106 results on '"Crow MT"'
Search Results
2. Simvastatin Protection Against Monocrotaline-Induced Pulmonary Hypertension Is Dose-Dependent.
- Author
-
Boueiz, A, primary, Mozammel, S, additional, Sloan, D, additional, Kesari, P, additional, Zaiman, A, additional, Champion, HC, additional, El-Haddad, H, additional, Crow, MT, additional, Angelini, D, additional, Hassoun, PM, additional, and Girgis, RE, additional
- Published
- 2009
- Full Text
- View/download PDF
3. Lung Injury Induces a Skeletal Muscle Atrophy Program That Is Qualitatively Different from That Induced by Starvation.
- Author
-
Files, DC, primary, DeGorordo, A, additional, Kesari, P, additional, Johnston, L, additional, Tsushima, K, additional, Aggarwal, N, additional, Sidhaye, VK, additional, D'Alessio, F, additional, King, LS, additional, and Crow, MT, additional
- Published
- 2009
- Full Text
- View/download PDF
4. A critical role for the protein apoptosis repressor with caspase recruitment domain in hypoxia-induced pulmonary hypertension.
- Author
-
Zaiman AL, Damico R, Thoms-Chesley A, Files DC, Kesari P, Johnston L, Swaim M, Mozammel S, Myers AC, Halushka M, El-Haddad H, Shimoda LA, Peng CF, Hassoun PM, Champion HC, Kitsis RN, Crow MT, Zaiman, Ari L, Damico, Rachel, and Thoms-Chesley, Alan
- Published
- 2011
- Full Text
- View/download PDF
5. Pulmonary epithelial neuropilin-1 deletion enhances development of cigarette smoke-induced emphysema.
- Author
-
Le A, Zielinski R, He C, Crow MT, Biswal S, Tuder RM, Becker PM, Le, Anne, Zielinski, Rachel, He, Chaoxia, Crow, Michael T, Biswal, Shyam, Tuder, Rubin M, and Becker, Patrice M
- Abstract
Rationale: Cigarette smoke (CS) exposure is an important risk factor for chronic obstructive pulmonary disease; however, not all smokers develop disease, suggesting that other factors influence disease development.Objectives: We sought to determine whether neuropilin-1 (Nrp1), an integral component of receptor complexes mediating alveolar septation and vascular development, was involved in maintenance of normal alveolar structure, and/or altered susceptibility to the effects of CS.Methods: Transgenic mice were generated to achieve inducible lung-specific deletion of epithelial Nrp1. We determined whether conditional Nrp1 deletion altered airspace size, then compared the effects of chronic CS or filtered air exposure on airspace size, inflammation, and the balance between cell death and proliferation in conditionally Nrp1-deficient adult mice and littermate controls. Finally, we evaluated the effects of Nrp1 silencing on cell death after acute exposure of A549 cells to cigarette smoke extract or short chain ceramides.Measurements and Main Results: Genetic deletion of epithelial Nrp1 in either postnatal or adult lungs resulted in a small increase in airspace size. More notably, both airspace enlargement and apoptosis of type I and type II alveolar epithelial cells were significantly enhanced following chronic CS exposure in conditionally Nrp1-deficient adult mice. Silencing of Nrp1 in A549 cells did not alter cell survival after vehicle treatment but significantly augmented apoptosis after exposure to cigarette smoke extract or ceramide.Conclusions: These data support a role for epithelial Nrp1 in the maintenance of normal alveolar structure and suggest that dysregulation of Nrp1 expression may promote epithelial cell death in response to CS exposure, thereby enhancing emphysema development. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
6. Myosin light-chain expression during avian muscle development
- Author
-
Crow, MT, Olson, PS, and Stockdale, FE
- Abstract
Monoclonal antibodies to adult chicken myosin light chains were generated and used to quantitate the types of myosin light-chain (MLC) isoforms expressed during development of the pectoralis major (PM), anterior latissimus dorsi (ALD), and medial adductor (MA) muscles of the chicken. These are muscles which, in the adult, are composed predominantly of fast, slow, and a mixture of fiber types, respectively. Three distinct phases of MLC expression characterized the development of the PM and MA muscles. The first identifiable pase occurred during the period of 5-7 d of incubation in ovo. Extracts of muscles from the pectoral region (which included the presumptive PM muscle) contained only fast MLC isoforms. This period of exclusive fast light-chain synthesis was followed by a phase (8- 12 d of incubation in ovo) in which coexpression of both fast and slow MLC isoforms was apparent in both PM and MA muscles. During the period, the composition of both fast and slow MLC isoforms in the PM and MA muscles was identical. Beginning at day 12 in ovo, the ALD was also subjected to immunochemical analyses. The proportion of fast and slow MLCs in this muscle at day 12 was similar to that present in the other muscles studied. The third development phase of MLC expression began at approximately 12 d of incubation in ovo and encompassed the transition in MLC composition to the isoform patterns incubation in ovo and encompassed the transition in MLC composition to the isoform patterns typical of adult muscle. During this period, the relative proportion of slow MLC rose in both the MA and ALD and fell in the PM. By day 16, the third fast light chain, LC(3f), was apparent in extracts of both the PM and MA. These results show that there is a developmental progression in the expression of MLC in the two avian muscles studied from day 5 in ovo; first, only fast MLCs are accumulated, then both fast and slow MLC isoforms are expressed. Only during the latter third of development in ovo is the final MLC isoform pattern characteristic of a particular muscle type expressed.
- Published
- 1983
- Full Text
- View/download PDF
7. Adenovirus-mediated gene transfer of TIMP-2 inhibits neointimal hyperplasia after balloon injury
- Author
-
Cheng, L., Mantile, G., Pauly, R., Monticone, Re, Bilato, C., Nater, C., Crow, Mt, William Stetler-Stevenson, and Capogrossi, Mc
8. Adenovirus mediated gene transfer of human tissue inhibitor of metalloproteinase-2 inhibits vascular smooth muscle cell migration
- Author
-
Mantile, G., Crow, Mt, Bilato, C., Pauly, R., Cheng, L., William Stetler-Stevenson, and Capogrossi, Mc
9. Foxp3(+) regulatory T cells participate in repair of ischemic acute kidney injury.
- Author
-
Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Satpute SR, Crow MT, King LS, and Rabb H
- Abstract
T lymphocytes modulate early ischemia-reperfusion injury in the kidney; however, their role during repair is unknown. We studied the role of TCRbeta(+)CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs), known to blunt immune responses, in repair after ischemia-reperfusion injury to the kidney. Using a murine model of ischemic acute kidney injury we found that there was a significant trafficking of Tregs into the kidneys after 3 and 10 days. Post-ischemic kidneys had increased numbers of TCRbeta(+)CD4(+) and TCRbeta(+)CD8(+) T cells with enhanced pro-inflammatory cytokine production. Treg depletion starting 1 day after ischemic injury using anti-CD25 antibodies increased renal tubular damage, reduced tubular proliferation at both time points, enhanced infiltrating T lymphocyte cytokine production at 3 days and TNF-alpha generation by TCRbeta(+)CD4(+) T cells at 10 days. In separate mice, infusion of CD4(+)CD25(+) Tregs 1 day after initial injury reduced INF-gamma production by TCRbeta(+)CD4(+) T cells at 3 days, improved repair and reduced cytokine generation at 10 days. Treg manipulation had minimal effect on neutrophil and macrophage infiltration; Treg depletion worsened mortality and serum creatinine, while Treg infusion had a late beneficial effect on serum creatinine in bilateral ischemia. Our study demonstrates that Tregs infiltrate ischemic-reperfused kidneys during the healing process promoting repair, likely through modulation of pro-inflammatory cytokine production of other T cell subsets. Treg targeting could be a novel therapeutic approach to enhance recovery from ischemic acute kidney injury. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
10. From Bench to Bedside: A Team's Approach to Multidisciplinary Strategies to Combat Therapeutic Resistance in Head and Neck Squamous Cell Carcinoma.
- Author
-
Crossman BE, Harmon RL, Kostecki KL, McDaniel NK, Iida M, Corday LW, Glitchev CE, Crow MT, Harris MA, Lin CY, Adams JM, Longhurst CA, Nickel KP, Ong IM, Alexandridis RA, Yu M, Yang DT, Hu R, Morris ZS, Hartig GK, Glazer TA, Ramisetty S, Kulkarni P, Salgia R, Kimple RJ, Bruce JY, Harari PM, and Wheeler DL
- Abstract
Head and neck squamous cell carcinoma (HNSCC) is diagnosed in more than 71,000 patients each year in the United States, with nearly 16,000 associated deaths. One significant hurdle in the treatment of HNSCC is acquired and intrinsic resistance to existing therapeutic agents. Over the past several decades, the University of Wisconsin has formed a multidisciplinary team to move basic scientific discovery along the translational spectrum to impact the lives of HNSCC patients. In this review, we outline key discoveries made throughout the years at the University of Wisconsin to deepen our understanding of therapeutic resistance in HNSCC and how a strong, interdisciplinary team can make significant advances toward improving the lives of these patients by combatting resistance to established therapeutic modalities. We are profoundly grateful to the many scientific teams worldwide whose groundbreaking discoveries, alongside evolving clinical paradigms in head and neck oncology, have been instrumental in making our work possible.
- Published
- 2024
- Full Text
- View/download PDF
11. MerTK Drives Proliferation and Metastatic Potential in Triple-Negative Breast Cancer.
- Author
-
Iida M, Crossman BE, Kostecki KL, Glitchev CE, Kranjac CA, Crow MT, Adams JM, Liu P, Ong I, Yang DT, Kang I, Salgia R, and Wheeler DL
- Subjects
- Humans, Animals, Female, Mice, Cell Line, Tumor, Cell Movement genetics, Gene Expression Regulation, Neoplastic, Endoglin metabolism, Endoglin genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Lung Neoplasms genetics, Lung Neoplasms secondary, Neoplasm Metastasis, Signal Transduction, Apoptosis genetics, c-Mer Tyrosine Kinase metabolism, c-Mer Tyrosine Kinase genetics, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms genetics, Cell Proliferation
- Abstract
Triple-negative breast cancer (TNBC) is characterized by the absence of the estrogen receptor, progesterone receptor, and receptor tyrosine kinase HER2 expression. Due to the limited number of FDA-approved targeted therapies for TNBC, there is an ongoing need to understand the molecular underpinnings of TNBC for the development of novel combinatorial treatment strategies. This study evaluated the role of the MerTK receptor tyrosine kinase on proliferation and invasion/metastatic potential in TNBC. Immunohistochemical analysis demonstrated MerTK expression in 58% of patient-derived TNBC xenografts. The stable overexpression of MerTK in human TNBC cell lines induced an increase in proliferation rates, robust in vivo tumor growth, heightened migration/invasion potential, and enhanced lung metastases. NanoString nCounter analysis of MerTK-overexpressing SUM102 cells (SUM102-MerTK) revealed upregulation of several signaling pathways, which ultimately drive cell cycle progression, reduce apoptosis, and enhance cell survival. Proteomic profiling indicated increased endoglin (ENG) production in SUM102-MerTK clones, suggesting that MerTK creates a conducive environment for increased proliferative and metastatic activity via elevated ENG expression. To determine ENG's role in increasing proliferation and/or metastatic potential, we knocked out ENG in a SUM102-MerTK clone with CRISPR technology. Although this ENG knockout clone exhibited similar in vivo growth to the parental SUM102-MerTK clone, lung metastasis numbers were significantly decreased ~4-fold, indicating that MerTK enhances invasion and metastasis through ENG. Our data suggest that MerTK regulates a unique proliferative signature in TNBC, promoting robust tumor growth and increased metastatic potential through ENG upregulation. Targeting MerTK and ENG simultaneously may provide a novel therapeutic approach for TNBC patients.
- Published
- 2024
- Full Text
- View/download PDF
12. Apoptosis Repressor With Caspase Recruitment Domain Ameliorates Amyloid-Induced β-Cell Apoptosis and JNK Pathway Activation.
