1
|
Kuruppu S, Cheng LK, Avci R, Angeli-Gordon TR, Paskaranandavadivel N. Relationship Between Intestinal Slow-waves, Spike-bursts, and Motility, as Defined Through High-resolution Electrical and Video Mapping. J Neurogastroenterol Motil 2022; 28:664-677. [PMID: 36250373 PMCID: PMC9577564 DOI: 10.5056/jnm21183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/01/2022] [Accepted: 12/17/2022] [Indexed: 11/20/2022] Open
Abstract
Background/Aims High-resolution extracellular mapping has improved our understanding of bioelectric slow-wave and spike-burst activity in the small intestine. The spatiotemporal correlation of electrophysiology and motility patterns is of critical interest to intestinal function but remains incompletely defined. Methods Intestinal jejunum segments from in vivo pigs and rabbits were exteriorized, and simultaneous high-resolution extracellular recordings and video recordings were performed. Contractions were quantified with strain fields, and the frequencies and velocities of motility patterns were calculated. The amplitudes, frequencies, and velocities of slow-wave propagation patterns and spike-bursts were quantified and visualized. In addition, the duration, size and energy of spike-burst patches were quantified. Results Slow-wave associated spike-bursts activated periodically at 10.8 ± 4.0 cycles per minute (cpm) in pigs and 10.2 ± 3.2 cpm in rabbits, while independent spike-bursts activated at a frequency of 3.2 ± 1.8 cpm. Independent spike-bursts had higher amplitude and longer duration than slow-wave associated spike-bursts (1.4 ± 0.8 mV vs 0.1 ± 0.1 mV, P < 0.001; 1.8 ± 1.4 seconds vs 0.8 ± 0.3 seconds, P < 0.001 in pigs). Spike-bursts that activated as longitudinal or circumferential patches were associated with contractions in the respective directions. Spontaneous peristaltic contractions were elicited by independent spike-bursts and travelled slower than slow-wave velocity (3.7 ± 0.5 mm/sec vs 10.1 ± 4.7 mm/sec, P = 0.007). Cyclic peristaltic contractions were driven by slow-wave associated spike-bursts and were coupled to slow-wave velocity and frequency in rabbit (14.2 ± 2.3 mm/sec vs 11.5 ± 4.6 mm/sec, P = 0.162; 11.0 ± 0.6 cpm vs 10.8 ± 0.6 cpm, P = 0.970). Conclusions Motility patterns were dictated by patterns of spike-burst patches. When spike-bursts were coupled to slow-waves, periodic motility patterns were observed, while when spike-bursts were not coupled to slow-waves, spontaneous aperiodic motility patterns were captured.
Collapse
Affiliation(s)
- Sachira Kuruppu
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Leo K Cheng
- Auckland Bioengineering Institute, University of Auckland, New Zealand
- Riddet Institute, Center of Research Excellence, New Zealand
- Department of Surgery, Vanderbilt University, Nashville, USA
| | - Recep Avci
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | | | | |
Collapse
|
2
|
Lavenus SB, Vosatka KW, Caruso AP, Ullo MF, Khan A, Logue JS. Emerin regulation of nuclear stiffness is required for fast amoeboid migration in confined environments. J Cell Sci 2022; 135:274946. [PMID: 35362531 DOI: 10.1242/jcs.259493] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/17/2022] [Indexed: 11/20/2022] Open
Abstract
When metastasizing, tumor cells must traverse environments with diverse physicochemical properties. Recently, the cell nucleus has emerged as a major regulator of the transition from mesenchymal to fast amoeboid (leader bleb-based) migration. Here, in melanoma cells, we demonstrate that increasing nuclear stiffness through elevating Lamin A, inhibits fast amoeboid migration. Importantly, nuclei may respond to force through stiffening. A key factor in this process is the inner nuclear membrane (INM) protein, emerin. Accordingly, we determined the role of emerin in regulating fast amoeboid migration. Strikingly, we found that both the up- and down-regulation of emerin results in an inhibition of fast amoeboid migration. However, when key Src phosphorylation sites were removed, up-regulation of emerin no longer inhibited fast amoeboid migration. Interestingly, in confined cells, Src activity was low, as measured by a Src biosensor. Thus, the fast amoeboid migration of melanoma cells depends on the precise calibration of emerin activity.
Collapse
Affiliation(s)
- Sandrine B Lavenus
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Karl W Vosatka
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Alexa P Caruso
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Maria F Ullo
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Ayesha Khan
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Jeremy S Logue
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| |
Collapse
|
3
|
von Horsten S, Essen LO. Conformational Change of Tetratricopeptide Repeats Region Triggers Activation of Phytochrome-Associated Protein Phosphatase 5. FRONTIERS IN PLANT SCIENCE 2021; 12:733069. [PMID: 34721460 PMCID: PMC8551457 DOI: 10.3389/fpls.2021.733069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
Phytochrome activity is not only controlled by light but also by post-translational modifications, e. g. phosphorylation. One of the phosphatases responsible for plant phytochrome dephosphorylation and thereby increased activity is the phytochrome-associated protein phosphatase 5 (PAPP5). We show that PAPP5 recognizes phospho-site mimicking mutants of phytochrome B, when being activated by arachidonic acid (AA). Addition of AA to PAPP5 decreases the α-helical content as tracked by CD-spectroscopy. These changes correspond to conformational changes of the regulatory tetratricopeptide repeats (TPR) region as shown by mapping data from hydrogen deuterium exchange mass spectrometry onto a 3.0 Å crystal structure of PAPP5. Surprisingly, parts of the linker between the TPR and PP2A domains and of the so-called C-terminal inhibitory motif exhibit reduced deuterium uptake upon AA-binding. Molecular dynamics analyses of PAPP5 complexed to a phyB phosphopeptide show that this C-terminal motif remains associated with the TPR region in the substrate bound state, suggesting that this motif merely serves for restricting the orientations of the TPR region relative to the catalytic PP2A domain. Given the high similarity to mammalian PP5 these data from a plant ortholog show that the activation mode of these PPP-type protein phosphatases is highly conserved.
Collapse
Affiliation(s)
- Silke von Horsten
- Department of Biochemistry, Faculty of Chemistry, Philipps-University, Marburg, Germany
| | - Lars-Oliver Essen
- Department of Biochemistry, Faculty of Chemistry, Philipps-University, Marburg, Germany
- Center for Synthetic Microbiology, Philipps-University, Marburg, Germany
| |
Collapse
|
4
|
Ding K, Zhang L, Fan X, Zhuo P, Feng Q, Zhang S, Guo X, Liu X. Influence of an L-type SALMFamide neuropeptide on locomotory performance and muscle physiology in the sea cucumber Apostichopus japonicus. J Exp Biol 2021; 224:272337. [PMID: 34477872 DOI: 10.1242/jeb.242566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
Neuropeptides in the SALMFamide family serve as muscle relaxants in echinoderms and may affect locomotion, as the motor behavior in sea cucumbers involves alternating contraction and extension of the body wall, which is under the control of longitudinal muscle. We evaluated the effect of an L-type SALMFamide neuropeptide (LSA) on locomotory performance of Apostichopus japonicus. We also investigated the metabolites of longitudinal muscle tissue using ultra performance liquid chromatography and quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) to assess the potential physiological mechanisms underlying the effect of LSA. The hourly distance, cumulative duration and number of steps moved significantly increased in sea cucumbers in the fourth hour after injection with LSA. Also, the treatment enhanced the mean and maximum velocity by 9.8% and 17.8%, respectively, and increased the average stride by 12.4%. Levels of 27 metabolites in longitudinal muscle changed after LSA administration, and the increased concentration of pantothenic acid, arachidonic acid and lysophosphatidylethanolamine, and the altered phosphatidylethanolamine/phosphatidylcholine ratio are potential physiological mechanisms that could explain the observed effect of LSA on locomotor behavior in A. japonicus.
Collapse
Affiliation(s)
- Kui Ding
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China
| | - Libin Zhang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,University of Chinese Academy of Sciences, 100049 Beijing, China.,Shandong Province Key Laboratory of Experimental Marine Biology, 266071 Qingdao, China
| | - Xinhao Fan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China
| | - Pengji Zhuo
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Qiming Feng
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Shuangyan Zhang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Xueying Guo
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China
| | - Xiang Liu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, 266237 Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, 266071 Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, 266071 Qingdao, China
| |
Collapse
|
5
|
Kimizuka M, Tokinaga Y, Azumaguchi R, Hamada K, Kazuma S, Yamakage M. Effects of anesthetic agents on contractions of the pregnant rat myometrium in vivo and in vitro. J Anesth 2021; 35:68-80. [PMID: 33098452 PMCID: PMC7840642 DOI: 10.1007/s00540-020-02866-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/08/2020] [Indexed: 10/31/2022]
Abstract
BACKGROUND Several anesthetic agents are used in cesarean sections for both regional and general anesthesia purposes. However, there are no data comparing the in vivo effects of propofol, sevoflurane, and dexmedetomidine on the contraction of the myometrium in pregnant rats. The aim of this study was to investigate the effect of these anesthetic agents on myometrial contraction and elucidate the underlying mechanisms. METHODS Contraction force and frequency changes in response to propofol, dexmedetomidine, or sevoflurane were evaluated in vivo and in vitro. To test the effect of arachidonic acid on myometrial contraction enhanced by dexmedetomidine, changes in myometrial contraction with dexmedetomidine after administration of indomethacin were evaluated. The amount of phosphorylated myosin phosphatase target subunit 1 (MYPT1) in the membrane fraction was expressed as a percentage of the total fraction by Western blot analysis. RESULTS This study demonstrated that dexmedetomidine enhances oxytocin-induced contraction in the myometrium of pregnant rats, whereas propofol and sevoflurane attenuate these contractions. The dexmedetomidine-induced enhancement of myometrial contraction force was abolished by the administration of indomethacin. Propofol did not affect oxytocin-induced MYPT1 phosphorylation, whereas sevoflurane attenuated oxytocin-induced MYPT1 phosphorylation. CONCLUSIONS Inhibition of myofilament calcium sensitivity may underlie the inhibition of myometrial contraction induced by sevoflurane. Arachidonic acid may play an important role in the enhancement of myometrial contraction induced by dexmedetomidine by increasing myofilament calcium sensitivity. Dexmedetomidine may be used as a sedative agent to promote uterine muscle contraction and suppress bleeding after fetal delivery.
