct and peri-infarct regions but not in non-infarct regions. Inhibition of sEH resulted in significant improvements in mitochondrial respiration, ATP content, mitochondrial enzymatic activities and restored insulin sensitivity and PI3K activity. CONCLUSION: Inhibition or genetic deletion of sEH protects against long-term ischemia by preserving cardiac function and maintaining mitochondrial efficiency.

13.
Invest Ophthalmol Vis Sci ; 56(12): 7109-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26529045

ABSTRACT

PURPOSE: We investigated the interplay between photoreceptors expressing mutant ELOVL4 (responsible for Stargardt-like disease, STGD3) and RPE in the initial stages of retinal degeneration. METHODS: Using electron microscopy and electroretinogram (ERG), we assessed RPE and photoreceptor ultrastructure and function in transgenic ELOVL4 (TG1-2 line; TG) and wild-type (WT) littermates. Experiments were done at P30, 1 month before photoreceptor loss in TG and at P90, a time point with approximately 30% rod loss. To further elucidate the mechanism underlying our ultrastructural and functional results, we undertook Western blotting and immunohistochemistry of key proteins involved in phagocytosis of outer segments by RPE cells. RESULTS: Firstly, we showed that in TG mouse photoreceptors, endogenous ELOVL4 protein is not mislocalized in the presence of the mutated ELOVL4 protein. Secondly, we found evidence of RPE toxicity at P30, preceding any photoreceptor loss. Pathology in RPE cells was exacerbated at P90. Furthermore, higher proportions of phagosomes remained at the apical side of RPE cells. Subretinal lysosomal deposits were immunopositive for phagocytic proteins. Ultrastructural analysis of photoreceptor (rod) outer segments showed disrupted surface morphology consisting of disc spacing irregularities. Finally, rods and RPE exhibited signs of dysfunction as measured by the ERG a-wave leading edge (P30) and c-wave (P90), respectively. CONCLUSIONS: The presence of human mutant ELOVL4 in transgenic mouse photoreceptors leads to early outer segment disc pathology and RPE cytotoxicity. Defective processing of these abnormal discs by RPE cells ultimately may be responsible for outer segment truncation, photoreceptor death, and vision loss.


Subject(s)
Eye Proteins/genetics , Macular Degeneration/congenital , Mutation , RNA/genetics , Retinal Pigment Epithelium/ultrastructure , Rod Cell Outer Segment/ultrastructure , Animals , Blotting, Western , Cells, Cultured , DNA Mutational Analysis , Disease Models, Animal , Electroretinography , Eye Proteins/metabolism , Humans , Immunohistochemistry , Macular Degeneration/diagnosis , Macular Degeneration/metabolism , Macular Degeneration/physiopathology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiopathology , Rod Cell Outer Segment/metabolism
14.
PLoS One ; 10(6): e0129176, 2015.
Article in English | MEDLINE | ID: mdl-26076379

ABSTRACT

Although mammals are thought to lose their capacity to regenerate heart muscle shortly after birth, embryonic and neonatal cardiomyocytes in mammals are hyperplastic. During proliferation these cells need to selectively disassemble their myofibrils for successful cytokinesis. The mechanism of sarcomere disassembly is, however, not understood. To study this, we performed a series of immunofluorescence studies of multiple sarcomeric proteins in proliferating neonatal rat ventricular myocytes and correlated these observations with biochemical changes at different cell cycle stages. During myocyte mitosis, α-actinin and titin were disassembled as early as prometaphase. α-actinin (representing the sarcomeric Z-disk) disassembly precedes that of titin (M-line), suggesting that titin disassembly occurs secondary to the collapse of the Z-disk. Sarcomere disassembly was concurrent with the dissolution of the nuclear envelope. Inhibitors of several intracellular proteases could not block the disassembly of α-actinin or titin. There was a dramatic increase in both cytosolic (soluble) and sarcomeric α-actinin during mitosis, and cytosolic α-actinin exhibited decreased phosphorylation compared to sarcomeric α-actinin. Inhibition of cyclin-dependent kinase 1 (CDK1) induced the quick reassembly of the sarcomere. Sarcomere dis- and re-assembly in cardiomyocyte mitosis is CDK1-dependent and features dynamic differential post-translational modifications of sarcomeric and cytosolic α-actinin.