- Author
-
Templin AT, Samarasekera T, Meier DT, Hogan MF, Mellati M, Crow MT, Kitsis RN, Zraika S, Hull RL, and Kahn SE
- Subjects
- Animals, Apoptosis drug effects, Apoptosis genetics, Apoptosis Regulatory Proteins genetics, Blotting, Western, Cells, Cultured, Female, Immunoprecipitation, Insulin-Secreting Cells drug effects, JNK Mitogen-Activated Protein Kinases genetics, Male, Mice, Mice, Transgenic, Muscle Proteins genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Amyloid pharmacology, Apoptosis Regulatory Proteins chemistry, Apoptosis Regulatory Proteins metabolism, Insulin-Secreting Cells metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Muscle Proteins chemistry, Muscle Proteins metabolism
- Abstract
Islet amyloid is present in more than 90% of individuals with type 2 diabetes, where it contributes to β-cell apoptosis and insufficient insulin secretion. Apoptosis repressor with caspase recruitment domain (ARC) binds and inactivates components of the intrinsic and extrinsic apoptosis pathways and was recently found to be expressed in islet β-cells. Using a human islet amyloid polypeptide transgenic mouse model of islet amyloidosis, we show ARC knockdown increases amyloid-induced β-cell apoptosis and loss, while ARC overexpression decreases amyloid-induced apoptosis, thus preserving β-cells. These effects occurred in the absence of changes in islet amyloid deposition, indicating ARC acts downstream of amyloid formation. Because islet amyloid increases c-Jun N-terminal kinase (JNK) pathway activation, we investigated whether ARC affects JNK signaling in amyloid-forming islets. We found ARC knockdown enhances JNK pathway activation, whereas ARC overexpression reduces JNK, c-Jun phosphorylation, and c-Jun target gene expression ( Jun and Tnf ). Immunoprecipitation of ARC from mouse islet lysates showed ARC binds JNK, suggesting interaction between JNK and ARC decreases amyloid-induced JNK phosphorylation and downstream signaling. These data indicate that ARC overexpression diminishes amyloid-induced JNK pathway activation and apoptosis in the β-cell, a strategy that may reduce β-cell loss in type 2 diabetes., (© 2017 by the American Diabetes Association.)
- Published
- 2017
- Full Text
- View/download PDF
13. The aquaporin 1 C-terminal tail is required for migration and growth of pulmonary arterial myocytes.
- Author
-
Lai N, Lade J, Leggett K, Yun X, Baksh S, Chau E, Crow MT, Sidhaye V, Wang J, and Shimoda LA
- Subjects
- Animals, Aquaporin 1 genetics, Calcium metabolism, Cell Growth Processes genetics, Cell Growth Processes physiology, Cell Hypoxia physiology, Cell Movement genetics, Fluoresceins chemistry, Gene Knockdown Techniques, Male, Muscle Cells metabolism, Mutation, Pulmonary Artery metabolism, Rats, Rats, Wistar, Water metabolism, Aquaporin 1 metabolism, Cell Movement physiology, Muscle Cells physiology, Pulmonary Artery physiology
- Abstract
Pulmonary arterial smooth muscle cell (PASMC) proliferation and migration are important contributors to the vascular remodeling that occurs during development of pulmonary hypertension. We previously demonstrated that aquaporin (AQP)1, a member of the water channel family of proteins, was expressed in PASMCs and was necessary for hypoxia-induced migration; however, the mechanism by which AQP1 controls this response is unclear. The C-terminal tail of AQP1 contains putative calcium (EF-hand) and protein binding sites. Thus, we wanted to explore whether the C-terminal tail or the EF-hand motif of AQP1 was required for migration and proliferation. Rat PASMCs were isolated from distal pulmonary arteries, and proliferation and migration were measured using BrdU incorporation and transwell filters, respectively. To deplete AQP1, PASMCs were transfected with AQP1 small interference RNA (siRNA) or nontargeting siRNA. Knockdown of AQP1 reduced basal proliferation and hypoxia-induced migration and proliferation in PASMCs. In subsequent experiments, wild-type AQP1, AQP1 lacking the entire cytoplasmic C-terminal tail, or AQP1 with a mutation in the EF-hand motif were expressed in PASMCs using adenoviral constructs. For all AQP1 constructs, infection increased AQP1 protein levels, water permeability, and the change in cell volume induced by hypotonic challenge. Infection with wild-type and EF-hand mutated AQP1, but not C-terminal-deleted AQP1, increased PASMC migration and proliferation. Our results suggest that AQP1 controls proliferation and migration in PASMCs and that the mechanism requires the C-terminal tail of the protein but is independent of water transport or the EF-hand motif.
- Published
- 2014
- Full Text
- View/download PDF
14. Mitogen-activated protein kinase-activated protein kinase 2 mediates apoptosis during lung vascular permeability by regulating movement of cleaved caspase 3.
- Author
-
Damarla M, Parniani AR, Johnston L, Maredia H, Serebreni L, Hamdan O, Sidhaye VK, Shimoda LA, Myers AC, Crow MT, Schmidt EP, Machamer CE, Gaestel M, Rane MJ, Kolb TM, Kim BS, Damico RL, and Hassoun PM
- Subjects
- Active Transport, Cell Nucleus, Animals, Capillary Permeability, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Poly(ADP-ribose) Polymerases, Apoptosis physiology, Caspase 3 metabolism, Intracellular Signaling Peptides and Proteins metabolism, Lung blood supply, Protein Serine-Threonine Kinases metabolism
- Abstract
Apoptosis is a key pathologic feature in acute lung injury. Animal studies have demonstrated that pathways regulating apoptosis are necessary in the development of acute lung injury, and that activation of p38 mitogen-activated protein kinase (MAPK) is linked to the initiation of the apoptotic cascade. In this study, we assessed the role of the MAPK-activated protein kinase (MK) 2, one of p38 MAPK's immediate downstream effectors, in the development of apoptosis in an animal model of LPS-induced pulmonary vascular permeability. Our results indicate that wild-type (WT) mice exposed to LPS demonstrate increased apoptosis, as evidenced by cleavage of caspase 3 and poly (ADP-ribose) polymerase 1 and increased deoxynucleotidyl transferase-mediated dUDP nick-end labeling staining, which is accompanied by increases in markers of vascular permeability. In contrast, MK2(-/-) mice are protected from pulmonary vascular permeability and apoptosis in response to LPS. Although there was no difference in activation of caspase 3 in MK2(-/-) compared with WT mice, interestingly, cleaved caspase 3 translocated to the nucleus in WT mice while it remained in the cytosol of MK2(-/-) mice in response to LPS. In separate experiments, LPS-induced apoptosis in human lung microvascular endothelial cells was also associated with nuclear translocation of cleaved caspase 3 and apoptosis, which were both prevented by MK2 silencing. In conclusion, our data suggest that MK2 plays a critical role in the development of apoptosis and pulmonary vascular permeability, and its effects on apoptosis are in part related to its ability to regulate nuclear translocation of cleaved caspase 3.
- Published
- 2014
- Full Text
- View/download PDF
15. The apoptosis inhibitor ARC alleviates the ER stress response to promote β-cell survival.
- Author
-
McKimpson WM, Weinberger J, Czerski L, Zheng M, Crow MT, Pessin JE, Chua SC Jr, and Kitsis RN
- Subjects
- Animals, Apoptosis, Calcium metabolism, Cell Line, Tumor, Cell Survival, Humans, Mice, Transcription Factor CHOP physiology, Apoptosis Regulatory Proteins physiology, Endoplasmic Reticulum Stress, Insulin-Secreting Cells physiology, Muscle Proteins physiology
- Abstract
Type 2 diabetes involves insulin resistance and β-cell failure leading to inadequate insulin secretion. An important component of β-cell failure is cell loss by apoptosis. Apoptosis repressor with caspase recruitment domain (ARC) is an inhibitor of apoptosis that is expressed in cardiac and skeletal myocytes and neurons. ARC possesses the unusual property of antagonizing both the extrinsic (death receptor) and intrinsic (mitochondria/endoplasmic reticulum [ER]) cell death pathways. Here we report that ARC protein is abundant in cells of the endocrine pancreas, including >99.5% of mouse and 73% of human β-cells. Using genetic gain- and loss-of-function approaches, our data demonstrate that ARC inhibits β-cell apoptosis elicited by multiple inducers of cell death, including ER stressors tunicamycin, thapsigargin, and physiological concentrations of palmitate. Unexpectedly, ARC diminishes the ER stress response, acting distal to protein kinase RNA-like ER kinase (PERK) and inositol-requiring protein 1α, to suppress C/EBP homologous protein (CHOP) induction. Depletion of ARC in isolated islets augments palmitate-induced apoptosis, which is dramatically rescued by deletion of CHOP. These data demonstrate that ARC is a previously unrecognized inhibitor of apoptosis in β-cells and that its protective effects are mediated through suppression of the ER stress response pathway.
- Published
- 2013
- Full Text
- View/download PDF
16. Resolution of experimental lung injury by monocyte-derived inducible nitric oxide synthase.
- Author
-
D'Alessio FR, Tsushima K, Aggarwal NR, Mock JR, Eto Y, Garibaldi BT, Files DC, Avalos CR, Rodriguez JV, Waickman AT, Reddy SP, Pearse DB, Sidhaye VK, Hassoun PM, Crow MT, and King LS
- Subjects
- Acute Lung Injury immunology, Animals, B7-2 Antigen biosynthesis, Cell Line, Cell Line, Transformed, Disease Models, Animal, Inflammation Mediators metabolism, Inflammation Mediators therapeutic use, Macrophages, Peritoneal enzymology, Macrophages, Peritoneal immunology, Male, Mice, Mice, Congenic, Mice, Inbred C57BL, Mice, Knockout, Monocytes pathology, Nitric Oxide Synthase Type II deficiency, Acute Lung Injury enzymology, Acute Lung Injury therapy, Monocytes enzymology, Monocytes immunology, Nitric Oxide Synthase Type II therapeutic use
- Abstract
Although early events in the pathogenesis of acute lung injury (ALI) have been defined, little is known about the mechanisms mediating resolution. To search for determinants of resolution, we exposed wild type (WT) mice to intratracheal LPS and assessed the response at intervals to day 10, when injury had resolved. Inducible NO synthase (iNOS) was significantly upregulated in the lung at day 4 after LPS. When iNOS-/- mice were exposed to intratracheal LPS, early lung injury was attenuated; however, recovery was markedly impaired compared with WT mice. iNOS-/- mice had increased mortality and sustained increases in markers of lung injury. Adoptive transfer of WT (iNOS+/+) bone marrow-derived monocytes or direct adenoviral gene delivery of iNOS into injured iNOS-/- mice restored resolution of ALI. Irradiated bone marrow chimeras confirmed the protective effects of myeloid-derived iNOS but not of epithelial iNOS. Alveolar macrophages exhibited sustained expression of cosignaling molecule CD86 in iNOS-/- mice compared with WT mice. Ab-mediated blockade of CD86 in iNOS-/- mice improved survival and enhanced resolution of lung inflammation. Our findings show that monocyte-derived iNOS plays a pivotal role in mediating resolution of ALI by modulating lung immune responses, thus facilitating clearance of alveolar inflammation and promoting lung repair.
- Published
- 2012
- Full Text
- View/download PDF
17. Bax regulates primary necrosis through mitochondrial dynamics.
- Author
-
Whelan RS, Konstantinidis K, Wei AC, Chen Y, Reyna DE, Jha S, Yang Y, Calvert JW, Lindsten T, Thompson CB, Crow MT, Gavathiotis E, Dorn GW 2nd, O'Rourke B, and Kitsis RN
- Subjects
- Adenosine Triphosphate biosynthesis, Animals, Mice, Mice, Knockout, Myocardial Infarction genetics, Myocardial Infarction physiopathology, Necrosis, bcl-2-Associated X Protein genetics, Mitochondria physiology, bcl-2-Associated X Protein physiology
- Abstract
The defining event in apoptosis is mitochondrial outer membrane permeabilization (MOMP), allowing apoptogen release. In contrast, the triggering event in primary necrosis is early opening of the inner membrane mitochondrial permeability transition pore (mPTP), precipitating mitochondrial dysfunction and cessation of ATP synthesis. Bcl-2 proteins Bax and Bak are the principal activators of MOMP and apoptosis. Unexpectedly, we find that deletion of Bax and Bak dramatically reduces necrotic injury during myocardial infarction in vivo. Triple knockout mice lacking Bax/Bak and cyclophilin D, a key regulator of necrosis, fail to show further reduction in infarct size over those deficient in Bax/Bak. Absence of Bax/Bak renders cells resistant to mPTP opening and necrosis, effects confirmed in isolated mitochondria. Reconstitution of these cells or mitochondria with wild-type Bax, or an oligomerization-deficient mutant that cannot support MOMP and apoptosis, restores mPTP opening and necrosis, implicating distinct mechanisms for Bax-regulated necrosis and apoptosis. Both forms of Bax restore mitochondrial fusion in Bax/Bak-null cells, which otherwise exhibit fragmented mitochondria. Cells lacking mitofusin 2 (Mfn2), which exhibit similar fusion defects, are protected to the same extent as Bax/Bak-null cells. Conversely, restoration of fused mitochondria through inhibition of fission potentiates mPTP opening in the absence of Bax/Bak or Mfn2, indicating that the fused state itself is critical. These data demonstrate that Bax-driven fusion lowers the threshold for mPTP opening and necrosis. Thus, Bax and Bak play wider roles in cell death than previously appreciated and may be optimal therapeutic targets for diseases that involve both forms of cell death.