Collapse
Affiliation(s)
- Motonobu Kimizuka
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan.
| | - Yasuyuki Tokinaga
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Ryu Azumaguchi
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Kosuke Hamada
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Satoshi Kazuma
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Michiaki Yamakage
- Department of Anesthesiology, Sapporo Medical University School of Medicine, 291, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| |
Collapse
|
6
|
Ding K, Zhang L, Fan X, Guo X, Liu X, Yang H. The Effect of Pedal Peptide-Type Neuropeptide on Locomotor Behavior and Muscle Physiology in the Sea Cucumber Apostichopus japonicus. Front Physiol 2020; 11:559348. [PMID: 33192555 PMCID: PMC7642236 DOI: 10.3389/fphys.2020.559348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Neuropeptides are endogenous active substances that are present in nervous tissues and participate in behavioral and physiological processes of the animal system. Locomotor behavior is basic to predation, escape, reproduction in animals, and neuropeptides play an important role in locomotion. In this study, the function of pedal peptide-type neuropeptide (PDP) in the process of locomotor behavior of the sea cucumber Apostichopus japonicus was evaluated. The locomotor behavior of A. japonicus was recorded by infrared camera before and after PDP administration, and muscle physiology was studied by ultra performance liquid chromatography and quadrupole time-off-light mass spectrometry (UPLC-Q-TOF-MS) to clarify the potential physiological mechanisms. The results showed that PDP enhanced the cumulative duration of moving significantly at the 7th h after injection, and reduced the mean and maximum velocity by 16.90 and 14.22% in A. japonicus. The data of muscle metabolomics suggested that some significantly changed metabolites were related to locomotor behavior of sea cucumbers. The decreases of phosphatidylethanolamine (PE) and phosphatidylcholine (PC) might result in the increases of lysophosphatidylcholines (lysoPC) and lysophosphatidylethanolamine (lysoPE), and suggested the change of fluidity and permeability in the muscle cell membrane, which would affect the physiology and function of muscle cells, and finally alter the locomotor behavior. In addition, the increased level of arachidonic acid (ARA) might activate K+ ion channels and then affect the signaling of muscle cells, or promote the sensitivity of muscle cells to Ca2+ and then result in the contractility of longitudinal muscles in sea cucumbers. ARA was also involved in the linoleic acid metabolism which was the only pathway that disturbed significantly after PDP administration. In conclusion, PDP participated in the regulation of locomotor behavior in the sea cucumber, and the decreased PE and PC, increased lysoPC, lysoPE and ARA might be the potential physiological mechanisms that responsible for behavioral effects of PDP in A. japonicus.
Collapse
Affiliation(s)
- Kui Ding
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Libin Zhang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.,CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Satellite Ocean Environment Dynamics, Second Institute of Oceanography, Ministry of Natural Resources, Hangzhou, China
| | - Xinhao Fan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Xueying Guo
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Xiang Liu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Hongsheng Yang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
7
|
Lomakin AJ, Cattin CJ, Cuvelier D, Alraies Z, Molina M, Nader GPF, Srivastava N, Sáez PJ, Garcia-Arcos JM, Zhitnyak IY, Bhargava A, Driscoll MK, Welf ES, Fiolka R, Petrie RJ, De Silva NS, González-Granado JM, Manel N, Lennon-Duménil AM, Müller DJ, Piel M. The nucleus acts as a ruler tailoring cell responses to spatial constraints. Science 2020; 370:eaba2894. [PMID: 33060332 PMCID: PMC8059074 DOI: 10.1126/science.aba2894] [Citation(s) in RCA: 321] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/29/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
The microscopic environment inside a metazoan organism is highly crowded. Whether individual cells can tailor their behavior to the limited space remains unclear. In this study, we found that cells measure the degree of spatial confinement by using their largest and stiffest organelle, the nucleus. Cell confinement below a resting nucleus size deforms the nucleus, which expands and stretches its envelope. This activates signaling to the actomyosin cortex via nuclear envelope stretch-sensitive proteins, up-regulating cell contractility. We established that the tailored contractile response constitutes a nuclear ruler-based signaling pathway involved in migratory cell behaviors. Cells rely on the nuclear ruler to modulate the motive force that enables their passage through restrictive pores in complex three-dimensional environments, a process relevant to cancer cell invasion, immune responses, and embryonic development.
Collapse
Affiliation(s)
- A J Lomakin
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences (ÖAW), Vienna, Austria
- Medical University of Vienna (MUV), Vienna, Austria
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London, UK
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - C J Cattin
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - D Cuvelier
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Z Alraies
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - M Molina
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London, UK
| | - G P F Nader
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - N Srivastava
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - P J Sáez
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - J M Garcia-Arcos
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - I Y Zhitnyak
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- N.N. Blokhin Medical Research Center of Oncology, Moscow, Russia
| | - A Bhargava
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - M K Driscoll
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - E S Welf
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R Fiolka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R J Petrie
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - N S De Silva
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - J M González-Granado
- LamImSys Lab, Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - N Manel
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | | | - D J Müller
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
| | - M Piel
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France.
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| |
Collapse
|
8
|
Gao X, Luo X, You W, Ke C. Circadian movement behaviours and metabolism differences of the Pacific abalone Haliotis discus hannai. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 211:111994. [PMID: 32858337 DOI: 10.1016/j.jphotobiol.2020.111994] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/21/2020] [Accepted: 08/09/2020] [Indexed: 11/28/2022]
Abstract
Circadian rhythm is the most important and universal biological rhythm in marine organisms. In this research, the movement behaviour of abalone (Haliotis discus hannai) was continuously monitored under a light cycle of 12 L:12D. It was found that the cumulative movement distance and cumulative movement time of abalone reached was highest from 00:00-03:00 h. The minimum values of maximum movement velocity occurred between 21:00-00:00 h, and a significant circadian cosine rhythm was exhibited during these periods (P < 0.05). Metabolomic analysis of cerebral ganglions of abalone was conducted at 06:00 h (6 M), 14:00 h (14 M), and 22:00 h (22 M) and 380, 385, and 315 metabolites with significant differences were identified in 6 M vs 14 M, 14 M vs 22 M, and 6 M vs 22 M, respectively (P < 0.05). With the alternation of day and night, the expression levels of phosphatidylcholine, 5-HT, N-acetyl-5-hydroxytryptamine, indole-3-acetaldehyde, hypoxanthine, and deoxyinosine declined significantly, while those of Lysophosphatidylcholines (lysoPC) (20: 5 (5Z, 8Z, 11Z, 14Z, 17Z)), lysoPC (22: 4 (7Z, 10Z, 13Z, 16Z)), lysoPC (16: 1 (9Z) / 0: 0), phosphatidylethanolamine (PE) (18: 1 (11Z) 22: 2 (13Z, 16Z)), and guanosine 5'-phosphate rose significantly. These 11 metabolites can be used as differential metabolic markers. These findings not only quantitatively describe the circadian movement behaviours of abalone, but also provide an initial analysis of the circadian mechanism of the physiological metabolic conversion of abalone, which in turn provides guidelines for light control and feeding strategy for use in aquaculture production.
Collapse
Affiliation(s)
- Xiaolong Gao
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen 361102, China; College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Xuan Luo
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen 361102, China; College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Weiwei You
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen 361102, China; College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.
| | - Caihuan Ke
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China; Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen 361102, China; College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
9
|
Álvarez-Santos MD, Álvarez-González M, Estrada-Soto S, Bazán-Perkins B. Regulation of Myosin Light-Chain Phosphatase Activity to Generate Airway Smooth Muscle Hypercontractility. Front Physiol 2020; 11:701. [PMID: 32676037 PMCID: PMC7333668 DOI: 10.3389/fphys.2020.00701] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Smooth muscle is a central structure involved in the regulation of airway tone. In addition, it plays an important role in the development of some pathologies generated by alterations in contraction, such as hypercontractility and the airway hyperresponsiveness observed in asthma. The molecular processes associated with smooth muscle contraction are centered around myosin light chain (MLC) phosphorylation, which is controlled by a balance in the activity of myosin light-chain kinase (MLCK) and myosin light-chain phosphatase (MLCP). MLCK activation depends on increasing concentrations of intracellular Ca2+, while MLCP activation is independent of Ca2+. MLCP contains a phosphatase subunit (PP1c) that is regulated through myosin phosphatase target subunit 1 (MYPT1) and other subunits, such as glycogen-associated regulatory subunit and myosin-binding subunit 85 kDa. Interestingly, MLCP inhibition may contribute to exacerbation of smooth muscle contraction by increasing MLC phosphorylation to induce hypercontractility. Many pathways inhibiting MLCP activity in airway smooth muscle have been proposed and are focused on inhibition of PP1c, inhibitory phosphorylation of MYPT1 and dissociation of the PP1c-MYPT1 complex.