Subject(s)
Actinin/metabolism , Mitosis , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , Actinin/physiology , Animals , Cell Fractionation , Connectin/metabolism , Connectin/physiology , Cytosol/metabolism , Cytosol/ultrastructure , Flow Cytometry , Fluorescent Antibody Technique , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Phosphorylation , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sarcomeres/drug effects , Sarcomeres/ultrastructure
15.
Am J Physiol Heart Circ Physiol ; 306(5): H764-70, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24375642

ABSTRACT

Matrix metalloproteinase-2 (MMP-2) has been extensively studied in the context of extracellular matrix remodeling but is also localized within cells and can be activated by prooxidants to proteolyze specific intercellular targets. Although there are reports of MMP-2 in mitochondria, a critical source of cellular oxidative stress, these studies did not take into account the presence within their preparations of the mitochondria-associated membrane (MAM), a subdomain of the endoplasmic reticulum (ER). We hypothesized that MMP-2 is situated in the MAM and therefore investigated its subcellular distribution between mitochondria and the MAM. Immunogold electron microscopy revealed MMP-2 localized in mitochondria of heart sections from mice. In contrast, immunofluorescence analysis of an MMP-2:HaloTag fusion protein expressed in HL-1 cardiomyocytes showed an ER-like distribution, with greater colocalization with an ER marker (protein disulfide isomerase) relative to the mitochondrial marker, MitoTracker red. Although MMP-2 protein and enzymatic activity were present in crude mitochondrial fractions, once these were separated into purified mitochondria and MAM, MMP-2 was principally associated with the latter. Thus, although mitochondria may contain minimal levels of MMP-2, the majority of MMP-2 previously identified as "mitochondrial" is in fact associated with the MAM. We also found that calreticulin, an ER- and MAM-resident Ca(2+) handling protein and chaperone, could be proteolyzed by MMP-2 in vitro. MAM-localized MMP-2 could therefore potentially impact mitochondrial function by affecting ER-mitochondrial Ca(2+) signaling via its proteolysis of calreticulin.


Subject(s)
Endoplasmic Reticulum/enzymology , Intracellular Membranes/enzymology , Matrix Metalloproteinase 2/metabolism , Myocytes, Cardiac/enzymology , Animals , Calcium Signaling , Calreticulin/metabolism , Cell Line , Endoplasmic Reticulum/ultrastructure , Intracellular Membranes/ultrastructure , Male , Matrix Metalloproteinase 2/deficiency , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondrial Membranes/enzymology , Myocytes, Cardiac/ultrastructure , Proteolysis , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Transfection
16.
J Cardiovasc Pharmacol ; 61(3): 258-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403888

ABSTRACT

BACKGROUND: Caveolins (Cav) are structural proteins that insert into the plasma membrane to form caveolae that can bind molecules important in cardiac signal transduction and function. Cytochrome P450 epoxygenases can metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which have known cardioprotective effects. Subsequent metabolism of EETs by soluble epoxide hydrolase reduces the protective effect. AIMS: (1) To assess the effect of ischemia-reperfusion injury on expression and subcellular localization of caveolins. (2) To study the effect of EETs on caveolins. METHODS: Hearts from soluble epoxide hydrolase null (KO) and littermate control (WT) mice were perfused in Langendorff mode and subjected to 20 minutes ischemia followed by 40 minutes reperfusion. Immunohistochemistry, immunoblot, and electron microscopy were performed to study localization of caveolins and changes in ultrastructure. RESULTS: In WT heart, Cav-1 and Cav-3 were present in cardiomyocyte and capillary endothelial cell at baseline. After ischemia, Cav-1 but not Cav-3, disappeared from cardiomyocyte; moreover, caveolae were absent and mitochondrial cristae were damaged. Improved postischemic functional recovery observed in KO or WT hearts treated with 11,12-EET corresponded to higher Cav-1 expression and maintained caveolae structure. In addition, KO mice preserved the Cav-1 signaling after ischemia that lost in WT mice. CONCLUSIONS: Taken together, our data suggest that ischemia-reperfusion injury causes loss of Cav-1 and caveolins, and EETs-mediated cardioprotection involves preservation of Cav-1.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Caveolin 3/metabolism , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Biological Transport , Blotting, Western , Caveolae/ultrastructure , Epoxide Hydrolases/genetics , Heart/physiopathology , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/ultrastructure , Perfusion , Signal Transduction
17.
Crit Care Med ; 40(4): 1237-44, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22425819