- Published
- 2012
- Full Text
- View/download PDF
18. A critical role for muscle ring finger-1 in acute lung injury-associated skeletal muscle wasting.
- Author
-
Files DC, D'Alessio FR, Johnston LF, Kesari P, Aggarwal NR, Garibaldi BT, Mock JR, Simmers JL, DeGorordo A, Murdoch J, Willis MS, Patterson C, Tankersley CG, Messi ML, Liu C, Delbono O, Furlow JD, Bodine SC, Cohn RD, King LS, and Crow MT
- Subjects
- Animals, Blotting, Western, Bronchoalveolar Lavage Fluid chemistry, Disease Models, Animal, Down-Regulation, Gene Expression Regulation, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Multivariate Analysis, Muscle Strength physiology, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Muscular Atrophy pathology, RING Finger Domains genetics, Random Allocation, Sensitivity and Specificity, Tripartite Motif Proteins, Acute Lung Injury genetics, Acute Lung Injury pathology, Muscle Proteins genetics, Muscular Atrophy genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Rationale: Acute lung injury (ALI) is a debilitating condition associated with severe skeletal muscle weakness that persists in humans long after lung injury has resolved. The molecular mechanisms underlying this condition are unknown., Objectives: To identify the muscle-specific molecular mechanisms responsible for muscle wasting in a mouse model of ALI., Methods: Changes in skeletal muscle weight, fiber size, in vivo contractile performance, and expression of mRNAs and proteins encoding muscle atrophy-associated genes for muscle ring finger-1 (MuRF1) and atrogin1 were measured. Genetic inactivation of MuRF1 or electroporation-mediated transduction of miRNA-based short hairpin RNAs targeting either MuRF1 or atrogin1 were used to identify their role in ALI-associated skeletal muscle wasting., Measurements and Main Results: Mice with ALI developed profound muscle atrophy and preferential loss of muscle contractile proteins associated with reduced muscle function in vivo. Although mRNA expression of the muscle-specific ubiquitin ligases, MuRF1 and atrogin1, was increased in ALI mice, only MuRF1 protein levels were up-regulated. Consistent with these changes, suppression of MuRF1 by genetic or biochemical approaches prevented muscle fiber atrophy, whereas suppression of atrogin1 expression was without effect. Despite resolution of lung injury and down-regulation of MuRF1 and atrogin1, force generation in ALI mice remained suppressed., Conclusions: These data show that MuRF1 is responsible for mediating muscle atrophy that occurs during the period of active lung injury in ALI mice and that, as in humans, skeletal muscle dysfunction persists despite resolution of lung injury.
- Published
- 2012
- Full Text
- View/download PDF
19. Knockdown of lung phosphodiesterase 2A attenuates alveolar inflammation and protein leak in a two-hit mouse model of acute lung injury.
- Author
-
Rentsendorj O, Damarla M, Aggarwal NR, Choi JY, Johnston L, D'Alessio FR, Crow MT, and Pearse DB
- Subjects
- Adenoviridae enzymology, Adenoviridae genetics, Animals, Bronchoalveolar Lavage Fluid cytology, Cyclic AMP metabolism, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 2 genetics, Cyclic Nucleotide Phosphodiesterases, Type 2 metabolism, Inflammation metabolism, Inflammation pathology, Lipopolysaccharides administration & dosage, Lung virology, Male, Mice, Mice, Inbred C57BL, Neutrophils pathology, Nitric Oxide Synthase Type II metabolism, Proteins metabolism, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Tidal Volume, Time Factors, Trachea, Ventilator-Induced Lung Injury metabolism, Ventilator-Induced Lung Injury pathology, Acute Lung Injury etiology, Cyclic Nucleotide Phosphodiesterases, Type 2 deficiency, Lung metabolism, Pulmonary Alveoli metabolism, Pulmonary Alveoli pathology
- Abstract
Phosphodiesterase 2A (PDE2A) is stimulated by cGMP to hydrolyze cAMP, a potent endothelial barrier-protective molecule. We previously found that lung PDE2A contributed to a mouse model of ventilator-induced lung injury (VILI). The purpose of the present study was to determine the contribution of PDE2A in a two-hit mouse model of 1-day intratracheal (IT) LPS followed by 4 h of 20 ml/kg tidal volume ventilation. Compared with IT water controls, LPS alone (3.75 μg/g body wt) increased lung PDE2A mRNA and protein expression by 6 h with a persistent increase in protein through day 4 before decreasing to control levels on days 6 and 10. Similar to the PDE2A time course, the peak in bronchoalveolar lavage (BAL) neutrophils, lactate dehydrogenase (LDH), and protein concentration also occurred on day 4 post-LPS. IT LPS (1 day) and VILI caused a threefold increase in lung PDE2A and inducible nitric oxide synthase (iNOS) and a 24-fold increase in BAL neutrophilia. Compared with a control adenovirus, PDE2A knockdown with an adenovirus expressing a short hairpin RNA administered IT 3 days before LPS/VILI effectively decreased lung PDE2A expression and significantly attenuated BAL neutrophilia, LDH, protein, and chemokine levels. PDE2A knockdown also reduced lung iNOS expression by 53%, increased lung cAMP by nearly twofold, and improved survival from 47 to 100%. We conclude that in a mouse model of LPS/VILI, a synergistic increase in lung PDE2A expression increased lung iNOS and alveolar inflammation and contributed significantly to the ensuing acute lung injury.
- Published
- 2011
- Full Text
- View/download PDF
20. Induction of the apoptosis inhibitor ARC by Ras in human cancers.
- Author
-
Wu L, Nam YJ, Kung G, Crow MT, and Kitsis RN
- Subjects
- Animals, Caspases metabolism, Cell Line, Tumor, Cytoskeletal Proteins metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression Regulation, Neoplastic, Genes, ras genetics, Humans, MAP Kinase Kinase Kinases metabolism, Mice, Nerve Tissue Proteins metabolism, Proteasome Inhibitors, Rats, Apoptosis, Cytoskeletal Proteins physiology, Nerve Tissue Proteins physiology, Skin Neoplasms metabolism, ras Proteins metabolism
- Abstract
Inhibition of apoptosis is critical for carcinogenesis. ARC (apoptosis repressor with caspase recruitment domain) is an endogenous inhibitor of apoptosis that antagonizes both intrinsic and extrinsic apoptosis pathways. Although normally expressed in striated myocytes and neurons, ARC is markedly induced in a variety of primary human epithelial cancers and renders cancer cells resistant to killing. The mechanisms that mediate the induction of ARC in cancer are unknown. Herein we demonstrate that increases in ARC abundance are stimulated by Ras through effects on transcription and protein stability. Overexpression of activated N-Ras or H-Ras in normal cells is sufficient to increase ARC mRNA and protein levels. Similarly, transgenic expression of activated H-Ras induces ARC in both the normal mammary epithelium and resulting tumors of intact mice. Conversely, knockdown of endogenous N-Ras in breast and colon cancer cells significantly reduces ARC mRNA and protein levels. The promoter of the Nol3 locus, encoding ARC, is activated by N-Ras and H-Ras in a MEK/ERK-dependent manner. Ras also stabilizes ARC protein by suppressing its polyubiquitination and subsequent proteasomal degradation. In addition to the effects of Ras on ARC abundance, ARC mediates Ras-induced cell survival and cell cycle progression. Thus, Ras induces ARC in epithelial cancers, and ARC plays a role in the oncogenic actions of Ras.
- Published
- 2010
- Full Text
- View/download PDF
21. Mycophenolate mofetil modifies kidney tubular injury and Foxp3+ regulatory T cell trafficking during recovery from experimental ischemia-reperfusion.
- Author
-
Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Soloski MJ, Crow MT, and Rabb H
- Subjects
- Animals, Disease Models, Animal, Kidney drug effects, Kidney Tubular Necrosis, Acute pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mycophenolic Acid therapeutic use, Reperfusion Injury therapy, Forkhead Transcription Factors metabolism, Immunosuppressive Agents therapeutic use, Kidney pathology, Kidney Tubular Necrosis, Acute drug therapy, Mycophenolic Acid analogs & derivatives, Reperfusion Injury drug therapy, T-Lymphocytes, Regulatory cytology
- Abstract
Lymphocytes participate in the early pathogenesis of ischemia-reperfusion injury (IRI) in kidney; however, their role during repair is largely unknown. Recent data have shown that Foxp3(+) regulatory T cells (Tregs) traffic into kidney during healing from IRI and directly participate in repair. Since lymphocyte-targeting therapy is currently administered to prevent rejection during recovery from IRI in renal transplants, we hypothesized that mycophenolate mofetil (MMF) would alter Treg trafficking and kidney repair. C57BL/6J and T cell deficient mice underwent unilateral clamping of renal pedicle for 45 min, followed by reperfusion, and were sacrificed at day 10. Mice were treated with saline (C) or MMF (100mg/kg) i.p. daily starting at day 2 until sacrifice (n=5-12/group). MMF worsened kidney tubular damage compared to C at 10 days (cortex and outer medulla: p<0.05) in wild-type mice; tubular apoptotic index was increased in cortex in MMF group as well (p=0.01). MMF reduced the total number of kidney-infiltrating mononuclear cells (p<0.001 versus C) and the percentages of TCRbeta(+)CD4(+) and TCRbeta(+)CD8(+) T cells (p<0.01), but not natural killer (NK), NKT or B lymphocytes. MMF specifically reduced kidney Foxp3(+) Tregs (0.82+/-0.11% versus 1.75+/-0.17%, p<0.05). Tubular proliferative index and tissue levels of basic FGF were increased in MMF group (p<0.05), IL-10 and IL-6 were decreased (p<0.05). To evaluate if MMF effect occurred through non-lymphocytic cells, T cell deficient mice were treated with MMF. Tubular injury in T cell deficient mice was not affected by MMF treatment, though MMF-treated animals had increased VEGF and decreased PDGF-BB protein tissue levels compared to controls (p<0.05). Thus, MMF modifies the structural, epithelial proliferative and inflammatory response during healing, likely through effects on T cells and possibly Tregs. Kidney repair after IRI can be altered by agents that target lymphocytes., (Copyright (c) 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
22. Transcription factor Nrf2 is protective during ischemic and nephrotoxic acute kidney injury in mice.
- Author
-
Liu M, Grigoryev DN, Crow MT, Haas M, Yamamoto M, Reddy SP, and Rabb H
- Subjects
- Acetylcysteine pharmacology, Acute Kidney Injury chemically induced, Acute Kidney Injury pathology, Animals, Antineoplastic Agents toxicity, Capillary Permeability, Cisplatin toxicity, Glutathione pharmacology, Inflammation Mediators metabolism, Kidney pathology, Kidney Function Tests, Mice, Mice, Inbred ICR, Mice, Knockout, NF-E2-Related Factor 2 genetics, Oligonucleotide Array Sequence Analysis, Up-Regulation, Acute Kidney Injury metabolism, NF-E2-Related Factor 2 metabolism, Reperfusion Injury metabolism
- Abstract
Oxidative stress is involved in acute kidney injury due to ischemia-reperfusion and chemotherapy-induced nephrotoxicity. To investigate their basic mechanisms we studied the role of nuclear factor-erythroid 2-p45-related factor 2 (Nrf2), a redox-sensitive transcription factor that regulates expression of several antioxidant and cytoprotective genes. We compared the responses of Nrf2-knockout mice and their wild-type littermates in established mouse models of ischemia-reperfusion injury and cisplatin-induced nephrotoxicity. Several Nrf2-regulated genes encoding antioxidant enzymes/proteins were significantly upregulated in the kidneys of wild type but not Nrf2-knockout mice following renal ischemia. Renal function, histology, vascular permeability, and survival were each significantly worse in the Nrf2 knockout mice. Further, proinflammatory cytokine and chemokine expression tended to increase after ischemia in the knockout compared to the wild-type mice. Treatment of the knockout mice with the antioxidants N-acetyl-cysteine or glutathione improved renal function. The knockout mice were more susceptible to cisplatin-induced nephrotoxicity, and this was blunted by N-acetyl-cysteine pretreatment. Our study demonstrates that Nrf2-deficiency enhances susceptibility to both ischemic and nephrotoxic acute kidney injury, and identifies this transcription factor as a potential therapeutic target in these injuries.