Collapse
Affiliation(s)
- Mayra D Álvarez-Santos
- Biology Area, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marisol Álvarez-González
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Blanca Bazán-Perkins
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.,Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
10
|
Liu Z, Khalil RA. Evolving mechanisms of vascular smooth muscle contraction highlight key targets in vascular disease. Biochem Pharmacol 2018; 153:91-122. [PMID: 29452094 PMCID: PMC5959760 DOI: 10.1016/j.bcp.2018.02.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/12/2018] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle (VSM) plays an important role in the regulation of vascular function. Identifying the mechanisms of VSM contraction has been a major research goal in order to determine the causes of vascular dysfunction and exaggerated vasoconstriction in vascular disease. Major discoveries over several decades have helped to better understand the mechanisms of VSM contraction. Ca2+ has been established as a major regulator of VSM contraction, and its sources, cytosolic levels, homeostatic mechanisms and subcellular distribution have been defined. Biochemical studies have also suggested that stimulation of Gq protein-coupled membrane receptors activates phospholipase C and promotes the hydrolysis of membrane phospholipids into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 stimulates initial Ca2+ release from the sarcoplasmic reticulum, and is buttressed by Ca2+ influx through voltage-dependent, receptor-operated, transient receptor potential and store-operated channels. In order to prevent large increases in cytosolic Ca2+ concentration ([Ca2+]c), Ca2+ removal mechanisms promote Ca2+ extrusion via the plasmalemmal Ca2+ pump and Na+/Ca2+ exchanger, and Ca2+ uptake by the sarcoplasmic reticulum and mitochondria, and the coordinated activities of these Ca2+ handling mechanisms help to create subplasmalemmal Ca2+ domains. Threshold increases in [Ca2+]c form a Ca2+-calmodulin complex, which activates myosin light chain (MLC) kinase, and causes MLC phosphorylation, actin-myosin interaction, and VSM contraction. Dissociations in the relationships between [Ca2+]c, MLC phosphorylation, and force have suggested additional Ca2+ sensitization mechanisms. DAG activates protein kinase C (PKC) isoforms, which directly or indirectly via mitogen-activated protein kinase phosphorylate the actin-binding proteins calponin and caldesmon and thereby enhance the myofilaments force sensitivity to Ca2+. PKC-mediated phosphorylation of PKC-potentiated phosphatase inhibitor protein-17 (CPI-17), and RhoA-mediated activation of Rho-kinase (ROCK) inhibit MLC phosphatase and in turn increase MLC phosphorylation and VSM contraction. Abnormalities in the Ca2+ handling mechanisms and PKC and ROCK activity have been associated with vascular dysfunction in multiple vascular disorders. Modulators of [Ca2+]c, PKC and ROCK activity could be useful in mitigating the increased vasoconstriction associated with vascular disease.
Collapse
Affiliation(s)
- Zhongwei Liu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Anjum I, Denizalti M, Kandilci HB, Durlu-Kandilci NT, Sahin-Erdemli I. Enhancement of S1P-induced contractile response in detrusor smooth muscle of rats having cystitis. Eur J Pharmacol 2017; 814:343-351. [DOI: 10.1016/j.ejphar.2017.08.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 11/26/2022]
|
12
|
Irvine NA, Lillycrop KA, Fielding B, Torrens C, Hanson MA, Burdge GC. Polyunsaturated fatty acid biosynthesis is involved in phenylephrine-mediated calcium release in vascular smooth muscle cells. Prostaglandins Leukot Essent Fatty Acids 2015; 101:31-9. [PMID: 26324193 DOI: 10.1016/j.plefa.2015.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/23/2015] [Accepted: 08/11/2015] [Indexed: 10/24/2022]
Abstract
Stimulation of vascular smooth muscle (VSM) α1-adrenoceptors induces myosin phosphorylation and vasoconstriction via mobilisation of intracellular calcium and production of specific eicosanoids. Polyunsaturated fatty acid (PUFA) biosynthesis in VSM cells is involved, although the precise mechanism is not known. To address this, we characterised PUFA biosynthesis in VSM cells and determined its role in intracellular calcium release and eicosanoid production. Murine VSM cells converted 18:2n-6 to longer chain PUFA including 22:5n-6. Δ6 (D6d) and Δ5 (D5d) desaturase, and elongase (Elovl) 5 were expressed. Elovl2 was not detected in human, mouse or rat VSM cells, or in rat or mouse aortae, but tit was not associated with hypermethylation of its promoter. D6d or D5d inhibition reduced 18:3n-6 and 20:4n-6 synthesis, respectively, and induced concentration-related decrease in phenylephrine-mediated calcium release, and in PGE2 and PGF2α secretion. Together these findings suggest that PUFA biosynthesis in VSM cells is involved in calcium release associated with vasoconstriction.
Collapse
Affiliation(s)
- Nicola A Irvine
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Barbara Fielding
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christopher Torrens
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mark A Hanson
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
13
|
Abstract
MLCP (myosin light chain phosphatase) regulates platelet function through its ability to control myosin IIa phosphorylation. Recent evidence suggests that MLCP is a de facto target for signalling events stimulated by cAMP. In the present mini-review, we discuss the mechanisms by which cAMP signalling maintains MLCP in an active state to control platelet contractile machinery.
Collapse
|
14
|
A novel trigger for cholesterol-dependent smooth muscle contraction mediated by the sphingosylphosphorylcholine-Rho-kinase pathway in the rat basilar artery: a mechanistic role for lipid rafts. J Cereb Blood Flow Metab 2015; 35:835-42. [PMID: 25605290 PMCID: PMC4420858 DOI: 10.1038/jcbfm.2014.260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 02/04/2023]
Abstract
Hyperlipidemia is a risk factor for abnormal cerebrovascular events. Rafts are cholesterol-enriched membrane microdomains that influence signal transduction. We previously showed that Rho-kinase-mediated Ca(2+) sensitization of vascular smooth muscle (VSM) induced by sphingosylphosphorylcholine (SPC) has a pivotal role in cerebral vasospasm. The goals of the study were to show SPC-Rho-kinase-mediated VSM contraction in vivo and to link this effect to cholesterol and rafts. The SPC-induced VSM contraction measured using a cranial window model was reversed by Y-27632, a Rho-kinase inhibitor, in rats fed a control diet. The extent of SPC-induced contraction correlated with serum total cholesterol. Total cholesterol levels in the internal carotid artery (ICA) were significantly higher in rats fed a cholesterol diet compared with a control diet or a β-cyclodextrin diet, which depletes VSM cholesterol. Western blotting and real-time PCR revealed increases in flotillin-1, a raft marker, and flotillin-1 mRNA in the ICA in rats fed a cholesterol diet, but not in rats fed the β-cyclodextrin diet. Depletion of cholesterol decreased rafts in VSM cells, and prevention of an increase in cholesterol by β-cyclodextrin inhibited SPC-induced contraction in a cranial window model. These results indicate that cholesterol potentiates SPC-Rho-kinase-mediated contractions of importance in cerebral vasospasm and are compatible with a role for rafts in this process.
Collapse
|
15
|
Ihara E, Yu Q, Chappellaz M, MacDonald JA. ERK and p38MAPK pathways regulate myosin light chain phosphatase and contribute to Ca2+ sensitization of intestinal smooth muscle contraction. Neurogastroenterol Motil 2015; 27:135-46. [PMID: 25557225 DOI: 10.1111/nmo.12491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/20/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated protein kinase (ERK) and p38MAPK, are known regulators of smooth muscle contractility. The contraction of smooth muscle is mainly regulated by the phosphorylation of regulatory light chains of myosin II (LC20), which is driven by the balance between myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We hypothesized that one possible mechanism for MAPK-dependent modulation of intestinal smooth muscle contractility is via the regulation of MLCP activity. METHODS Contractile responses to carbachol (CCh) and effects of MAPK inhibitors on CCh-induced contractions were assessed with isolated rat ileal longitudinal smooth muscle strips. Biochemical assessments of MLCP activity and myosin phosphatse targeting subunit (MYPT1) and CPI-17 phosphorylations were completed. KEY RESULTS Treatment of ileal smooth muscle with PD98059 (10 μM; MEK inhibitor) or SB203580 (10 μM; p38MAPK inhibitor) significantly inhibited CCh-induced contractile force. Decreased MLCP activity was observed during sustained contractions induced by CCh; the MLCP activity was recovered by treatment with PD98059 and SB203580. However, MYPT1 (Thr697 and Thr855) and CPI-17 (Thr38) phosphorylations were not affected. Application of ML-7 (MLCK inhibitor) during CCh-induced sustained contraction elicited an MLCP-dependent relaxation, the rate of which was accelerated by application of PD98059 and SB203580 with proportional changes in LC20 phosphorylation levels but not MYPT1 phosphorylation (Thr697 or Thr855). CONCLUSIONS & INFERENCES ERK and p38MAPK contribute to CCh-induced sustained contraction in a LC20 phosphorylation dependent manner. Moreover, both kinases inhibit MLCP activity possibly by a novel mechanism.
Collapse
Affiliation(s)
- E Ihara
- Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta, Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada; Department of Medicine and Bioregulatory Science, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | |
Collapse
|
16
|
Khasnis M, Nakatomi A, Gumpper K, Eto M. Reconstituted human myosin light chain phosphatase reveals distinct roles of two inhibitory phosphorylation sites of the regulatory subunit, MYPT1. Biochemistry 2014; 53:2701-9. [PMID: 24712327 PMCID: PMC4010256 DOI: 10.1021/bi5001728] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The myosin light chain phosphatase
(MLCP) is a cytoskeleton-associated
protein phosphatase-1 (PP1) holoenzyme and a RhoA/ROCK effector, regulating
cytoskeletal reorganization. ROCK-induced phosphorylation of the MLCP
regulatory subunit (MYPT1) at two sites, Thr696 and Thr853, suppresses
the activity, although little is known about the difference in the
role. Here, we developed a new method for the preparation of the recombinant
human MLCP complex and determined the molecular and cellular basis
of inhibitory phosphorylation. The recombinant MLCP partially purified
from mammalian cell lysates retained characteristics of the native
enzyme, such that it was fully active without Mn2+ and
sensitive to PP1 inhibitor compounds. Selective thio-phosphorylation
of MYPT1 at Thr696 with ROCK inhibited the MLCP activity 30%, whereas
the Thr853 thio-phosphorylation did not alter the phosphatase activity.
Interference with the docking of phospho-Thr696 at the active site
weakened the inhibition, suggesting selective autoinhibition induced
by phospho-Thr696. Both Thr696 and Thr853 sites underwent autodephosphorylation.
Compared with that of Thr853, phosphorylation of Thr696 was more stable,
and it facilitated Thr853 phosphorylation. Endogenous MYPT1 at Thr696
was spontaneously phosphorylated in quiescent human leiomyosarcoma
cells. Serum stimulation of the cells resulted in dissociation of
MYPT1 from myosin and PP1C in parallel with an increase in the level
of Thr853 phosphorylation. The C-terminal domain of human MYPT1(495–1030)
was responsible for the binding to the N-terminal portion of myosin
light meromyosin. The spontaneous phosphorylation at Thr696 may adjust
the basal activity of cellular MLCP and affect the temporal phosphorylation
at Thr853 that is synchronized with myosin targeting.