ABSTRACT

OBJECTIVES: Asphyxiated neonates often have myocardial depression, which is a significant cause of morbidity and mortality. Cardioprotective effects of cyclosporine have been observed in adult patients and animals with myocardial infarction. However, the cardioprotective effect of cyclosporine in neonates has not yet been studied. We hypothesize that cyclosporine will improve cardiac function and reduce myocardial injury in asphyxiated newborn piglets. DESIGN: Thirty-six piglets (1-4 days old, weighing 1.4-2.5 kg) were acutely instrumented for continuous monitoring of cardiac output and systemic arterial pressure. After stabilization, normocapnic alveolar hypoxia (10% to 15% oxygen) was instituted for 2 hrs followed by reoxygenation with 100% oxygen for 0.5 hrs and then 21% for 3.5 hrs. A nonasphyxiated, sham-operated group was included (n = 4) to control for effects of the surgical model. Plasma troponin and myocardial lactate concentrations were determined as well as morphologic examinations. SETTING: Neonatal asphyxia and reoxygenation. SUBJECTS: Newborn (1-4 days old) piglets. INTERVENTIONS: Piglets were block-randomized to receive intravenous boluses of cyclosporine A (2.5, 10, or 25 mg/kg) or normal saline (control) at 5 mins of reoxygenation (n = 8/group). MEASUREMENTS AND MAIN RESULTS: Cardiac index, heart rate, systemic oxygenation, plasma troponin, and left ventricular lactate were measured. Hypoxic piglets had cardiogenic shock (cardiac output 40% to 48% of baseline), hypotension (mean arterial pressure 27-31 mm Hg), and acidosis (pH 7.04). Cyclosporine treatment caused bell-shaped improvements in cardiac output, stroke volume, and systemic oxygen delivery (p < .05 vs. controls). Plasma troponin and left ventricle lactate were higher in controls than that of 2.5 and 10 mg/kg cyclosporine-treated groups (p < .05). Although histologic features of myocardial injury were not different among groups, severe damage was observed in mitochondria of control piglets but attenuated in that of cyclosporine (10 mg/kg) treatment. CONCLUSIONS: Postresuscitation administration of cyclosporine causes preservation of cardiac function and attenuates myocardial injury in newborn piglets after asphyxia-reoxygenation.


Subject(s)
Asphyxia/drug therapy , Cyclosporine/therapeutic use , Heart/drug effects , Hemodynamics/drug effects , Immunosuppressive Agents/therapeutic use , Animals , Animals, Newborn , Asphyxia/physiopathology , Blood Gas Analysis , Cardiopulmonary Resuscitation/methods , Cyclosporine/administration & dosage , Dose-Response Relationship, Drug , Heart/physiopathology , Hemodynamics/physiology , Immunosuppressive Agents/administration & dosage , Lactates/blood , Microscopy, Electron, Transmission , Myocardium/ultrastructure , Swine , Troponin I/blood
18.
Intensive Care Med ; 38(3): 482-90, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22143394

ABSTRACT

PURPOSE: Asphyxia-related intestinal injury in neonates may present similar to necrotizing enterocolitis (NEC) and is partially associated with hypoxia-reoxygenation injury. Cyclosporine has been shown to reduce myocardial cell death following ischemia-reperfusion. We hypothesize that cyclosporine treatment may attenuate NEC-like intestinal injury in asphyxiated newborn piglets during reoxygenation. METHODS: Twenty piglets (1-4 days old) were acutely anesthetized and instrumented for continuous monitoring of systemic hemodynamics and superior mesenteric arterial (SMA) flow. After stabilization, normocapnic alveolar hypoxia (10-15% oxygen) was instituted for 2 h followed by reoxygenation with 100% oxygen for 0.5 h, then 21% for 3.5 h. The piglets were blindly block-randomized to receive cyclosporine (10 mg/kg) or placebo (normal saline) boluses at 5 min of reoxygenation (n = 8/group). A sham-operated group was included (n = 4) and received no hypoxia-reoxygenation. Intestinal samples were collected for tissue lactate and histological assessment (Park's criteria). RESULTS: At 2 h of hypoxia, piglets had cardiogenic shock (cardiac output 45% of baseline), hypotension (mean arterial pressure 30 mmHg), acidosis (pH 7.04), and decreased superior mesenteric perfusion (all P < 0.05 vs. sham-operated group, ANOVA). Cyclosporine treatment increased SMA flow (114 ± 6 vs. 78 ± 19% of baseline of controls, respectively) with improved SMA oxygen delivery and intestinal tissue lactate (all P < 0.05). Some control piglets had NEC-like injuries including pneumatosis intestinalis, which were attenuated in cyclosporine-treated piglets (P < 0.05 vs. controls). CONCLUSIONS: This is the first study to demonstrate that post-resuscitation administration of cyclosporine improves mesenteric perfusion and attenuates NEC-like intestinal injury in newborn piglets following asphyxia-reoxygenation.