- Published
- 2009
- Full Text
- View/download PDF
23. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMalpha) induces the vascular and hemodynamic changes of pulmonary hypertension.
- Author
-
Angelini DJ, Su Q, Yamaji-Kegan K, Fan C, Skinner JT, Champion HC, Crow MT, and Johns RA
- Subjects
- Animals, Cell Line, Chronic Disease, Disease Models, Animal, Gene Knockdown Techniques, Gene Transfer Techniques, Humans, Hypertension, Pulmonary pathology, Lung pathology, Lung physiopathology, Male, Rats, Rats, Sprague-Dawley, Blood Vessels physiopathology, Hemodynamics physiology, Hypertension, Pulmonary complications, Hypertension, Pulmonary physiopathology, Hypoxia complications, Nerve Growth Factor metabolism
- Abstract
Pulmonary hypertension (PH) is a serious disease of multiple etiologies mediated by hypoxia, immune stimuli, and elevated pulmonary pressure that leads to vascular thickening and eventual right heart failure. In a chronic hypoxia model of PH, we previously reported the induction of a novel pleiotropic cytokine, hypoxia-induced mitogenic factor (HIMF), that exhibits mitogenic, vasculogenic, contractile, and chemokine properties during PH-associated vascular remodeling. To examine the role of HIMF in hypoxia-induced vascular remodeling, we performed in vivo knockdown of HIMF using short hairpin RNA directed at rat HIMF in the chronic hypoxia model of PH. Knockdown of HIMF partially blocked increases in mean pulmonary artery pressure, pulmonary vascular resistance, right heart hypertrophy, and vascular remodeling caused by chronic hypoxia. To demonstrate a direct role for HIMF in the mechanism of PH development, we performed HIMF-gene transfer into the lungs of rats using a HIMF-expressing adeno-associated virus (AAV). AAV-HIMF alone caused development of PH similar to that of chronic hypoxia with increased mean pulmonary artery pressure and pulmonary vascular resistance, right heart hypertrophy, and neomuscularization and thickening of small pulmonary arterioles. The findings suggest that HIMF represents a critical cytokine-like growth factor in the development of PH.
- Published
- 2009
- Full Text
- View/download PDF
24. Expression, activity, and pro-hypertrophic effects of PDE5A in cardiac myocytes.
- Author
-
Zhang M, Koitabashi N, Nagayama T, Rambaran R, Feng N, Takimoto E, Koenke T, O'Rourke B, Champion HC, Crow MT, and Kass DA
- Subjects
- Animals, Base Sequence, Cells, Cultured, Cyclic GMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 genetics, Endothelin-1 pharmacology, Gene Knockdown Techniques, Gene Silencing, Luminescent Proteins analysis, Luminescent Proteins metabolism, MicroRNAs metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Natriuretic Peptides metabolism, Nitric Oxide Synthase Type III metabolism, Phenylephrine pharmacology, Phosphodiesterase 5 Inhibitors, Piperazines pharmacology, Purines pharmacology, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Sildenafil Citrate, Sulfones pharmacology, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Myocytes, Cardiac enzymology
- Abstract
Cyclic GMP-selective phosphodiesterase type 5 (PDE5) has been traditionally thought to play a little role in cardiac myocytes, yet recent studies using selective inhibitors such as sildenafil suggest it can potently modulate acute and chronic cardiac stress responses. To date, evidence for myocyte PDE5 expression and regulation has relied on small-molecule inhibitors and anti-sera, leaving open concerns regarding non-specific immune-reactivity, and off-target drug effects. To directly address both issues, we engineered a robust PDE5-gene silencing shRNA (inserted into miRNA-155 cassette) and DsRed-PDE5 fusion protein, both coupled to a CMV promoter and incorporated into adenoviral vectors. PDE5 mRNA and protein knock-down eliminated anti-sera positivity on immunoblots and fluorescent immuno-histochemistry in neonatal and adult cardiomyocytes, and suppressed PDE5 enzyme activity. Stimulation of myocyte hypertrophy by phenylephrine was blunted by PDE5 gene silencing in a protein kinase G dependent manner, and this effect was similar to that from sildenafil with no additive response by both combined. DsRed-PDE5 fusion protein expression showed normal z-band localization in adult myocytes but was diffused in eNOS(-/-) myocytes; echoing reported findings with anti-sera. PDE5 overexpression increased enzyme activity and amplified natriuretic peptide gene expression from phenylephrine stimulation. These data confirm PDE5 expression, activity, and targeted inhibition by sildenafil in cardiomyocytes, as well as the role of this PDE in cardiomyocyte hypertrophy modulation.
- Published
- 2008
- Full Text
- View/download PDF
25. Sphingosine 1-phosphate rescues canine LPS-induced acute lung injury and alters systemic inflammatory cytokine production in vivo.
- Author
-
Szczepaniak WS, Zhang Y, Hagerty S, Crow MT, Kesari P, Garcia JG, Choi AM, Simon BA, and McVerry BJ
- Subjects
- Acute Lung Injury chemically induced, Animals, Bronchoalveolar Lavage Fluid cytology, Bronchoalveolar Lavage Fluid immunology, Cytokines metabolism, Disease Models, Animal, Dogs, Lipopolysaccharides pharmacology, Lung immunology, Male, Neutrophils immunology, Respiration, Artificial, Sphingosine pharmacology, Acute Lung Injury drug therapy, Acute Lung Injury immunology, Cytokines immunology, Lysophospholipids pharmacology, Sphingosine analogs & derivatives
- Abstract
S1P has been demonstrated to protect against the formation of lipopolysaccharide (LPS)-induced lung edema when administered concomitantly with LPS. In the current study, we sought to determine the effectiveness of S1P to attenuate lung injury in a translationally relevant canine model of ALI when administered as rescue therapy. Secondarily, we examined whether the attenuation of LPS-induced physiologic lung injury after administration of S1P was, at least in part, caused by an alteration in local and/or systemic inflammatory cytokine expression. We examined 18, 1-year-old male beagles prospectively in which we instilled bacterial LPS (2-4 mg/kg) intratracheally followed in 1 h with intravenous S1P (85 microg/kg) or vehicle and 8 h of high-tidal-volume mechanical ventilation. S1P attenuated the formation of Q(s)/Q(t) (32%), and both the presence of protein (72%) and neutrophils (95%) in BAL fluid compared with vehicle controls. Although lung tissue inflammatory cytokine production was found to vary regionally throughout the LPS-injured lung, S1P did not alter the expression pattern. Similarly, BAL cytokine production was not altered significantly by intravenous S1P in this model. Interestingly, S1P potentiated the LPS-induced systemic production of 3 inflammatory cytokines, TNF-alpha (6-fold), KC (1.2-fold), and IL-6 (3-fold), without resulting in end-organ dysfunction. In conclusion, intravenous S1P reduces inflammatory lung injury when administered as rescue therapy in our canine model of LPS-induced ALI. This improvement is observed in the absence of changes in local pulmonary inflammatory cytokine production and an augmentation of systemic inflammation.
- Published
- 2008
- Full Text
- View/download PDF
26. Alveolar cell apoptosis is dependent on p38 MAP kinase-mediated activation of xanthine oxidoreductase in ventilator-induced lung injury.
- Author
-
Le A, Damico R, Damarla M, Boueiz A, Pae HH, Skirball J, Hasan E, Peng X, Chesley A, Crow MT, Reddy SP, Tuder RM, and Hassoun PM
- Subjects
- Animals, Capillary Permeability, Caspase 3 metabolism, Caspase 7 metabolism, Disease Models, Animal, Enzyme Inhibitors pharmacology, Lung Diseases enzymology, Lung Diseases etiology, Lung Diseases prevention & control, Male, Mice, Mice, Inbred C57BL, Phosphorylation, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases metabolism, Pulmonary Alveoli blood supply, Pulmonary Alveoli drug effects, Pulmonary Alveoli enzymology, Pulmonary Alveoli injuries, Respiration, Artificial adverse effects, Time Factors, Xanthine Dehydrogenase antagonists & inhibitors, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Apoptosis drug effects, Lung Diseases pathology, Pulmonary Alveoli pathology, Xanthine Dehydrogenase metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Signaling via p38 MAP kinase has been implicated in the mechanotransduction associated with mechanical stress and ventilator-induced lung injury (VILI). However, the critical downstream mediators of alveolar injury remain incompletely defined. We provide evidence that high-tidal volume mechanical ventilation (HVt MV) rapidly activates caspases within the lung, resulting in increased alveolar cell apoptosis. Antagonism of MV-induced p38 MAP kinase activity with SB-203580 suppresses both MV-induced caspase activity and alveolar apoptosis, placing p38 MAP kinase upstream of MV-induced caspase activation and programmed cell death. The reactive oxygen species (ROS)-producing enzyme xanthine oxidoreductase (XOR) is activated in a p38 MAP kinase-dependent manner following HVt MV. Allopurinol, a XOR inhibitor, also suppresses HVt MV-induced apoptosis, implicating HVt MV-induced ROS in the induction of alveolar cell apoptosis. Finally, systemic administration of the pan-caspase inhibitor, z-VAD-fmk, but not its inactive peptidyl analog, z-FA-fmk, blocks ventilator-induced apoptosis of alveolar cells and alveolar-capillary leak, indicating that caspase-dependent cell death is necessary for VILI-associated barrier dysfunction in vivo.
- Published
- 2008
- Full Text
- View/download PDF
27. Macrophage migration inhibitory factor governs endothelial cell sensitivity to LPS-induced apoptosis.
- Author
-
Damico RL, Chesley A, Johnston L, Bind EP, Amaro E, Nijmeh J, Karakas B, Welsh L, Pearse DB, Garcia JG, and Crow MT
- Subjects
- CASP8 and FADD-Like Apoptosis Regulating Protein genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Cell Death drug effects, DNA Primers, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Gene Expression Regulation drug effects, Humans, Macrophage Migration-Inhibitory Factors physiology, Pulmonary Artery cytology, Pulmonary Artery drug effects, RNA Interference, RNA, Messenger genetics, RNA, Small Interfering genetics, Respiratory Mucosa drug effects, Reverse Transcriptase Polymerase Chain Reaction, Apoptosis drug effects, Endothelium, Vascular physiology, Lipopolysaccharides toxicity, Macrophage Migration-Inhibitory Factors genetics, Pulmonary Artery physiology, Respiratory Mucosa cytology, Respiratory Mucosa physiology
- Abstract
Human endothelial cells (EC) are typically resistant to the apoptotic effects of stimuli associated with lung disease. The determinants of this resistance remain incompletely understood. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine produced by human pulmonary artery EC (HPAEC). Its expression increases in response to various death-inducing stimuli, including lipopolysaccharide (LPS). We show here that silencing MIF expression by RNA interference (MIF siRNA) dramatically reduces MIF mRNA expression and the LPS-induced increase in MIF protein levels, thereby sensitizing HPAECs to LPS-induced cell death. Addition of recombinant human MIF (rhMIF) protein prevents the death-sensitizing effect of MIF siRNA. A common mediator of apoptosis resistance in ECs is the death effector domain (DED)-containing protein, FLIP (FLICE-like inhibitory protein). We show that LPS induces a transcription-independent increase in the short isoform of FLIP (FLIP(s)). This increase is blocked by MIF siRNA but restored with the addition of recombinant MIF protein (rHMIF). While FLIP(s) siRNA also sensitizes HPAECs to LPS-induced death, the addition of rhMIF does not affect this sensitization, placing MIF upstream of FLIP(s) in preventing HPAEC death. These studies demonstrate that MIF is an endogenous pro-survival factor in HPAECs and identify a novel mechanism for its role in apoptosis resistance through the regulation of FLIP(s). These results show that MIF can protect vascular endothelial cells from inflammation-associated cell damage.