Collapse
Affiliation(s)
- Mukta Khasnis
- Department of Molecular Physiology and Biophysics, Thomas Jefferson University Jefferson Medical School , and Kimmel Cancer Center , 1020 Locust Street, Philadelphia, Pennsylvania 19107, United States
| | | | | | | |
Collapse
|
17
|
Floyd CN, Ferro A. The platelet fibrinogen receptor: from megakaryocyte to the mortuary. JRSM Cardiovasc Dis 2012; 1:10.1258_cvd.2012.012007. [PMID: 24175064 PMCID: PMC3738324 DOI: 10.1258/cvd.2012.012007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Platelets are integral to normal haemostatic function and act to control vascular haemorrhage with the formation of a stable clot. The fibrinogen receptor (glycoprotein IIb/IIIa [GPIIb/IIIa]) is the most abundant platelet integrin and, by binding fibrinogen, facilitates irreversible binding of platelets to the exposed extracellular matrix and enables the cross-linking of adjacent platelets. The vital role of GPIIb/IIIa requires tight control of both its synthesis and function. After transcription from distinct domains on chromosome 17, the two subunits of the heterodimer are carefully directed through organelles with intricate regulatory steps designed to prevent the cellular expression of a dysfunctional receptor. Similarly, exquisite control of platelet activation via bidirectional signalling acts to limit the inappropriate and excessive formation of platelet-mediated thrombus. However, the enormous diversity of genetic mutations in the fibrinogen receptor has resulted in a number of allelic variants becoming established. The Pro33 polymorphism in GPIIIa is associated with increased cardiovascular risk due to a pathological persistence of outside-in signalling once fibrinogen has dissociated from the receptor. The polymorphism has also been associated with the phenomenon of aspirin resistance, although larger epidemiological studies are required to establish this conclusively. A failure of appropriate receptor function due to a diverse range of mutations in both structural and signalling domains, results in the bleeding diathesis Glanzmann's thrombasthaenia. GPIIb/IIIa inhibitors were the first rationally designed anti-platelet drugs and have proven to be a successful therapeutic option in high-risk primary coronary intervention. As our understanding of bidirectional signalling improves, more subtle and directed therapeutic strategies may be developed.
Collapse
Affiliation(s)
- Christopher N Floyd
- Department of Clinical Pharmacology, Cardiovascular Division, King's College London , London SE1 9NH , UK
| | | |
Collapse
|
18
|
Abstract
Myometrial smooth muscle contractility is regulated predominantly through the reversible phosphorylation of MYLs (myosin light chains), catalysed by MYLK (MYL kinase) and MYLP (MYL phosphatase) activities. MYLK is activated by Ca2+-calmodulin, and most uterotonic agonists operate through myometrial receptors that increase [Ca2+]i (intracellular Ca2+ concentration). Moreover, there is substantial evidence for Ca2+-independent inhibition of MYLP in smooth muscle, leading to generation of increased MYL phosphorylation and force for a given [Ca2+]i, a phenomenon known as 'Ca2+-sensitization'. ROCK (Rho-associated kinase)-mediated phosphorylation and inhibition of MYLP has been proposed as a mechanism for Ca2+-sensitization in smooth muscle. However, it is unclear to date whether the mechanisms that sensitize the contractile machinery to Ca2+ are important in the myometrium, as they appear to be in vascular and respiratory smooth muscle. In the present paper, we discuss the signalling pathways regulating MYLP activity and the involvement of ROCK in myometrial contractility, and present recent data from our laboratory which support a role for Ca2+-sensitization in human myometrium.
Collapse
|
19
|
Pattabiraman PP, Lih FB, Tomer KB, Rao PV. The role of calcium-independent phospholipase A2γ in modulation of aqueous humor drainage and Ca2+ sensitization of trabecular meshwork contraction. Am J Physiol Cell Physiol 2012; 302:C979-91. [PMID: 22237407 PMCID: PMC3330734 DOI: 10.1152/ajpcell.00396.2011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 01/06/2012] [Indexed: 11/22/2022]
Abstract
The contractile and relaxation characteristics of trabecular meshwork (TM) are presumed to influence aqueous humor (AH) drainage and intraocular pressure. The mechanisms underlying regulation of TM cell contractile properties, however, are not well understood. This study investigates the role of calcium-independent phospholipase A(2) (iPLA(2)), which controls eicosanoid synthesis, in regulation of TM cell contraction and AH outflow using mechanism-based isoform specific inhibitors (R)-bromoenol lactone (R-BEL, iPLA(2)γ specific) and (S)-bromoenol lactone (S-BEL, iPLA(2)β specific). Immunohistochemical analysis revealed intense staining for both iPLA(2)β and γ isoforms throughout the TM, juxtacanalicular tissue, and Schlemm's canal of human eye. Inhibition of iPLA(2)γ by R-BEL or small interfering RNA-mediated silencing of iPLA(2)γ expression induced dramatic changes in TM cell morphology, and decreased actin stress fibers, focal adhesions, and myosin light-chain (MLC) phosphorylation. AH outflow facility increased progressively and significantly in enucleated porcine eyes perfused with R-BEL. This response was associated with a significant decrease in TM tissue MLC phosphorylation and alterations in the morphology of aqueous plexi in R-BEL-perfused eyes. In contrast, S-BEL did not affect either of these parameters. Additionally, R-BEL-induced cellular relaxation of the TM was associated with a significant decrease in the levels of active Rho GTPase, phospho-MLC phosphatase, phospho-CPI-17, and arachidonic acid. Taken together, these observations demonstrate that iPLA(2)γ plays a significant and isoform-specific role in regulation of AH outflow facility by altering the contractile characteristics of the TM. The effects of iPLA(2)γ on TM contractile status appear to involve arachidonic acid and Rho GTPase signaling pathways.
Collapse
|
20
|
Lee JH, Palaia T, Ragolia L. Impaired insulin-stimulated myosin phosphatase Rho-interacting protein signaling in diabetic Goto-Kakizaki vascular smooth muscle cells. Am J Physiol Cell Physiol 2012; 302:C1371-81. [PMID: 22322972 DOI: 10.1152/ajpcell.00254.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance associated with Type 2 diabetes contributes to impaired vasorelaxation and therefore contributes to the enhanced incidence of hypertension observed in diabetes. In this study, we examined the role of insulin on the association of the myosin-binding subunit of myosin phosphatase (MYPT1) to myosin phosphatase Rho-interacting protein (MRIP), a relatively novel member of the myosin phosphatase complex that directly binds RhoA in vascular smooth muscle cells (VSMCs). Through a series of molecular and cellular studies, we investigated whether insulin stimulates the binding of MRIP to MYPT1 and compared the results generated from VSMCs isolated from both Wistar-Kyoto (WKY) control and Goto-Kakizaki (GK) diabetic rats. We demonstrate for the first time that insulin stimulates the binding of MRIP to MYPT1 in a dose- and time-dependent manner, as determined by immunoprecipitation, implying a regulatory role for MRIP in insulin-induced vasodilation signaling via MYPT1 interaction. VSMCs from GK model of Type 2 diabetes had impaired insulin-induced MRIP/MYPT1 binding as well as reduced MRIP expression. Adenovirus-mediated overexpression of MRIP in GK VSMCs led to significantly improved insulin-stimulated MRIP/MYPT1 binding. Finally, insulin-stimulated MRIP translocation out of stress fibers, which was observed in control VSMCs, was impaired in GK VSMCs. We believe the impaired expression of MRIP, and therefore decreased insulin-stimulated MRIP/MYPT1 association, in the GK diabetic model may contribute to the impaired insulin-mediated vasodilation observed in the diabetic vasculature and provides a novel therapeutic strategy for the treatment of Type 2 diabetes.
Collapse
Affiliation(s)
- Jin Hee Lee
- Vascular Biology Institute, Winthrop-University Hospital, Mineola, New York 11501, USA
| | | | | |
Collapse
|
21
|
Ruiz-Loredo AY, López-Colomé AM. New insights into the regulation of myosin light chain phosphorylation in retinal pigment epithelial cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 293:85-121. [PMID: 22251559 DOI: 10.1016/b978-0-12-394304-0.00008-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The retinal pigment epithelium (RPE) plays an essential role in the function of the neural retina and the maintenance of vision. Most of the functions displayed by RPE require a dynamic organization of the acto-myosin cytoskeleton. Myosin II, a main cytoskeletal component in muscle and non-muscle cells, is directly involved in force generation required for organelle movement, selective molecule transport within cell compartments, exocytosis, endocytosis, phagocytosis, and cell division, among others. Contractile processes are triggered by the phosphorylation of myosin II light chains (MLCs), which promotes actin-myosin interaction and the assembly of contractile fibers. Considerable evidence indicates that non-muscle myosin II activation is critically involved in various pathological states, increasing the interest in studying the signaling pathways controlling MLC phosphorylation. Particularly, recent findings suggest a role for non-muscle myosin II-induced contraction in RPE cell transformation involved in the establishment of numerous retinal diseases. This review summarizes the current knowledge regarding myosin function in RPE cells, as well as the signaling networks leading to MLC phosphorylation under pathological conditions. Understanding the molecular mechanisms underlying RPE dysfunction would improve the development of new therapies for the treatment or prevention of different ocular disorders leading to blindness.
Collapse
Affiliation(s)
- Ariadna Yolanda Ruiz-Loredo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico DF, Mexico
| | | |
Collapse
|
22
|
Cheng J, Zhang XM, Shen ZY, Chen WH, Cai WJ, Ying J, Hu GR, Liu RH. [Immunoregulatory mechanisms of an optimal Chinese herbal monomer compound in mice with allergic rhinitis]. ACTA ACUST UNITED AC 2011; 9:1360-6. [PMID: 22152776 DOI: 10.3736/jcim20111213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To study the immunoregulatory effect of an optimal Chinese herbal monomer compound, which consists of three monomers, namely, icariin, baicalin and Astragalus saponin I, in a mouse model of allergic rhinitis. METHODS A mouse model of allergic rhinitis was established by intraperitoneal injection of ovalbumin and aluminum hydroxide gel suspension. The splenic lymphocytes of the mice were separated, cultured in 96-well plates and divided into three groups: control group, concanavalin A group and compound group. Splenic lymphocyte proliferation was detected by cell counting kit-8 method at different time points. Cell cycle distribution was observed by flow cytometry (FCM) also at different time points. The changes of intracellular calcium concentration of splenic lymphocytes were measured by fluorescence microplate reader after the cells were incubated with fluorescence probe Fluo-3/AM. RESULTS The Chinese herbal monomer compound could inhibit cell proliferation induced by concanavalin A (P<0.01). And the inhibition presented a time-effect relationship. With extending of the action time, the inhibition rate gradually increased and reached peak at the 48th hour. FCM test revealed the fact that concanavalin A could promote cells to enter into the mitosis by reducing the percentage of cells in G0/G1 phases while increasing the percentage of cells in S and G(2)/M phases. Compared with the concanavalin A, the compound could increase the percentage of cells in G(0)/G(1) phases and at the same time reduce the percentage of cells in S and G(2)/M phases at different time points, with the effect most significant at the 24th hour (P<0.05 or P<0.01). The results of the test taken by the fluorescence microplate reader revealed that the fluorescence value of the concanavalin A group increased with time in the previous 24 h while the compound could reduce this trend obviously, thus reduce the intracellular calcium concentration (P<0.01). CONCLUSION The Chinese herbal monomer compound can inhibit the proliferation of cultured splenic lymphocytes of mice with allergic rhinitis. The effects of the compound of lowering intracellular calcium concentration and arresting cell cycle at G(0)/G(1) phases from entering into S and G(2)/M phases are responsible for its antiproliferation activity.