Subject(s)
Asphyxia/drug therapy , Cyclosporine/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Hypoxia/drug therapy , Mesenteric Arteries/drug effects , Alberta , Analysis of Variance , Animals , Animals, Newborn , Cyclosporine/administration & dosage , Enterocolitis, Necrotizing/etiology , Hemodynamics/drug effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Intestines/drug effects , Intestines/injuries , Mesenteric Arteries/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Random Allocation , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology , Resuscitation/methods , Swine
19.
Arterioscler Thromb Vasc Biol ; 32(3): 662-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22199370

ABSTRACT

OBJECTIVE: Matrix metalloproteinase (MMP)-2 is activated in aorta during endotoxemia and plays a role in the hypocontractility to vasoconstrictors. Calponin-1 is a regulator of vascular smooth muscle tone with similarities to troponin, a cardiac myocyte protein that is cleaved by MMP-2 in myocardial oxidative stress injuries. We hypothesized that calponin-1 may be proteolyzed by MMP-2 in endotoxemia-induced vascular hypocontractility. METHODS AND RESULTS: Rats were given a nonlethal dose of bacterial lipopolysaccharide (LPS) or vehicle. Some rats were given the MMP inhibitors ONO-4817 or doxycycline. Six hours later, plasma nitrate+nitrite increased >15-fold in LPS-treated rats, an effect unchanged by doxycycline. Both ONO-4817 and doxycycline prevented LPS-induced aortic hypocontractility to phenylephrine. LPS activated MMP-2 in the aorta by S-glutathiolation. Calponin-1 levels decreased by 25% in endotoxemic aortae, which was prevented by doxycycline. Calponin-1 and MMP-2 coimmunoprecipitated and both exhibited uniform cytosolic staining in medial vascular smooth muscle cells. In vitro incubation of calponin-1 with MMP-2 led to calponin-1 degradation and appearance of its cleavage product. CONCLUSION: Calponin-1 is a target of MMP-2, which contributes to endotoxemia-induced vascular hypocontractility.


Subject(s)
Calcium-Binding Proteins/metabolism , Endotoxemia/enzymology , Endotoxemia/physiopathology , Matrix Metalloproteinase 2/metabolism , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Vasoconstriction , Animals , Aorta/enzymology , Aorta/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Endotoxemia/chemically induced , Glutathione/metabolism , Immunoprecipitation , Lipopolysaccharides , Male , Matrix Metalloproteinase Inhibitors , Muscle, Smooth, Vascular/drug effects , Phenyl Ethers/pharmacology , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Calponins
20.
Circulation ; 122(20): 2039-47, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21041693

ABSTRACT

BACKGROUND: Titin is the largest mammalian (≈3000 to 4000 kDa) and myofilament protein that acts as a molecular spring in the cardiac sarcomere and determines systolic and diastolic function. Loss of titin in ischemic hearts has been reported, but the mechanism of titin degradation is not well understood. Matrix metalloproteinase-2 (MMP-2) is localized to the cardiac sarcomere and, on activation in ischemia/reperfusion injury, proteolyzes specific myofilament proteins. Here we determine whether titin is an intracellular substrate for MMP-2 and if its degradation during ischemia/reperfusion contributes to cardiac contractile dysfunction. METHODS AND RESULTS: Immunohistochemistry and confocal microscopy in rat and human hearts showed discrete colocalization between MMP-2 and titin in the Z-disk region of titin and that MMP-2 is localized mainly to titin near the Z disk of the cardiac sarcomere. Both purified titin and titin in skinned cardiomyocytes were proteolyzed when incubated with MMP-2 in a concentration-dependent manner, and this was prevented by MMP inhibitors. Isolated rat hearts subjected to ischemia/reperfusion injury showed cleavage of titin in ventricular extracts by gel electrophoresis, which was confirmed by reduced titin immunostaining in tissue sections. Inhibition of MMP activity with ONO-4817 prevented ischemia/reperfusion-induced titin degradation and improved the recovery of myocardial contractile function. Titin degradation was also reduced in hearts from MMP-2 knockout mice subjected to ischemia/reperfusion in vivo compared with wild-type controls. CONCLUSION: MMP-2 localizes to titin at the Z-disk region of the cardiac sarcomere and contributes to titin degradation in myocardial ischemia/reperfusion injury.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Muscle Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Protein Kinases/metabolism , Animals , Connectin , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenyl Ethers/pharmacology , Protein Kinases/genetics , Rats , Rats, Sprague-Dawley , Sarcolemma/genetics , Sarcolemma/metabolism , Sarcolemma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...