- Published
- 2008
- Full Text
- View/download PDF
28. Genetic and pharmacologic evidence links oxidative stress to ventilator-induced lung injury in mice.
- Author
-
Papaiahgari S, Yerrapureddy A, Reddy SR, Reddy NM, Dodd-O JM, Crow MT, Grigoryev DN, Barnes K, Tuder RM, Yamamoto M, Kensler TW, Biswal S, Mitzner W, Hassoun PM, and Reddy SP
- Subjects
- Acetylcysteine pharmacology, Animals, Capillary Permeability drug effects, Capillary Permeability physiology, Cytokines immunology, Disease Models, Animal, Free Radical Scavengers pharmacology, Inflammation prevention & control, Male, Mice, Mice, Knockout, Oxidative Stress immunology, Respiratory Distress Syndrome drug therapy, Respiratory Distress Syndrome physiopathology, Antioxidants metabolism, NF-E2-Related Factor 2 genetics, Oxidative Stress drug effects, Respiratory Distress Syndrome immunology, Ventilators, Mechanical adverse effects
- Abstract
Rationale: Mechanical ventilation (MV) is an indispensable therapy for critically ill patients with acute lung injury and the adult respiratory distress syndrome. However, the mechanisms by which conventional MV induces lung injury remain unclear., Objectives: We hypothesized that disruption of the gene encoding Nrf2, a transcription factor that regulates the induction of several antioxidant enzymes, enhances susceptibility to ventilator-induced lung injury (VILI) and that antioxidant supplementation attenuates this effect., Methods: To test our hypothesis and to examine the relevance of oxidative stress in VILI, we assessed lung injury and inflammatory responses in Nrf2-deficient (Nrf2(-/-)) mice and wild-type (Nrf2(+/+)) mice after an acute (2-h) injurious model of MV with or without administration of antioxidant., Measurements and Main Results: Nrf2(-/-) mice displayed greater levels of lung alveolar and vascular permeability and inflammatory responses to MV as compared with Nrf2(+/+) mice. Nrf2 deficiency enhances the levels of several proinflammatory cytokines implicated in the pathogenesis of VILI. We found diminished levels of critical antioxidant enzymes and redox imbalance by MV in the lungs of Nrf2(-/-) mice; however, antioxidant supplementation to Nrf2(-/-) mice remarkably attenuated VILI. When subjected to a clinically relevant prolong period of MV, Nrf2(-/-) mice displayed greater levels of VILI than Nrf2(+/+) mice. Expression profiling revealed lack of induction of several VILI genes, stress response and solute carrier proteins, and phosphatases in Nrf2(-/-) mice., Conclusions: Our data demonstrate for the first time a critical role for Nrf2 in VILI, which confers protection against cellular responses induced by MV by modulating oxidative stress.
- Published
- 2007
- Full Text
- View/download PDF
29. The pro-angiogenic cytokine pleiotrophin potentiates cardiomyocyte apoptosis through inhibition of endogenous AKT/PKB activity.
- Author
-
Li J, Wei H, Chesley A, Moon C, Krawczyk M, Volkova M, Ziman B, Margulies KB, Talan M, Crow MT, and Boheler KR
- Subjects
- Animals, Apoptosis, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Hypoxia, Male, Myocardium metabolism, Phosphorylation, RNA, Small Interfering metabolism, Rats, Rats, Sprague-Dawley, Signal Transduction, Carrier Proteins metabolism, Cytokines metabolism, Myocytes, Cardiac metabolism, Neovascularization, Pathologic, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Pleiotrophin is a development-regulated cytokine and growth factor that can promote angiogenesis, cell proliferation, or differentiation, and it has been reported to have neovasculogenic effects in damaged heart. Developmentally, it is prominently expressed in fetal and neonatal hearts, but it is minimally expressed in normal adult heart. Conversely, we show in a rat model of myocardial infarction and in human dilated cardiomyopathy that pleiotrophin is markedly up-regulated. To elucidate the effects of pleiotrophin on cardiac contractile cells, we employed primary cultures of rat neonatal and adult cardiomyocytes. We show that pleiotrophin is released from cardiomyocytes in vitro in response to hypoxia and that the addition of recombinant pleiotrophin promotes caspase-mediated genomic DNA fragmentation in a dose- and time-dependent manner. Functionally, it potentiates the apoptotic response of neonatal cardiomyocytes to hypoxic stress and to ultraviolet irradiation and of adult cardiomyocytes to hypoxia-reoxygenation. Moreover, UV-induced apoptosis in neonatal cardiomyocytes can be partially inhibited by small interfering RNA-mediated knockdown of endogenous pleiotrophin. Mechanistically, pleiotrophin antagonizes IGF-1 associated Ser-473 phosphorylation of AKT/PKB, and it concomitantly decreases both BAD and GSK3beta phosphorylation. Adenoviral expression of constitutively active AKT and lithium chloride-mediated inhibition of GSK3beta reduce the potentiated programmed cell death elicited by pleiotrophin. These latter data indicate that pleiotrophin potentiates cardiomyocyte cell death, at least partially, through inhibition of AKT signaling. In conclusion, we have uncovered a novel function for pleiotrophin on heart cells following injury. It fosters cardiomyocyte programmed cell death in response to pro-apoptotic stress, which may be critical to myocardial injury repair.
- Published
- 2007
- Full Text
- View/download PDF
30. Activation of CaMKIIdeltaC is a common intermediate of diverse death stimuli-induced heart muscle cell apoptosis.
- Author
-
Zhu W, Woo AY, Yang D, Cheng H, Crow MT, and Xiao RP
- Subjects
- Adrenergic beta-1 Receptor Agonists, Animals, Benzylamines pharmacology, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Calcium-Calmodulin-Dependent Protein Kinases genetics, Cells, Cultured, Cytochromes c metabolism, Enzyme Activation drug effects, Enzyme Activation genetics, Gene Expression, Mitochondria, Heart enzymology, Myocytes, Cardiac cytology, Protein Kinase Inhibitors, Rats, Rats, Sprague-Dawley, Receptors, Adrenergic, beta-1 metabolism, Sulfonamides pharmacology, bcl-X Protein metabolism, Apoptosis drug effects, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Myocytes, Cardiac enzymology, Signal Transduction drug effects, Signal Transduction genetics
- Abstract
Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) is expressed in many mammalian cells, with the delta isoform predominantly expressed in cardiomyocytes. Previous studies have shown that inhibition of CaMKII protects cardiomyocytes against beta(1)-adrenergic receptor-mediated apoptosis. However, it is unclear whether activation of CaMKII is sufficient to cause cardiomyocyte apoptosis and whether CaMKII signaling is important in heart muscle cell apoptosis mediated by other stimuli. Here, we specifically enhanced or suppressed CaMKII activity using adenoviral gene transfer of constitutively active (CA-CaMKII(deltaC)) or dominant negative (DN-CaMKII(deltaC)) mutants of CaMKII(deltaC) in cultured adult rat cardiomyocytes. Expression of CA-CaMKII(deltaC) promoted cardiomyocyte apoptosis that was associated with increased mitochondrial cytochrome c release and attenuated by co-expression of Bcl-X(L). Importantly, isoform-specific suppression of CaMKII(deltaC) with the DN-CaMKII(deltaC) mutant similar to nonselective CaMKII inhibition by the pharmacological inhibitors (KN-93 or AIP) not only prevented CA-CaMKII(deltaC)-mediated apoptosis but also protected cells from multiple death-inducing stimuli. Thus, activation of CaMKII(deltaC) constitutes a common intermediate by which various death-inducing stimuli trigger cardiomyocyte apoptosis via the primary mitochondrial death pathway.
- Published
- 2007
- Full Text
- View/download PDF
31. Revisiting p53 and its effectors in ischemic heart injury.
- Author
-
Crow MT
- Subjects
- Animals, Apoptosis physiology, Heart Rupture, Post-Infarction metabolism, Heart Rupture, Post-Infarction pathology, Humans, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Mice, Mice, Knockout, Models, Animal, Myocardial Ischemia pathology, Myocardium pathology, Tumor Suppressor Protein p53 genetics, Myocardial Ischemia metabolism, Myocardium metabolism, Tumor Suppressor Protein p53 physiology
- Published
- 2006
- Full Text
- View/download PDF
32. Apoptosis repressor with caspase recruitment domain (ARC) is expressed in cancer cells and localizes to nuclei.
- Author
-
Wang M, Qanungo S, Crow MT, Watanabe M, and Nieminen AL
- Subjects
- Active Transport, Cell Nucleus, Animals, Apoptosis Regulatory Proteins, Cell Line, Tumor, Humans, Rats, Cell Nucleus metabolism, Gene Expression Regulation, Neoplastic, Muscle Proteins metabolism, Neoplasms metabolism, Neoplasms pathology
- Abstract
Apoptosis repressor with caspase recruitment domain is expressed at high levels in brain and myogenic tissues, consistent with a role to inhibit apoptosis in the terminally differentiated cells. Expression of ARC in cancers is not known. In this study, we reported that ARC was highly expressed in various non-myogenic and non-neurogenic human and rat cancer cell lines. Unexpectedly, ARC was localized almost exclusively to the nuclei of cancer cells, which was unlike the cytoplasmic localization of ARC in non-cancer cells. Furthermore, nuclear ARC in cancer cells did not co-localize with nucleolus protein of 30 kDa, an alternatively spliced ARC isoform. These findings indicate that ARC is distributed differently in cancer cells than non-cancer cells and thus might play a role in neoplastic transformation.
- Published
- 2005
- Full Text
- View/download PDF
33. Heart failure after long-term supravalvular aortic constriction in rats.
- Author
-
Boluyt MO, Robinson KG, Meredith AL, Sen S, Lakatta EG, Crow MT, Brooks WW, Conrad CH, and Bing OH
- Subjects
- Animals, Aorta surgery, Cardiac Output, Low metabolism, Cardiomegaly etiology, Chronic Disease, Constriction, Pathologic, Contractile Proteins genetics, Elasticity, Extracellular Matrix Proteins genetics, Fibrosis, Gene Expression, Heart physiopathology, In Vitro Techniques, Ligation, Male, Myocardial Contraction, Myocardium pathology, Papillary Muscles, RNA, Messenger metabolism, Rats, Rats, Inbred WKY, Aorta physiopathology, Cardiac Output, Low etiology
- Abstract
Background: Pressure overload in humans follows a chronic and progressive course, often resulting in eventual cardiac decompensation and death. Animal models of heart failure generally fail to mimic the temporal features observed in human disease often covering a major portion of the life span, and findings of short-term studies are of uncertain applicability. The purpose was to determine whether chronic pressure overload introduced gradually in young normotensive rats would lead predictably to heart failure and to characterize specific phenotype features that have been well documented in another model of heart failure., Methods: Rats underwent banding of the ascending aorta at 7 weeks of age such that the hemodynamic load increased gradually with ontogenic growth. Two groups of hypertrophied hearts from aortic-banded rats, with and without signs of heart failure, were compared with those of control rats at a mean age of 11 months., Results: Hearts of aorta-banded rats underwent a transition from stable compensated hypertrophy to heart failure that was characterized by augmented hypertrophy, depressed contractile function, elevated fibrosis, increased myocardial stiffness, and marked alterations in the expression of genes encoding contractile, regulatory, and extracellular matrix proteins., Conclusions: Gradual constriction of the rat aorta resulted in heart failure after a variable length of time (3 to 18 months). Despite differences in genotype, the ultimate phenotype associated with the transition to failure in the aorta-banded rat is nearly identical to that observed in the aged spontaneously hypertensive rat (SHR), with a few notable differences. The findings suggest that a common heart failure phenotype follows long-term pressure overload regardless of the underlying etiology.
- Published
- 2005
- Full Text
- View/download PDF
34. Phosphatidylinositol 3-kinase offsets cAMP-mediated positive inotropic effect via inhibiting Ca2+ influx in cardiomyocytes.