Collapse
Affiliation(s)
- Jian Cheng
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
d'Emmanuele di Villa Bianca R, Sorrentino R, Coletta C, Mitidieri E, Rossi A, Vellecco V, Pinto A, Cirino G, Sorrentino R. Hydrogen sulfide-induced dual vascular effect involves arachidonic acid cascade in rat mesenteric arterial bed. J Pharmacol Exp Ther 2011; 337:59-64. [PMID: 21228064 DOI: 10.1124/jpet.110.176016] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hydrogen sulfide (H(2)S), a novel gaseous transmitter, is considered a physiological regulator of vascular homeostasis. Recent evidence suggests H(2)S as an endothelium-hyperpolarizing factor (EDHF) candidate. To address this issue, we evaluated the vascular effect of sodium hydrogen sulfide (NaHS), an H(2)S donor, on the rat mesenteric arterial bed. NaHS concentration-response curve was performed on preconstricted mesenteric arterial bed. To assess the contribution of EDHF, we performed a pharmacologic dissection using indomethacin, N(G)-nitro-l-arginine methyl ester (l-NAME), or apamin and charybdotoxin as cyclooxygenase, nitric-oxide synthase, and calcium-dependent potassium channel inhibitors, respectively. In another set of experiments, we used 4-(4-octadecylphenyl)-4-oxobutenoic acid, baicalein, or proadifen as phospholipase A(2) (PLA(2)), lipoxygenase, and cytochrome P450 inhibitors, respectively. Finally, an immunofluorescence study was performed to support the involvement of PLA(2) in mesenteric artery challenged by NaHS. NaHS promoted a dual vascular effect (i.e., vasoconstriction and vasodilation). l-NAME or baicalein administration affected neither NaHS-mediated vasodilation nor vasoconstriction, whereas apamin and charybdotoxin significantly inhibited NaHS-induced relaxation. Pretreatment with PLA(2) inhibitor abolished both the contracting and the relaxant effect, whereas P450 cytochrome blocker significantly reduced NaHS-mediated relaxation. The immunofluorescence study showed that NaHS caused a migration of cytosolic PLA(2) close to the nucleus, which implicates activation of this enzyme. Our data indicate that H(2)S could activate PLA(2), which in turn releases arachidonic acid leading, initially, to vasoconstriction followed by vasodilation mediated by cytochrome P450-derived metabolites. Because EDHF has been presumed to be a cytochrome P450 derivative of the arachidonic acid, our results suggest that H(2)S acts through EHDF release.
Collapse
|
24
|
RNAi-mediated knockdown of protein kinase C-alpha inhibits cell migration in MM-RU human metastatic melanoma cell line. Melanoma Res 2010; 20:171-8. [PMID: 20216103 DOI: 10.1097/cmr.0b013e32832f1581] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Protein kinase C (PKC) is a multigene family of serine/threonine protein kinases involved in cell signaling pathways of proliferation and motility. PKC interacts with Rho GTPases in the regulation of the actin cytoskeleton. The PKC-alpha isozyme binds the Rho GTPase cdc42, and both are coordinated with the Rac-phosphatidylinositol-3 kinase (PI3K) signaling pathway in melanoma cell invasion and migration on extracellular matrix proteins. To further define the role of PKC-alpha in melanoma cell migration, we tested the effect of PDBu and Ca dependent activation of PKC-alpha as well as treatment with the PKC-alpha inhibitors calphostin C and Go6976. Furthermore, we transfected siRNA targeted against PKC-alpha into human melanoma cells and performed time-lapse analysis of cell migration followed by western immunoblotting. We found that significant enhancement of cell migration at 0.5 h after PDBu treatment directly correlated with Ca dependent activation of PKC-alpha and was inhibited by the PKC-alpha inhibitor calphostin C. PKC-alpha siRNA transfection nearly abrogated PKC-alpha expression and significantly reduced melanoma cell migration compared with siRNA controls. These findings provide further evidence that PKC-alpha plays an important role in melanoma cell migration and may have implications in therapies designed to disrupt melanoma cell motility by alteration of PKC-alpha signaling.
Collapse
|
25
|
Katayama T, Watanabe M, Tanaka H, Hino M, Miyakawa T, Ohki T, Ye LH, Xie C, Yoshiyama S, Nakamura A, Ishikawa R, Tanokura M, Oiwa K, Kohama K. Stimulatory effects of arachidonic acid on myosin ATPase activity and contraction of smooth muscle via myosin motor domain. Am J Physiol Heart Circ Physiol 2010; 298:H505-14. [DOI: 10.1152/ajpheart.00577.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have been searching for a mechanism to induce smooth muscle contraction that is not associated with phosphorylation of the regulatory light chain (RLC) of smooth muscle myosin (Nakamura A, Xie C, Zhang Y, Gao Y, Wang HH, Ye LH, Kishi H, Okagaki T, Yoshiyama S, Hayakawa K, Ishikawa R, Kohama K. Biochem Biophys Res Commun 369: 135–143, 2008). In this article, we report that arachidonic acid (AA) stimulates ATPase activity of unphosphorylated smooth muscle myosin with maximal stimulation (Rmax) of 6.84 ± 0.51 relative to stimulation by the vehicle and with a half-maximal effective concentration (EC50) of 50.3 ± 4.2 μM. In the presence of actin, Rmax was 1.72 ± 0.08 and EC50 was 26.3 ± 2.3 μM. Our experiments with eicosanoids consisting of the AA cascade suggested that they neither stimulated nor inhibited the activity. Under conditions that did not allow RLC to be phosphorylated, AA stimulated contraction of smooth muscle tissue with an Rmax of 1.45 ± 0.07 and an EC50 of 27.0 ± 4.4 μM. In addition to the ATPase activities of the myosin, AA stimulated those of heavy meromyosin, subfragment 1 (S1), S1 from which the RLC was removed, and a recombinant heavy chain consisting of the myosin head. The stimulatory effects of AA on these preparations were about twofold. The site of AA action was indicated to be the step-releasing inorganic phosphate (Pi) from the reaction intermediate of the myosin-ADP-Pi complex. The enhancement of Pi release by AA was supported by computer simulation indicating that AA docked in the actin-binding cleft of the myosin motor domain. The stimulatory effect of AA was detectable with both unphosphorylated myosin and the myosin in which RLC was fully phosphorylated. The AA effect on both myosin forms was suggested to cause excess contraction such as vasospasm.
Collapse
Affiliation(s)
- Takeshi Katayama
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | | | - Hideyuki Tanaka
- Department of Research Science, Gunma University School of Health Sciences, Gunma
| | - Mizuki Hino
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Takashi Ohki
- Department of Physics, School of Science and Engineering, Waseda University, Tokyo
| | - Li-Hong Ye
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
- Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, China; and
| | - Ce Xie
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | - Shinji Yoshiyama
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | - Akio Nakamura
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | - Ryoki Ishikawa
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | | | - Kazuhiro Kohama
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Gunma
- Department of Biological Sciences, Marshall University, Huntington, West Virginia
| |
Collapse
|
26
|
Xie Z, Gong MC, Su W, Xie D, Turk J, Guo Z. Role of calcium-independent phospholipase A2beta in high glucose-induced activation of RhoA, Rho kinase, and CPI-17 in cultured vascular smooth muscle cells and vascular smooth muscle hypercontractility in diabetic animals. J Biol Chem 2010; 285:8628-38. [PMID: 20086008 DOI: 10.1074/jbc.m109.057711] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Previous studies suggest that high glucose-induced RhoA/Rho kinase/CPI-17 activation is involved in diabetes-associated vascular smooth muscle hypercontractility. However, the upstream signaling that links high glucose and RhoA/Rho kinase/CPI-17 activation is unknown. Here we report that calcium-independent phospholipase A(2)beta (iPLA(2)beta) is required for high glucose-induced RhoA/Rho kinase/CPI-17 activation and thereby contributes to diabetes-associated vascular smooth muscle hypercontractility. We demonstrate that high glucose increases iPLA(2)beta mRNA, protein, and iPLA(2) activity in a time-dependent manner. Protein kinase C is involved in high glucose-induced iPLA(2)beta protein up-regulation. Inhibiting iPLA(2)beta activity with bromoenol lactone or preventing its expression by genetic deletion abolishes high glucose-induced RhoA/Rho kinase/CPI-17 activation, and restoring expression of iPLA(2)beta in iPLA(2)beta-deficient cells also restores high glucose-induced CPI-17 phosphorylation. Pharmacological and genetic inhibition of 12/15-lipoxygenases has effects on high glucose-induced CPI-17 phosphorylation similar to iPLA(2)beta inhibition. Moreover, increases in iPLA(2) activity and iPLA(2)beta protein expression are also observed in both type 1 and type 2 diabetic vasculature. Pharmacological and genetic inhibition of iPLA(2)beta, but not iPLA(2)gamma, diminishes diabetes-associated vascular smooth muscle hypercontractility. In summary, our results reveal a novel mechanism by which high glucose-induced, protein kinase C-mediated iPLA(2)beta up-regulation activates the RhoA/Rho kinase/CPI-17 via 12/15-lipoxygenases and thereby contributes to diabetes-associated vascular smooth muscle hypercontractility.
Collapse
Affiliation(s)
- Zhongwen Xie
- Department of Physiology, University of Kentucky School of Medicine, Lexington, Kentucky 40536, USA
| | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Injeti ER, Sandoval RJ, Williams JM, Smolensky AV, Ford LE, Pearce WJ. Maximal stimulation-induced in situ myosin light chain kinase activity is upregulated in fetal compared with adult ovine carotid arteries. Am J Physiol Heart Circ Physiol 2008; 295:H2289-98. [PMID: 18835918 DOI: 10.1152/ajpheart.00606.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Postnatal decreases in vascular reactivity involve decreases in the thick filament component of myofilament calcium sensitivity, which is measured as the relationship between cytosolic calcium concentration and myosin light chain (MLC20) phosphorylation. The present study tests the hypothesis that downregulation of thick filament reactivity is due to downregulation of myosin light chain kinase (MLCK) activity in adult compared with fetal arteries. Total MLCK activity, calculated as %MLC20 phosphorylated per second in intact arteries during optimal inhibition of myosin light chain phosphatase activity, was significantly less in adult (6.56+/-0.29%) than in fetal preparations (7.39+/-0.53%). In situ MLC20 concentrations (microM) in adult (198+/-28) and fetal arteries (236+/-44) did not differ significantly. In situ MLCK concentrations (microM), however, were significantly greater in adult (8.21+/-0.59) than in fetal arteries (1.83+/-0.13). In situ MLCK activities (ng MLC20 phosphorylated.s(-1).ng MLCK(-1)) were significantly less in adult (0.26+/-0.01) than in fetal arteries (1.52+/-0.11). In contrast, MLCK activities in adult (15.8+/-1.5) and fetal artery homogenates (17.3+/-1.3) were not significantly different. When in situ fractional activation was calculated, adult values (1.72+/-0.17%) were significantly less than fetal values (9.08+/-0.83%). Together, these results indicate that decreased thick filament reactivity in adult compared with fetal ovine carotid arteries is due at least in part to greater MLCK activity in fetal arteries, which in turn cannot be explained by differences in MLCK, MLC20, or calmodulin concentrations. Instead, this difference appears to involve age-related differences in fractional activation of the MLCK enzyme.
Collapse
Affiliation(s)
- Elisha R Injeti
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | | | | | | | | | | |
Collapse
|
29
|
Li T, Liu L, Liu J, Ming J, Xu J, Yang G, Zhang Y. Mechanisms of Rho kinase regulation of vascular reactivity following hemorrhagic shock in rats. Shock 2008; 29:65-70. [PMID: 17666953 DOI: 10.1097/shk.0b013e318063e477] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our previous research showed that Rho kinase took part in the regulation of vascular hyporeactivity after shock. The objective of the present study was to investigate its mechanism. With isolated superior mesenteric artery (SMA) from hemorrhagic shock rats, we studied the relationship of Rho kinase regulating vascular reactivity to calcium sensitivity and myosin light chain phosphatase (MLCP) and myosin light chain kinase (MLCK). The vascular reactivity and calcium sensitivity of SMA were observed by measuring the contraction initiated by accumulative norepinephrine (NE) and calcium under depolarizing condition (120 mM K(+)) with an isolated organ perfusion system. Hypoxia-treated vascular smooth muscle cells (VSMCs) were used to study the effects of Rho kinase on the activity of MLCP and MLCK and the phosphorylation of 20-kDa myosin light chain (MLC(20)). Myosin light chain (20 kDa) phosphorylation of VSMC in mesenteric artery was detected by immunoprecipitation and Western blotting. The activity of MLCP and MLCK was assayed by enzymatic catalysis. The contractile response of VSMC was measured by the ratio of accumulative infiltration of fluorescent isothiocyanate-conjugated bovine serum albumin through transwell. The results indicated that the vascular reactivity and calcium sensitivity of SMA to NE and calcium following hemorrhagic shock and the contractile response of VSMC to NE following hypoxia were significantly decreased. Angiotensin II (Ang-II), the Rho kinase stimulator, significantly improved hypoxia or hemorrhagic shock-induced decrease of vascular reactivity and calcium sensitivity. These effects of Ang-II on vascular reactivity were abolished by Y-27632, the specific Rho kinase inhibitor. Calyculin A, the MLCP inhibitor, further enhanced Ang-II-induced increase of calcium sensitivity, but ML-9, the MLCK inhibitor, had no effect. Further studies showed Ang-II reversed the hypoxia-induced increase of MLCP activity and increased the hypoxia-induced decrease of MLC(20) phosphorylation in VSMC. It was suggested that Rho kinase played an important role in the regulation of vascular reactivity after hemorrhagic shock. The mechanisms may be related to its calcium sensitivity regulation. Rho kinase up-regulates calcium sensitivity of VSMC possibly through inhibiting the activity of MLCP and increasing the phosphorylation of MLC(20).
Collapse
Affiliation(s)
- Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, The 2nd Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, PR China.
| | | | | | | | | | | | | |
Collapse
|
30
|
Lee E, Hayes DB, Langsetmo K, Sundberg EJ, Tao TC. Interactions between the leucine-zipper motif of cGMP-dependent protein kinase and the C-terminal region of the targeting subunit of myosin light chain phosphatase. J Mol Biol 2007; 373:1198-212. [PMID: 17904578 PMCID: PMC4310484 DOI: 10.1016/j.jmb.2007.08.049] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 07/12/2007] [Accepted: 08/21/2007] [Indexed: 11/29/2022]
Abstract
Nitric oxide induces vasodilation by elevating the production of cGMP, an activator of cGMP-dependent protein kinase (PKG). PKG subsequently causes smooth muscle relaxation in part via activation of myosin light chain phosphatase (MLCP). To date, the interaction between PKG and the targeting subunit of MLCP (MYPT1) is not fully understood. Earlier studies by one group of workers showed that the binding of PKG to MYPT1 is mediated by the leucine-zipper motifs at the N and C termini, respectively, of the two proteins. Another group, however, reported that binding of PKG to MYPT1 did not require the leucine-zipper motif of MYPT1. In this work we fully characterized the interaction between PKG and MYPT1 using biophysical techniques. For this purpose we constructed a recombinant PKG peptide corresponding to a predicted coiled coil region that contains the leucine-zipper motif. We further constructed various C-terminal MYPT1 peptides bearing various combinations of a predicted coiled coil region, extensions preceding this coiled coil region, and the leucine-zipper motif. Our results show, firstly, that while the leucine-zipper motif at the N terminus of PKG forms a homodimeric coiled coil, the one at the C terminus of MYPT1 is monomeric and non-helical. Secondly, the leucine-zipper motif of PKG binds to that of MYPT1 to form a heterodimer. Thirdly, when the leucine-zipper motif of MYPT1 is absent, the PKG leucine-zipper motif binds to the coiled coil region and upstream segments of MYPT1 via formation of a heterotetramer. These results provide rationalization of some of the findings by others using alternative binding analyses.
Collapse
Affiliation(s)
- Eunhee Lee
- Cardiovascular Program, Boston Biomedical Research Institute, Watertown, MA 02472
| | - David B. Hayes
- Cardiovascular Program, Boston Biomedical Research Institute, Watertown, MA 02472
| | - Knut Langsetmo
- Cardiovascular Program, Boston Biomedical Research Institute, Watertown, MA 02472
| | - Eric J. Sundberg
- Cardiovascular Program, Boston Biomedical Research Institute, Watertown, MA 02472
| | - Terence C. Tao
- Cardiovascular Program, Boston Biomedical Research Institute, Watertown, MA 02472
- Department of Neurology, Harvard Medical School, Boston, MA 02115
- Department of Biochemistry, Tufts University School of Medicine, Boston, MA 02111
- *Corresponding author; e-mail address:
| |
Collapse
|
31
|
Sandoval RJ, Injeti ER, Gerthoffer WT, Pearce WJ. Postnatal maturation modulates relationships among cytosolic Ca2+, myosin light chain phosphorylation, and contractile tone in ovine cerebral arteries. Am J Physiol Heart Circ Physiol 2007; 293:H2183-92. [PMID: 17660392 DOI: 10.1152/ajpheart.00647.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study tests the hypothesis that age-related changes in patterns of agonist-induced myofilament Ca(2+) sensitization involve corresponding differences in the relative contributions of thick- and thin-filament regulation to overall myofilament Ca(2+) sensitivity. Posterior communicating cerebral arteries from term fetal and nonpregnant adult sheep were used in measurements of cytosolic Ca(2+), myosin light chain (MLC) phosphorylation, and contractile tensions induced by varying concentrations of K(+) or serotonin [5-hydroxytryptamine (5-HT)]. The results were used to assess the relative contributions of the relationships between cytosolic Ca(2+) and MLC phosphorylation (thick-filament reactivity), along with the relationships between MLC phosphorylation and contractile tension (thin-filament reactivity), to overall myofilament Ca(2+) sensitivity. For K(+)-induced contractions, both fetal and adult arteries exhibited similar basal myofilament Ca(2+) sensitivity. Despite this similarity, thick-filament reactivity was greater in fetal arteries, whereas thin-filament reactivity was greater in adult arteries. In contrast, 5-HT-induced contractions exhibited increased myofilament Ca(2+) sensitivity compared with K(+)-induced contractions for both fetal and adult cerebral arteries, and the magnitude of this effect was greater in fetal compared with adult arteries. When interpreted together with our previous studies of 5-HT-induced myofilament Ca(2+) sensitization, we attributed the present effects to agonist enhancement of thick-filament reactivity in fetal arteries mediated by G protein receptor activation of a PKC-independent but RhoA-dependent pathway. In adult arteries, agonist stimulation enhanced thin-filament reactivity was also probably mediated through G protein-coupled activation of RhoA-dependent and PKC-independent mechanisms. Overall, the present data demonstrate that agonist-enhanced myofilament Ca(2+) sensitivity can be partitioned into separate thick- and thin-filament effects, the magnitudes of which are different between fetal and adult cerebral arteries.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/metabolism
- Aging/metabolism
- Animals
- Calcium/metabolism
- Calcium Signaling/drug effects
- Cerebral Arteries/drug effects
- Cerebral Arteries/enzymology
- Cerebral Arteries/growth & development
- Cerebral Arteries/metabolism
- Cytosol/metabolism
- Dose-Response Relationship, Drug
- Fetus/blood supply
- Heat-Shock Proteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Myosin-Light-Chain Phosphatase/metabolism
- Phosphorylation
- Potassium/metabolism
- Protein Kinase C/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Serotonin/pharmacology
- Sheep
- Tissue Culture Techniques
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/metabolism
- Vasoconstrictor Agents/pharmacology
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Renan J Sandoval
- Department of Physiology and Pharmacology, Center for Perinatal Biology, Loma Linda University, School of Medicine, Loma Linda, California 92354, USA
| | | | | | | |
Collapse
|
32
|
Takeuchi T, Nakajima H, Hata F, Azuma YT. A minor role for Ca2+ sensitization in sustained contraction through activation of muscarinic receptor in circular muscle of rat distal colon. Pflugers Arch 2007; 454:565-74. [PMID: 17318645 DOI: 10.1007/s00424-007-0221-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Revised: 12/06/2006] [Accepted: 01/25/2007] [Indexed: 10/23/2022]
Abstract
We previously demonstrated that Ca(2+) sensitization has an essential role for carbachol-induced contraction in the longitudinal muscle of the rat distal colon. In the present study, we extended these studies to clarify the role of Ca(2+) sensitization in contraction induced by the activation of muscarinic receptors in the circular muscle of the rat distal colon. Carbachol induced a rapid phasic contraction followed by a sustained contraction that was significantly lower than the phasic and was superimposed with the rhythmic contractions. The extent of increase in intracellular Ca(2+) concentration that was measured simultaneously with tension recording was dissociated from the phasic contraction, whereas it exhibited to a similar extent as sustained contraction. In alpha-toxin-permeabilized preparations, Ca(2+) induced contraction comprising a rapid phasic and a subsequent low sustained component. After Ca(2+)-induced sustained contraction reached a constant level, guanosine triphosphate (GTP) addition resulted in the enhancement of contractile force in a concentration-dependent manner. Carbachol in the presence of GTP caused a further minimal increase in tension (Ca(2+) sensitization). Chelerythrine, a protein kinase C (PKC) inhibitor, inhibited carbachol-induced Ca(2+) sensitization but not GTP-induced Ca(2+) sensitization. In contrast, Y-27632, a Rho kinase inhibitor, inhibited GTP-induced Ca(2+) sensitization but not that induced by carbachol. Phorbol 12,13-dibutyrate, a PKC activator, increased the sustained contraction. These results suggest that the activation of muscarinic receptor with carbachol induces Ca(2+) sensitization via activation of PKC, but this action is minor in the circular muscle of the rat distal colon as a result of limited coupling between muscarinic receptors and Ca(2+) sensitization via the PKC pathway.