- Author
-
Leblais V, Jo SH, Chakir K, Maltsev V, Zheng M, Crow MT, Wang W, Lakatta EG, and Xiao RP
- Subjects
- Androstadienes pharmacology, Animals, Calcium Signaling physiology, Cells, Cultured drug effects, Cells, Cultured enzymology, Cells, Cultured metabolism, Chromones pharmacology, Colforsin pharmacology, Cyclic AMP pharmacology, Cyclic AMP-Dependent Protein Kinases pharmacology, Enzyme Activation drug effects, GTP-Binding Protein alpha Subunits, Gi-Go antagonists & inhibitors, GTP-Binding Protein alpha Subunits, Gi-Go physiology, GTP-Binding Protein beta Subunits physiology, GTP-Binding Protein gamma Subunits physiology, Insulin-Like Growth Factor I pharmacology, Ion Transport drug effects, Isoproterenol pharmacology, Morpholines pharmacology, Myocardial Contraction, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Norepinephrine pharmacology, Patch-Clamp Techniques, Peptide Fragments pharmacology, Pertussis Toxin pharmacology, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Prazosin pharmacology, Propanolamines pharmacology, Protein Processing, Post-Translational drug effects, Rats, Receptors, Adrenergic, beta-1 physiology, Recombinant Proteins pharmacology, Wortmannin, Calcium metabolism, Calcium Channels, L-Type metabolism, Cyclic AMP antagonists & inhibitors, Cyclic AMP-Dependent Protein Kinases physiology, Myocytes, Cardiac enzymology, Phosphatidylinositol 3-Kinases physiology, Second Messenger Systems physiology, Signal Transduction physiology
- Abstract
Phosphoinositide 3-kinase (PI3K) has been implicated in beta2-adrenergic receptor (beta2-AR)/G(i)-mediated compartmentation of the concurrent G(s)-cAMP signaling, negating beta2-AR-induced phospholamban phosphorylation and the positive inotropic and lusitropic responses in cardiomyocytes. However, it is unclear whether PI3K crosstalks with the beta1-AR signal transduction, and even more generally, with the cAMP/PKA pathway. In this study, we show that selective beta1-AR stimulation markedly increases PI3K activity in adult rat cardiomyocytes. Inhibition of PI3K by LY294002 significantly enhances beta1-AR-induced increases in L-type Ca2+ currents, intracellular Ca2+ transients, and myocyte contractility, without altering the receptor-mediated phosphorylation of phospholamban. The LY294002 potentiating effects are completely prevented by betaARK-ct, a peptide inhibitor of beta-adrenergic receptor kinase-1 (betaARK1) as well as G(betagamma) signaling, but not by disrupting G(i) function with pertussis toxin. Moreover, forskolin, an adenylyl cyclase activator, also elevates PI3K activity and inhibition of PI3K enhances forskolin-induced contractile response in a betaARK-ct sensitive manner. In contrast, PI3K inhibition affects neither the basal contractility nor high extracellular Ca2+-induced increase in myocyte contraction. These results suggest that beta1-AR stimulation activates PI3K via a PKA-dependent mechanism, and that G(betagamma) and the subsequent activation of betaARK1 are critically involved in the PKA-induced PI3K signaling which, in turn, negates cAMP-induced positive inotropic effect via inhibiting sarcolemmal Ca2+ influx and the subsequent increase in intracellular Ca2+ transients, without altering the receptor-mediated phospholamban phosphorylation, in intact cardiomyocytes.
- Published
- 2004
- Full Text
- View/download PDF
35. The mitochondrial death pathway and cardiac myocyte apoptosis.
- Author
-
Crow MT, Mani K, Nam YJ, and Kitsis RN
- Subjects
- Animals, Apoptosis Inducing Factor, Caspases physiology, Cytochromes c metabolism, Endoplasmic Reticulum physiology, Enzyme Activation, Flavoproteins pharmacology, Gene Expression Regulation, Heart Diseases pathology, Humans, Ion Channels physiology, Membrane Proteins pharmacology, Mitochondrial Membrane Transport Proteins, Mitochondrial Permeability Transition Pore, Protein Structure, Tertiary, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins physiology, Rodentia, Stress, Physiological pathology, Tumor Suppressor Protein p53 physiology, Apoptosis physiology, Mitochondria, Heart physiology, Myocytes, Cardiac cytology
- Abstract
Apoptosis has been causally linked to the pathogenesis of myocardial infarction and heart failure in rodent models. This death process is mediated by two central pathways, an extrinsic pathway involving cell surface receptors and an intrinsic pathway using mitochondria and the endoplasmic reticulum. Each of these pathways has been implicated in myocardial pathology. In this review, we summarize recent advances in the understanding of the intrinsic pathway and how it relates to cardiac myocyte death and heart disease.
- Published
- 2004
- Full Text
- View/download PDF
36. Sir-viving cardiac stress: cardioprotection mediated by a longevity gene.
- Author
-
Crow MT
- Subjects
- Acetylation, Animals, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins physiology, Cell Survival, Forkhead Transcription Factors, Gene Silencing, Histone Deacetylases physiology, Insulin physiology, Mice, Myocytes, Cardiac cytology, Nuclear Proteins physiology, Protein Processing, Post-Translational genetics, Rats, Rats, Wistar, Receptor, Insulin genetics, Receptor, Insulin physiology, Signal Transduction genetics, Silent Information Regulator Proteins, Saccharomyces cerevisiae physiology, Sirtuin 1, Sirtuin 2, Sirtuins genetics, Species Specificity, Stress, Physiological complications, Stress, Physiological genetics, Stress, Physiological metabolism, Transcription Factors physiology, Transcription, Genetic, Longevity genetics, Myocardium metabolism, Sirtuins physiology
- Published
- 2004
- Full Text
- View/download PDF
37. Sustained beta1-adrenergic stimulation modulates cardiac contractility by Ca2+/calmodulin kinase signaling pathway.
- Author
-
Wang W, Zhu W, Wang S, Yang D, Crow MT, Xiao RP, and Cheng H
- Subjects
- Adrenergic alpha-Agonists pharmacology, Animals, Calcium Signaling drug effects, Calcium-Binding Proteins metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Calcium-Calmodulin-Dependent Protein Kinases genetics, Cell Size drug effects, Cells, Cultured drug effects, Cells, Cultured physiology, Humans, Male, Mutagenesis, Site-Directed, Myocardial Contraction drug effects, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology, Norepinephrine pharmacology, Phosphorylation, Prazosin pharmacology, Protein Processing, Post-Translational, Rats, Rats, Sprague-Dawley, Receptors, Adrenergic, beta-1 drug effects, Recombinant Fusion Proteins physiology, Second Messenger Systems drug effects, Calcium Signaling physiology, Calcium-Calmodulin-Dependent Protein Kinases physiology, Cyclic AMP physiology, Cyclic AMP-Dependent Protein Kinases physiology, Myocardial Contraction physiology, Receptors, Adrenergic, beta-1 physiology, Second Messenger Systems physiology
- Abstract
A tenet of beta1-adrenergic receptor (beta1AR) signaling is that stimulation of the receptor activates the adenylate cyclase-cAMP-protein kinase A (PKA) pathway, resulting in positive inotropic and relaxant effects in the heart. However, recent studies have suggested the involvement of Ca2+/calmodulin-dependent protein kinase II (CaMKII) in beta1AR-stimulated cardiac apoptosis. In this study, we determined roles of CaMKII and PKA in sustained versus short-term beta1AR modulation of excitation-contraction (E-C) coupling in cardiac myocytes. Short-term (10-minute) and sustained (24-hour) beta1AR stimulation with norepinephrine similarly enhanced cell contraction and Ca2+ transients, in contrast to anticipated receptor desensitization. More importantly, the sustained responses were largely PKA-independent, and were sensitive to specific CaMKII inhibitors or adenoviral expression of a dominant-negative CaMKII mutant. Biochemical assays revealed that a progressive and persistent CaMKII activation was associated with a rapid desensitization of the cAMP/PKA signaling. Concomitantly, phosphorylation of phospholamban, an SR Ca2+ cycling regulatory protein, was shifted from its PKA site (16Ser) to CaMKII site (17Thr). Thus, beta1AR stimulation activates dual signaling pathways mediated by cAMP/PKA and CaMKII, the former undergoing desensitization and the latter exhibiting sensitization. This finding may bear important etiological and therapeutical ramifications in understanding beta1AR signaling in chronic heart failure.
- Published
- 2004
- Full Text
- View/download PDF
38. Regulation of c-Jun N-terminal kinase and p38 kinase pathways in endothelial cells.
- Author
-
Wadgaonkar R, Pierce JW, Somnay K, Damico RL, Crow MT, Collins T, and Garcia JG
- Subjects
- Antineoplastic Agents pharmacology, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Nucleus metabolism, Cells, Cultured, Cycloheximide pharmacology, Cytoplasm metabolism, Dual Specificity Phosphatase 1, Dual-Specificity Phosphatases, E-Selectin metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Enzyme Inhibitors pharmacology, Humans, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Interleukin-1 metabolism, Interleukin-1 pharmacology, JNK Mitogen-Activated Protein Kinases, Luciferases, Mitogen-Activated Protein Kinase Phosphatases, Mitogen-Activated Protein Kinases antagonists & inhibitors, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Phosphorylation, Promoter Regions, Genetic genetics, Protein Phosphatase 1, Protein Synthesis Inhibitors, Protein Tyrosine Phosphatases genetics, Protein Tyrosine Phosphatases metabolism, RNA, Small Interfering pharmacology, Tumor Necrosis Factor-alpha pharmacology, Umbilical Veins, p38 Mitogen-Activated Protein Kinases, E-Selectin genetics, Endothelial Cells drug effects, Gene Expression Regulation, Mitogen-Activated Protein Kinases metabolism, Signal Transduction
- Abstract
The rapid and transient induction of E-selectin gene expression by inflammatory tumor necrosis factor (TNF)-alpha in endothelial cells is mediated by signaling pathways which involve c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) kinase pathways. To explore this regulation, we first observed that in the continuous presence of cytokine TNF, activation of JNK-1 in both nuclear and cytoplasmic compartments peaked at 15-30 min, with activity returning to uninduced levels by 60 min. Phosphorylation of both the p38 kinase and its molecular target, the nuclear transcription factor, activating transcription factor-2, were transient after TNF-alpha or interleukin (IL)-1beta induction. However, cycloheximide treatment prolonged the TNF-alpha-induced JNK-1 kinase activity beyond 60 min, suggesting that protein synthesis is required to limit this signaling cascade. We investigated the possible role of the dual-specificity phosphatases MAPK phosphatase (MKP)-1 and MKP-2 in limiting cytokine-induced MAPK signaling. Maximum induction of MKP-1 mRNA and nuclear protein levels by TNF-alpha or IL-1beta were noted at 60 min and their expression correlated with the termination of JNK kinase activity, whereas nuclear levels of MKP-2 were not significantly affected by treatment with TNF-alpha or IL-1beta. Transient overexpression of MKP-1 demonstrated significant specific inhibition of E-selectin promoter activity consistent with a regulatory role for dual-specificity phosphatases. Inhibition of MKP-1 expression through the use of small interfering RNAs prolonged the cytokine-induced p38 and JNK kinase phosphorylation. Our results suggest that endogenous inhibitors of the MAPK cascade, such as the dual-specificity phosphatases like MKP-1 may be important for the postinduction repression of MAPK activity and E-selectin transcription in endothelial cells. Thus, these inhibitors may play an important role in limiting the inflammatory effects of TNF-alpha and IL-1beta.