Collapse
Affiliation(s)
- Tadayoshi Takeuchi
- Laboratory of Veterinary Pharmacology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| | | | | | | |
Collapse
|
33
|
Lee JH, Ragolia L. AKT phosphorylation is essential for insulin-induced relaxation of rat vascular smooth muscle cells. Am J Physiol Cell Physiol 2006; 291:C1355-65. [PMID: 16855220 PMCID: PMC1636679 DOI: 10.1152/ajpcell.00125.2006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Insulin resistance, a major factor in the development of type 2 diabetes, is known to be associated with defects in blood vessel relaxation. The role of Akt on insulin-induced relaxation of vascular smooth muscle cell (VSMC) was investigated using siRNA targeting Akt (siAKTc) and adenovirus constructing myristilated Akt to either suppress endogenous Akt or overexpress constitutively active Akt, respectively. siAKTc decreased both basal and insulin-induced phosphorylations of Akt and glycogen synthase kinase 3beta, abolishing insulin-induced nitric oxide synthase (iNOS) expression. cGMP-dependent kinase 1alpha (cGK1alpha) and myosin-bound phosphatase (MBP) activities, both downstream of iNOS, were also decreased. siAKTc treatment resulted in increased insulin and ANG II-stimulated phosphorylation of contractile apparatus, such as MBP substrate (MYPT1) and myosin light chain (MLC20), accompanied by increased Rho-associated kinase alpha (ROKalpha) activity, demonstrating the requirement of Akt for insulin-induced vasorelaxation. Corroborating these results, constitutively active Akt upregulated the signaling molecules involved in insulin-induced relaxation such as iNOS, cGK1alpha, and MBP activity, even in the absence of insulin stimulation. On the contrary, the contractile response involving the phosphorylation of MYPT1 and MLC20, and increased ROKalpha activity stimulated by ANG II were all abolished by overexpressing active Akt. In conclusion, we demonstrated here that insulin-induced VSMC relaxation is dependent on Akt activation via iNOS, cGK1alpha, and MBP activation, as well as the decreased phosphorylations of MYPT1 and MLC20 and decreased ROKalpha activity.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Animals
- Carrier Proteins/metabolism
- Cells, Cultured
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Humans
- Insulin/metabolism
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myosin Light Chains/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Phosphoprotein Phosphatases/metabolism
- Phosphorylation
- Protein Phosphatase 1
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Inbred WKY
- Vasodilation/physiology
- rho-Associated Kinases
Collapse
Affiliation(s)
- Jin Hee Lee
- Vascular Biology Institute, Winthrop-University Hospital, Mineola NY 11501
| | - Louis Ragolia
- Vascular Biology Institute, Winthrop-University Hospital, Mineola NY 11501
- School of Medicine, State University of New York at Stony Brook, Stony Brook, New York 11794
| |
Collapse
|
34
|
Liu Y, Taylor CW. Stimulation of arachidonic acid release by vasopressin in A7r5 vascular smooth muscle cells mediated by Ca2+-stimulated phospholipase A2. FEBS Lett 2006; 580:4114-20. [PMID: 16828086 DOI: 10.1016/j.febslet.2006.06.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Accepted: 06/19/2006] [Indexed: 11/22/2022]
Abstract
Arachidonic acid (AA) regulates many aspects of vascular smooth muscle behaviour, but the mechanisms linking receptors to AA release are unclear. In A7r5 vascular smooth muscle cells pre-labelled with (3)H-AA, vasopressin caused a concentration-dependent stimulation of 3H-AA release that required phospholipase C and an increase in cytosolic [Ca2+]. Ca2+ release from intracellular stores and Ca2+ entry via L-type channels or the capacitative Ca2+ entry pathway were each effective to varying degrees. Selective inhibitors of PLA2 inhibited the 3H-AA release evoked by vasopressin, though not the underlying Ca2+ signals, and established that cPLA2 mediates the release of AA. We conclude that in A7r5 cells vasopressin stimulates AA release via a Ca2+-dependent activation of cPLA2.
Collapse
Affiliation(s)
- Yingjie Liu
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | | |
Collapse
|
35
|
Kamata K, Hosokawa M, Matsumoto T, Kobayashi T. Altered arachidonic acid-mediated responses in the perfused kidney of the streptozotocin-induced diabetic rat. J Smooth Muscle Res 2006; 42:171-87. [PMID: 17159333 DOI: 10.1540/jsmr.42.171] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Using perfused kidneys isolated from age-matched controls and streptozotocin (STZ)-induced diabetic rats, we investigated the effects of arachidonic acid (AA) on perfusion pressure in the presence of methoxamine. AA elicited a transient contraction followed by a sustained relaxation in each group. The amplitude of contraction was smaller in the diabetic group than in the control group, whereas the amplitude of the sustained relaxation was greater in the former than in the latter group. In the diabetic group, the AA-induced sustained relaxation was completely inhibited by indomethacin [cyclooxygenase (COX) inhibitor], SKF525A [cytochrome P450 (CYP450) inhibitor], or clotrimazole (epoxygenase inhibitor), but not by furegrelate [thromboxane A(2) (TXA(2))-synthase inhibitor], SQ29548 (TXA(2)-receptor antagonist), or baicalein [lipoxygenase (LOX) inhibitor]. In the diabetic kidney, more-or-less additive inhibitions of the AA-induced relaxation were seen when indomethacin was given with either SKF525A or clotrimazole. These results suggest that in the STZ-induced diabetic perfused kidney, vasorelaxant metabolites derived from AA (probably COX and/or CYP450 metabolites) are increased, and may serve to regulate vascular tone.
Collapse
Affiliation(s)
- Katsuo Kamata
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan.
| | | | | | | |
Collapse
|
36
|
Yang JX, Lin Y. The action of PKA on smooth muscle myosin phosphorylation. Life Sci 2005; 77:2669-75. [PMID: 15961117 DOI: 10.1016/j.lfs.2005.04.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2004] [Revised: 02/20/2005] [Accepted: 04/14/2005] [Indexed: 10/25/2022]
Abstract
The aim of the study is to reveal the characterization of PKA acting on myosin. We found: (a) in the absence of Ca(2+)/CaM, PKA slightly phosphorylated MLC(20) and stimulated the Mg(2+)-ATPase activity of myosin, which was strengthened significantly by arachidonic acid (ACAD); (b) Ca(2+)-independent phosphorylation of myosin by PKA was obviously less efficient than both Ca(2+)-dependent and independent phosphorylation of myosin by MLCK; (c) micro-amount of calponin could not increase the precipitation of myosin phosphorylated by PKA, but it increased the precipitation of myosin phosphorylated by MLCK, suggesting the presence of conformational differences between the myosins phosphorylated by PKA and by MLCK.