- Published
- 2004
- Full Text
- View/download PDF
39. Modulation of vascular smooth muscle cell migration by calcium/ calmodulin-dependent protein kinase II-delta 2.
- Author
-
Pfleiderer PJ, Lu KK, Crow MT, Keller RS, and Singer HA
- Subjects
- Animals, Calcium metabolism, Calcium pharmacology, Calcium Signaling drug effects, Calcium Signaling genetics, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Calcium-Calmodulin-Dependent Protein Kinases genetics, Calmodulin metabolism, Calmodulin pharmacology, Cell Movement drug effects, Cell Movement genetics, Cells, Cultured, Enzyme Inhibitors pharmacology, Humans, Isoenzymes genetics, Isoenzymes metabolism, Muscle, Smooth, Vascular drug effects, Mutation genetics, Phosphorylation drug effects, Platelet-Derived Growth Factor pharmacology, Rats, Rats, Sprague-Dawley, Threonine metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Movement physiology, Muscle, Smooth, Vascular enzymology
- Abstract
Previous studies demonstrated a requirement for multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in PDGF-stimulated vascular smooth muscle (VSM) cell migration. In the present study, molecular approaches were used specifically to assess the role of the predominant CaMKII isoform (delta(2) or delta(C)) on VSM cell migration. Kinase-negative (K43A) and constitutively active (T287D) mutant forms of CaMKII delta(2) were expressed using recombinant adenoviruses. CaMKII activities were evaluated in vitro by using a peptide substrate and in intact cells by assessing the phosphorylation of overexpressed phospholamban on Thr(17), a CaMKII-selective phosphorylation site. Expression of kinase-negative CaMKII delta(2) inhibited substrate phosphorylation both in vitro and in the intact cell, indicating a dominant-negative function with respect to exogenous substrate. However, overexpression of the kinase-negative mutant failed to inhibit endogenous CaMKII delta(2) autophosphorylation on Thr(287) after activation of cells with ionomycin, and in fact, these subunits served as a substrate for the endogenous kinase. Constitutively active CaMKII delta(2) phosphorylated substrate in vitro without added Ca(2+)/calmodulin and in the intact cell without added Ca(2+)-dependent stimuli, but it inhibited autophosphorylation of endogenous CaMKII delta(2) on Thr(287). Basal and PDGF-stimulated cell migration was significantly enhanced in cells expressing kinase-negative CaMKII delta(2), an effect opposite that of KN-93, a chemical inhibitor of CaMKII activation. Expression of the constitutively active CaMKII delta(2) mutant inhibited PDGF-stimulated cell migration. These studies point to a role for the CaMKII delta(2) isoform in regulating VSM cell migration. An inclusive interpretation of results using both pharmacological and molecular approaches raises the hypothesis that CaMKII delta(2) autophosphorylation may play an important role in PDGF-stimulated VSM cell migration.
- Published
- 2004
- Full Text
- View/download PDF
40. Apoptosis repressor with caspase recruitment domain protects against cell death by interfering with Bax activation.
- Author
-
Gustafsson AB, Tsai JG, Logue SE, Crow MT, and Gottlieb RA
- Subjects
- Animals, Apoptosis Regulatory Proteins, Cell Line, Creatine Kinase metabolism, Cytochromes c metabolism, Myocardial Infarction enzymology, Myocardial Infarction prevention & control, Myocardium metabolism, Rats, Recombinant Fusion Proteins metabolism, bcl-2-Associated X Protein, Apoptosis physiology, Caspase Inhibitors, Cell Death physiology, Muscle Proteins metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2
- Abstract
Myocardial ischemia/reperfusion (I/R) is associated with an extensive loss of myocardial cells. The apoptosis repressor with caspase recruitment domain (ARC) is a protein that is highly expressed in heart and skeletal muscle and has been demonstrated to protect the heart against I/R injury (Gustafsson, A. B., Sayen, M. R., Williams, S. D., Crow, M. T., and Gottlieb, R. A. (2002) Circulation 106, 735-739). In this study, we have shown that transduction of TAT-ARCL31F, a mutant of ARC in the caspase recruitment domain, did not reduce creatine kinase release and infarct size after I/R. TAT-ARCL31F also failed to protect against hydrogen peroxide-mediated cell death in H9c2 cells, suggesting that the caspase recruitment domain is important in mediating ARC's protective effects. In addition, we report that ARC co-immunoprecipitated with the pro-apoptotic protein Bax, which causes cytochrome c release when activated. TAT-ARC, but not TAT-ARCL31F, prevented Bax activation and cytochrome c release in hydrogen peroxide-treated H9c2 cells. TAT-ARC was also effective in blocking cytochrome c release after ischemia and reperfusion, whereas TAT-ARCL31F had no effect on cytochrome c release. In addition, recombinant ARC protein abrogated Bax-induced cytochrome c release from isolated mitochondria. This suggests that ARC can protect against cell death by interfering with activation of the mitochondrial death pathway through the interaction with Bax, preventing mitochondrial dysfunction and release of pro-apoptotic factors.
- Published
- 2004
- Full Text
- View/download PDF
41. Phorbol esters increase MLC phosphorylation and actin remodeling in bovine lung endothelium without increased contraction.
- Author
-
Bogatcheva NV, Verin AD, Wang P, Birukova AA, Birukov KG, Mirzopoyazova T, Adyshev DM, Chiang ET, Crow MT, and Garcia JG
- Subjects
- Animals, Base Sequence, Cattle, Cells, Cultured, DNA Primers, Electric Conductivity, Endothelium, Vascular drug effects, Lung drug effects, Mutagenesis, Myosin Light Chains metabolism, Phosphorylation, Polymerase Chain Reaction, Pulmonary Circulation drug effects, RNA, Messenger genetics, Transcription, Genetic drug effects, Xenopus laevis, Actins genetics, Endothelium, Vascular physiology, Lung physiology, Myosin Light Chains genetics, Pulmonary Circulation physiology, Tetradecanoylphorbol Acetate pharmacology
- Abstract
Direct protein kinase C (PKC) activation with phorbol myristate acetate (PMA) results in the loss of endothelial monolayer integrity in bovine lung endothelial cells (EC) but produces barrier enhancement in human lung endothelium. To extend these findings, we studied EC contractile events and observed a 40% increase in myosin light chain (MLC) phosphorylation in bovine endothelium following PMA challenge. The increase in PMA-mediated MLC phosphorylation occurred at sites distinct from Ser19/Thr18, sites catalyzed by MLC kinase (MLCK), and immunoblotting with antibodies specific to phosphorylated Ser19/Thr18 demonstrated profound time-dependent Ser19/Thr18 dephosphorylation. These events occurred in conjunction with rearrangement of stress fibers into a grid-like network, but without an increase in cellular contraction as measured by silicone membrane wrinkling assay. The PMA-induced MLC dephosphorylation was not due to kinase inhibition but, rather, correlated with rapid increases in myosin-associated phosphatase 1 (PPase 1) activity. These data suggest that PMA-mediated EC barrier regulation may involve dual mechanisms that alter MLC phosphorylation. The increase in bovine MLC phosphorylation likely occurs via direct PKC-dependent MLC phosphorylation in conjunction with decreases in Ser19/Thr18 phosphorylation catalyzed by MLCK due to PMA-induced increases in PPase 1 activity. Together, these events result in stress fiber destabilization and profound actin rearrangement in bovine endothelium, which may result in the physiological alterations observed in these models.
- Published
- 2003
- Full Text
- View/download PDF
42. Interaction of filamin A with the insulin receptor alters insulin-dependent activation of the mitogen-activated protein kinase pathway.
- Author
-
He HJ, Kole S, Kwon YK, Crow MT, and Bernier M
- Subjects
- Base Sequence, Cell Line, Contractile Proteins chemistry, Contractile Proteins genetics, DNA genetics, Filamins, Humans, In Vitro Techniques, Insulin Receptor Substrate Proteins, Membrane Microdomains metabolism, Microfilament Proteins chemistry, Microfilament Proteins genetics, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Phosphoproteins metabolism, Phosphorylation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Receptor, Insulin genetics, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Subcellular Fractions metabolism, Transcriptional Activation, Transfection, ets-Domain Protein Elk-1, Contractile Proteins metabolism, DNA-Binding Proteins, Insulin metabolism, MAP Kinase Signaling System physiology, Microfilament Proteins metabolism, Receptor, Insulin metabolism, Transcription Factors
- Abstract
The biological actions of insulin are associated with a rapid reorganization of the actin cytoskeleton within cells in culture. Even though this event requires the participation of actin-binding proteins, the effect of filamin A (FLNa) on insulin-mediated signaling events is still unknown. We report here that human melanoma M2 cells lacking FLNa expression exhibited normal insulin receptor (IR) signaling, whereas FLNa-expressing A7 cells were unable to elicit insulin-dependent Shc tyrosine phosphorylation and p42/44 MAPK activation despite no significant defect in IR-stimulated phosphorylation of insulin receptor substrate-1 or activation of the phosphatidylinositol 3-kinase/AKT cascade. Insulin-dependent translocation of Shc, SOS1, and MAPK to lipid raft microdomains was markedly attenuated by FLNa expression. Coimmunoprecipitation experiments and in vitro binding assays demonstrated that FLNa binds constitutively to IR and that neither insulin nor depolymerization of actin by cytochalasin D affected this interaction. The colocalization of endogenous FLNa with IR was detected at the surface of HepG2 cells. Ectopic expression of a C-terminal fragment of FLNa (FLNaCT) in HepG2 cells blocked the endogenous IR-FLNa interaction and potentiated insulin-stimulated MAPK phosphorylation and transactivation of Elk-1 compared with vector-transfected cells. Expression of FLNaCT had no major effect on insulin-induced phosphorylation of the IR, insulin receptor substrate-1, or AKT, but it elicited changes in actin cytoskeletal structure and ruffle formation in HepG2 cells. Taken together, these results indicate that FLNa interacts constitutively with the IR to exert an inhibitory tone along the MAPK activation pathway.
- Published
- 2003
- Full Text
- View/download PDF
43. Role of CaM kinase II and ERK activation in thrombin-induced endothelial cell barrier dysfunction.
- Author
-
Borbiev T, Verin AD, Birukova A, Liu F, Crow MT, and Garcia JG
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Calmodulin-Binding Proteins metabolism, Cattle, Cell Membrane Permeability drug effects, Cell Membrane Permeability physiology, Electric Impedance, Endothelium, Vascular cytology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Myosin Light Chains metabolism, Phosphorylation, Pulmonary Artery cytology, Stress Fibers enzymology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Endothelium, Vascular enzymology, Hemostatics pharmacology, Mitogen-Activated Protein Kinases metabolism, Pulmonary Artery enzymology, Thrombin pharmacology
- Abstract
We have previously shown that thrombin-induced endothelial cell barrier dysfunction involves cytoskeletal rearrangement and contraction, and we have elucidated the important role of endothelial cell myosin light chain kinase and the actin- and myosin-binding protein caldesmon. We evaluated the contribution of calmodulin (CaM) kinase II and extracellular signal-regulated kinase (ERK) activation in thrombin-mediated bovine pulmonary artery endothelial cell contraction and barrier dysfunction. Similar to thrombin, infection with a constitutively active adenoviral alpha-CaM kinase II construct induced significant ERK activation, indicating that CaM kinase II activation lies upstream of ERK. Thrombin-induced ERK-dependent caldesmon phosphorylation (Ser789) was inhibited by either KN-93, a specific CaM kinase II inhibitor, or U0126, an inhibitor of MEK activation. Immunofluorescence microscopy studies revealed phosphocaldesmon colocalization within thrombin-induced actin stress fibers. Pretreatment with either U0126 or KN-93 attenuated thrombin-mediated cytoskeletal rearrangement and evoked declines in transendothelial electrical resistance while reversing thrombin-induced dissociation of myosin from nondenaturing caldesmon immunoprecipitates. These results strongly suggest the involvement of CaM kinase II and ERK activities in thrombin-mediated caldesmon phosphorylation and both contractile and barrier regulation.
- Published
- 2003
- Full Text
- View/download PDF
44. Central involvement of Rho family GTPases in TNF-alpha-mediated bovine pulmonary endothelial cell apoptosis.
- Author
-
Petrache I, Crow MT, Neuss M, and Garcia JG
- Subjects
- Actins metabolism, Animals, Antigens, CD metabolism, Caspases metabolism, Cattle, Cells, Cultured, Cytoskeleton metabolism, Endothelium, Vascular cytology, Enzyme Inhibitors pharmacology, Intracellular Signaling Peptides and Proteins, Myosins metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases metabolism, Pulmonary Artery cytology, Receptors, Tumor Necrosis Factor metabolism, Receptors, Tumor Necrosis Factor, Type I, rho-Associated Kinases, Apoptosis drug effects, Apoptosis physiology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Pulmonary Artery drug effects, Pulmonary Artery metabolism, Tumor Necrosis Factor-alpha pharmacology, rho GTP-Binding Proteins metabolism
- Abstract
In our recent studies, we defined a critical role for increased levels of myosin light chain (MLC) phosphorylation, a regulatory event in the interaction between actin and myosin in TNF-alpha-induced pulmonary endothelial cell actomyosin rearrangement and apoptosis. The Rho GTPase effector, Rho kinase is an important signaling effector governing levels of MLC phosphorylation which contributes to plasma membrane blebbing in several models of apoptosis. In this study, we directly assessed the role of Rho kinase in TNF-alpha-induced endothelial cell microfilament rearrangement and apoptosis. Inhibition of RhoA GTPase activity by the overexpression of dominant negative RhoA attenuates TNF-alpha-triggered stress fiber formation, consistent with Rho activation as a key event in TNF-alpha-induced cytoskeletal rearrangement. Furthermore, pharmacologic inhibition of Rho kinase as well as dominant negative RhoA overexpression dramatically reduced TNF-alpha-induced bovine endothelial apoptosis reflected by nucleosomal fragmentation as well as caspase 7, 3, and 8 activation. These results indicate that Rho kinase-dependent cytoskeletal rearrangement is critical for early apoptotic events, possibly in the assembly of the death-inducing signaling complex leading to initiator and effector caspase activation, and suggest a novel role for Rho GTPases in endothelial cell apoptosis.
- Published
- 2003
- Full Text
- View/download PDF
45. Blocking the development of postischemic cardiomyopathy with viral gene transfer of the apoptosis repressor with caspase recruitment domain.
- Author
-
Chatterjee S, Bish LT, Jayasankar V, Stewart AS, Woo YJ, Crow MT, Gardner TJ, and Sweeney HL
- Subjects
- Adenoviridae genetics, Animals, Apoptosis physiology, Cardiomyopathies etiology, Myocardial Reperfusion Injury, Rabbits, Ventricular Function, Left physiology, Ventricular Remodeling, Cardiomyopathies prevention & control, Gene Transfer Techniques, Muscle Proteins genetics, Myocardial Ischemia complications
- Abstract
Objectives: Apoptosis caused by acute ischemia and subsequent ventricular remodeling is implicated as a mediator of heart failure. This study was designed to assess the efficacy of in vivo viral gene transfer of the antiapoptotic factor apoptosis repressor with caspase recruitment domain to block apoptosis and preserve ventricular geometry and function., Methods: In a rabbit model of regional ischemia followed by reperfusion, an experimental group treated with adenovirus-apoptosis repressor with caspase recruitment domain was compared with empty vector adenovirus-null controls. Cardiac function was assessed by echocardiography and sonomicrometry of the border zone compared with the normal left ventricle. Animals were killed at 6 weeks with measurements of ventricular geometry and apoptosis., Results: Animals with the apoptosis repressor with caspase recruitment domain (ARC group) maintained higher ejection fractions at 4 and 6 weeks, and sonomicrometry demonstrated greater protection of border zone fractional shortening at 6 weeks compared with the control group. The ARC group maintained superior preservation of left ventricular geometry with less ventricular dilation and wall thinning. Finally, there was reduced apoptosis in the rabbits treated with apoptosis repressor with caspase recruitment domain compared with the controls., Conclusions: Gene transfer of apoptosis repressor with caspase recruitment domain preserves left ventricular function after ischemia. The benefit at 6 weeks is postulated to result from an apoptosis repressor with caspase recruitment domain-mediated reduction in apoptosis and ventricular remodeling. Adenovirus-apoptosis repressor with caspase recruitment domain administration offers a potential strategy after myocardial ischemia to protect the heart from late postischemic cardiomyopathy.
- Published
- 2003
- Full Text
- View/download PDF
46. Caspase-dependent cleavage of myosin light chain kinase (MLCK) is involved in TNF-alpha-mediated bovine pulmonary endothelial cell apoptosis.
- Author
-
Petrache I, Birukov K, Zaiman AL, Crow MT, Deng H, Wadgaonkar R, Romer LH, and Garcia JG
- Subjects
- Animals, Caspase 3, Caspase 8, Caspase 9, Caspases analysis, Cattle, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Myosin-Light-Chain Kinase analysis, Myosin-Light-Chain Kinase physiology, Stress Fibers chemistry, Apoptosis, Caspases metabolism, Endothelium, Vascular enzymology, Myosin-Light-Chain Kinase metabolism, Pulmonary Artery cytology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Cytoskeletal proteins are key participants in the cellular progression to apoptosis. Our previous work demonstrated the critical dependence of actomyosin rearrangement and MLC phosphorylation in TNF-alpha-induced endothelial cell apoptosis. As these events reflect the activation of the multifunctional endothelial cell (EC) MLCK isoform, we assessed the direct role of EC MLCK in the regulation of TNF-alpha-induced apoptosis. Bovine pulmonary artery endothelial cells expressing either an adenovirus encoding antisense MLCK cDNA (Ad.GFP-AS MLCK) or a dominant/negative EC MLCK construct (EC MLCK-ATPdel) resulted in marked reductions in MLCK activity and TNF-alpha-mediated apoptosis. In contrast, a constitutively active EC MLCK lacking the carboxyl-terminal autoinhibitory domains (EC MLCK-1745) markedly enhanced the apoptotic response to TNF-alpha. Immunostaining in GFP-EC MLCK-expressing cells revealed colocalization of caspase 8 and EC MLCK along actin stress fibers after TNF-alpha. TNF-alpha induced the caspase-dependent cleavage of EC MLCK-1745 in transfected endothelial cells, which was confirmed by mass spectroscopy with in vitro cleavage by caspase 3 at LKKD (D1703). The resulting MLCK fragments displayed significant calmodulin-independent kinase activity. These studies convincingly demonstrate that novel interactions between the apoptotic machinery and EC MLCK exist that regulate the endothelial contractile apparatus in TNF-alpha-induced apoptosis.
- Published
- 2003
- Full Text
- View/download PDF
47. Beta-adrenergic receptor signaling pathways mediating cell survival in cardiomyocytes: a role for PKC epsilon inhibition?
- Author
-
Crow MT
- Subjects
- Animals, Cell Survival, Humans, Mice, Myocardium cytology, Rabbits, Rats, Myocardium metabolism, Myocytes, Cardiac metabolism, Protein Kinase C metabolism, Receptors, Adrenergic, beta metabolism, Signal Transduction
- Published
- 2002
- Full Text
- View/download PDF
48. Hypoxia, BNip3 proteins, and the mitochondrial death pathway in cardiomyocytes.
- Author
-
Crow MT
- Subjects
- Animals, Caspases metabolism, Cell Hypoxia, Models, Cardiovascular, Myocardial Ischemia metabolism, Myocardial Ischemia pathology, Myocardium cytology, Proto-Oncogene Proteins c-bcl-2 metabolism, Rats, Signal Transduction, Apoptosis, Membrane Proteins physiology, Mitochondria metabolism, Myocardium metabolism, Proto-Oncogene Proteins, Tumor Suppressor Proteins
- Published
- 2002
- Full Text
- View/download PDF
49. TAT protein transduction into isolated perfused hearts: TAT-apoptosis repressor with caspase recruitment domain is cardioprotective.
- Author
-
Gustafsson AB, Sayen MR, Williams SD, Crow MT, and Gottlieb RA
- Subjects
- Animals, Apoptosis, Apoptosis Regulatory Proteins, Cardiotonic Agents metabolism, Cell Line, Creatine Kinase metabolism, Gene Products, tat chemistry, Male, Myocardial Infarction pathology, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury enzymology, Myocardial Reperfusion Injury pathology, Myocardium metabolism, Organ Culture Techniques, Perfusion, Protein Structure, Tertiary, Rats, Rats, Sprague-Dawley, Recombinant Fusion Proteins genetics, Transduction, Genetic, beta-Galactosidase genetics, Gene Products, tat genetics, Muscle Proteins genetics, Myocardial Reperfusion Injury prevention & control
- Abstract
Background: Linkage of the 11-amino-acid transduction domain of HIV TAT to a heterologous protein allows the protein to be transduced readily into cells., Methods and Results: In this study, we inserted the apoptosis repressor with caspase recruitment domain (ARC) or beta-galactosidase (beta-gal) cDNA into the pTAT-hemagglutinin bacterial expression vector to produce genetic in-frame TAT-ARC or TAT-beta-gal fusion proteins for use in cell culture and in Langendorff perfusion of adult rat hearts. TAT-beta-gal and TAT-ARC were conjugated with Texas Red and could be detected in >95% of cells. TAT-ARC was able to protect H9c2 cells against cell death mediated by hydrogen peroxide, as measured by protection against the loss of mitochondrial membrane potential and preservation of nuclear morphology. Isolated adult hearts were perfused with recombinant TAT-beta-gal or TAT-ARC (20 nmol/L) for 15 minutes and then subjected to 30 minutes of global no-flow ischemia, followed by 2 hours of reperfusion. Protein transduction was assessed by Western blotting of cell lysates and cytosolic and mitochondrial fractions and by fluorescence microscopy of Texas Red-conjugated TAT proteins. TAT-beta-gal and TAT-ARC readily transduced into perfused hearts and were homogeneously distributed. Infarct size was determined by 2,3,5-triphenyltetrazolium chloride staining, and creatine kinase release was measured. Transduction of TAT-ARC was cardioprotective when administered before global ischemia and reperfusion., Conclusions: Our results demonstrate that TAT-linked fusion protein transduction into the myocardium is feasible and that transduction of TAT-ARC is protective in cell culture and in the perfused heart.
- Published
- 2002
- Full Text
- View/download PDF
50. Phosphatidylinositol 3-kinase functionally compartmentalizes the concurrent G(s) signaling during beta2-adrenergic stimulation.
- Author
-
Jo SH, Leblais V, Wang PH, Crow MT, and Xiao RP
- Subjects
- Adrenergic beta-Agonists pharmacology, Adrenergic beta-Antagonists pharmacology, Androstadienes pharmacology, Animals, Calcium metabolism, Calcium-Binding Proteins metabolism, Cell Size drug effects, Cells, Cultured, Chromones pharmacology, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Enzyme Inhibitors pharmacology, Ethanolamines pharmacology, Heart Ventricles cytology, Heart Ventricles drug effects, Heart Ventricles metabolism, Heterotrimeric GTP-Binding Proteins metabolism, Imidazoles pharmacology, Insulin-Like Growth Factor I pharmacology, Morpholines pharmacology, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Rats, Receptors, Adrenergic, beta-2 drug effects, Serine metabolism, Wortmannin, GTP-Binding Protein alpha Subunits, Gs metabolism, Phosphatidylinositol 3-Kinases metabolism, Receptors, Adrenergic, beta-2 metabolism, Signal Transduction
- Abstract
Compartmentation of intracellular signaling pathways serves as an important mechanism conferring the specificity of G protein-coupled receptor (GPCR) signaling. In the heart, stimulation of beta2-adrenoceptor (beta2-AR), a prototypical GPCR, activates a tightly localized protein kinase A (PKA) signaling, which regulates substrates at cell surface membranes, bypassing cytosolic target proteins (eg, phospholamban). Although a concurrent activation of beta2-AR-coupled G(i) proteins has been implicated in the functional compartmentation of PKA signaling, the exact mechanism underlying the restriction of the beta2-AR-PKA pathway remains unclear. In the present study, we demonstrate that phosphatidylinositol 3-kinase (PI3K) plays an essential role in confining the beta2-AR-PKA signaling. Inhibition of PI3K with LY294002 or wortmannin enables beta2-AR-PKA signaling to reach intracellular substrates, as manifested by a robust increase in phosphorylation of phospholamban, and markedly enhances the receptor-mediated positive contractile and relaxant responses in cardiac myocytes. These potentiating effects of PI3K inhibitors are not accompanied by an increase in beta2-AR-induced cAMP formation. Blocking G(i) or Gbetagamma signaling with pertussis toxin or betaARK-ct, a peptide inhibitor of Gbetagamma, completely prevents the potentiating effects induced by PI3K inhibition, indicating that the pathway responsible for the functional compartmentation of beta2-AR-PKA signaling sequentially involves G(i), Gbetagamma, and PI3K. Thus, PI3K constitutes a key downstream event of beta2-AR-G(i) signaling, which confines and negates the concurrent beta2-AR/G(s)-mediated PKA signaling.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.