Collapse
Affiliation(s)
- Jing Xian Yang
- Department of Pharmacology, Dalian Medical University, 465 Zhongshan Road, Dalian 116027, PR China.
| | | |
Collapse
|
37
|
Yamada A, Hirose K, Hashimoto A, Iino M. Real-time imaging of myosin II regulatory light-chain phosphorylation using a new protein biosensor. Biochem J 2005; 385:589-94. [PMID: 15362955 PMCID: PMC1134733 DOI: 10.1042/bj20040778] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphorylation of the RMLC (regulatory myosin light chain) regulates the activity of myosin II, which is critically involved in the motility of both muscle and non-muscle cells. There are both Ca2+-dependent and -independent pathways for RMLC phosphorylation in smooth-muscle cells, and the latter pathway is often involved in an abnormal contractility in pathological states such as asthma and hypertension. Therefore pharmacological interventions of RMLC phosphorylation may have a therapeutic value. In the present study, we developed a new genetically encoded biosensor, termed CRCit (ECFP-RMLC-Citrine, where ECFP is enhanced cyan fluorescent protein), that detects RMLC phosphorylation using fluorescence resonance energy transfer between two variants of the green fluorescent protein fused to both the N- and C-termini of RMLC. When expressed in primary cultured vascular smooth-muscle cells, CRCit detected the Ca2+-dependent RMLC phosphorylation with a high spatiotemporal resolution. Furthermore, we could specifically assay the agonist-induced Ca2+-independent phosphorylation of RMLC when Ca2+ signalling in cells expressing CRCit was suppressed. Thus CRCit may also be used for the high throughput screening of compounds that inhibit abnormal smooth-muscle contraction.
Collapse
Affiliation(s)
- Aki Yamada
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Akiko Hashimoto
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Masamitsu Iino
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
38
|
Abstract
The present review is an attempt to put into perspective the available information on the putative changes in cellular mechanisms of the contractile properties of the aging gastrointestinal (GI) smooth muscle. Information on smooth muscle of the GI tract is scanty. Smooth muscle cells from old rats (32 months old) exhibit limited cell length distribution and diminished contractility. The observed reduced contractile response may be due to the effect of aging on signal transduction pathways, especially an inhibition of the tyrosine kinase-Src kinase pathway, a reduced activation of the PKCalpha pathway, a reduced association of contractile proteins (HSP27-tropomyosin, HSP27-actin, and actin-myosin). Levels of HSP27-phosphorylation are also reduced compared to adult rats. Regulation of GI motility is a complex mechanism of signal transduction and interaction of signaling and contractile proteins. It is suggested that further studies to elucidate the role of HSP27 in aging smooth muscle of the GI tract are needed.
Collapse
Affiliation(s)
- Khalil N Bitar
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
39
|
Vijayan KV, Liu Y, Sun W, Ito M, Bray PF. The Pro33 isoform of integrin beta3 enhances outside-in signaling in human platelets by regulating the activation of serine/threonine phosphatases. J Biol Chem 2005; 280:21756-62. [PMID: 15826939 DOI: 10.1074/jbc.m500872200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Integrin beta(3) is polymorphic at residue 33 (Leu(33) or Pro(33)), and the Pro(33)-positive platelets display enhanced aggregation, P-selectin secretion, and shorter bleeding times. Because outside-in signaling is critical for platelet function, we hypothesized that the Pro(33) variant provides a more efficient signaling than the Leu(33) isoform. When compared with Pro(33)-negative platelets, Pro(33)-positive platelets demonstrated significantly greater serine/threonine phosphorylation of extracellular signal-regulated kinase (ERK2) and myosin light chain (MLC) but not cytoplasmic phospholipase A2 upon thrombin-induced aggregation. Tyrosine phosphorylation of integrin beta(3) and the adaptor protein Shc was no different in the fibrinogen-engaged platelets from both genotypes. The addition of Integrilin (alpha(IIb)beta(3)-fibrinogen blocker) or okadaic acid (serine/threonine phosphatase inhibitor) dramatically enhanced ERK2 and MLC phosphorylation in the Pro(33)-negative platelets when compared with Pro(33)-positive platelets, suggesting that integrin engagement during platelet aggregation activates serine/threonine phosphatases. The phosphatase activity of myosin phosphatase (MP) that dephosphorylates MLC is inactivated by phosphorylation of the myosin binding subunit of MP at Thr(696), and aggregating Pro(33)-positive platelets exhibited an increased Thr(696) phosphorylation of MP. These studies highlight a role for the dephosphorylation events via the serine/threonine phosphatases during the integrin outside-in signaling mechanism, and the Leu(33) --> Pro polymorphism regulates this process. Furthermore, these findings support a mechanism whereby the reported enhanced alpha granule secretion in the Pro(33)-positive platelets could be mediated by an increased phosphorylation of MLC, which in turn is caused by an increased phosphorylation and subsequent inactivation of myosin phosphatase.
Collapse
Affiliation(s)
- K Vinod Vijayan
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 286, N1319, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
40
|
Koga Y, Ikebe M. p116Rip Decreases Myosin II Phosphorylation by Activating Myosin Light Chain Phosphatase and by Inactivating RhoA. J Biol Chem 2005; 280:4983-91. [PMID: 15545284 DOI: 10.1074/jbc.m410909200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p116Rip was originally found to be a RhoA-binding protein, but its function has been unknown. Here, we clarify the function of p116Rip. Two critical findings were made. First, we found that p116Rip activated the GTPase activity of RhoA in vitro and that p116Rip overexpression in cells consistently diminished the epidermal growth factor-induced increase in GTP-bound RhoA. Second, p116Rip activated the myosin light chain phosphatase (MLCP) activity of the holoenzyme. p116Rip did not activate the catalytic subunit alone, indicating that the activation is due to the binding of p116Rip to the myosin phosphatase targeting subunit MYPT1. Interestingly, the activation of phosphatase was specific to myosin as substrate, and p116Rip directly bound to myosin, thus facilitating myosin/MLCP interaction. The gene silencing of p116Rip consistently and significantly increased myosin phosphorylation as well as stress fiber formation in cells. Based upon these findings, we propose that p116Rip is an important regulatory component that controls the RhoA signaling pathway, thus regulating MLCP activity and myosin phosphorylation in cells.
Collapse
Affiliation(s)
- Yasuhiko Koga
- Department of Physiology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | |
Collapse
|
41
|
Xiao D, Longo LD, Zhang L. Alpha1-adrenoceptor-mediated phosphorylation of MYPT-1 and CPI-17 in the uterine artery: role of ERK/PKC. Am J Physiol Heart Circ Physiol 2005; 288:H2828-35. [PMID: 15665049 DOI: 10.1152/ajpheart.01189.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously demonstrated that ERK/PKC signaling pathways play a key role in regulation of Ca(2+) sensitivity and contractility of the uterine artery. The present study tested the hypothesis that ERK and PKC differentially regulated myosin light chain phosphatase activity by phosphorylation of myosin phosphatase target protein-1 (MYPT-1) and CPI-17. Agonist-induced contractions and phosphorylation of MYPT-1/Thr(696), MYPT-1/Thr(850), and CPI-17/Thr(38) were measured simultaneously in the same tissues of isolated near-term pregnant ovine uterine arteries. Phenylephrine produced time-dependent concurrent increases in the phosphorylation of ERK(44/42) and MYPT-1/Thr(850) that preceded contractions. In addition, phenylephrine induced phosphorylation of CPI-17/Thr(38) that was concurrent with the contractions. In contrast, phenylephrine did not induce phosphorylation of MYPT-1/Thr(696) in the uterine artery. PD-098059 inhibited phosphorylation of ERK(44/42) and the initial peak phosphorylation of MYPT-1/Thr(850) but did not affect CPI-17/Thr(38) phosphorylation. Activation of PKC by phorbol 12,13-dibutyrate induced a time-dependent phosphorylation of CPI-17/Thr(38) that preceded contractions of the uterine artery. In addition, phorbol 12,13-dibutyrate activated PKC-alpha and induced a coimmunoprecipitation of PKC-alpha with caldesmon. The results suggest that phosphorylation of MYPT-1/Thr(850) and CPI-17/Thr(38) play important roles in regulation of agonist-mediated Ca(2+) sensitivity in the uterine artery, in part by ERK and PKC, respectively. In addition, phosphorylated CPI-17 may regulate Ca(2+) sensitivity by interacting with caldesmon and reversing its inhibitory effect on myosin ATPase.
Collapse
Affiliation(s)
- Daliao Xiao
- Center for Perinatal Biology, Dept. of Pharmacology & Physiology, Loma Linda Univ. School of Medicine, Loma Linda, CA 92350, USA
| | | | | |
Collapse
|
42
|
Ding X, Murray PA. Regulation of pulmonary venous tone in response to muscarinic receptor activation. Am J Physiol Lung Cell Mol Physiol 2005; 288:L131-40. [PMID: 15377494 DOI: 10.1152/ajplung.00230.2004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated cellular mechanisms that mediate or modulate the vascular response to muscarinic receptor activation (ACh) in pulmonary veins (PV). Isometric tension was measured in isolated canine PV rings with endothelium (E+) and without endothelium (E−). Tension and intracellular Ca2+concentration ([Ca2+]i) were measured simultaneously in fura-2-loaded E− PV strips. In the absence of preconstriction, ACh (0.01–10 μM) caused dose-dependent contraction in E+ and E− rings. ACh contraction was potentiated by removing the endothelium or by nitric oxide (NO) synthase inhibition ( N-nitro-l-arginine methyl ester, P = 0.001). Cyclooxygenase inhibition (indomethacin) reduced ACh contraction in both E+ and E− PV rings ( P = 0.013 and P = 0.037, respectively). ACh contraction was attenuated by inhibitors of voltage-operated Ca2+channels (nifedipine, P < 0.001), inositol-1,4,5-trisphosphate (IP3)-mediated Ca2+release (2-aminoethoxydiphenyl borate, P = 0.001), PKC (bisindolylmaleimide I, P = 0.001), Rho-kinase (Y-27632, P = 0.002), and tyrosine kinase (TK; tyrphostin 47, P = 0.015) in E− PV rings. ACh (1 μM) caused a leftward shift in the [Ca2+]i-tension relationship ( P = 0.015), i.e., ACh increased myofilament Ca2+sensitivity. Inhibition of PKC, Rho-kinase, and TK attenuated the ACh-induced increase in myofilament Ca2+sensitivity ( P < 0.001, P < 0.001, and P = 0.024, respectively). These findings indicate that in canine PV, ACh contraction is modulated by NO and partially mediated by metabolites of the cyclooxygenase pathway and involves Ca2+influx through voltage-operated Ca2+channels and IP3-mediated Ca2+release. In addition, ACh induces increased myofilament Ca2+sensitivity, which requires the PKC, Rho-kinase, and TK pathways.
Collapse
Affiliation(s)
- Xueqin Ding
- Center for Anesthesiology Research, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | |
Collapse
|
43
|
Cassis LA, Huang J, Gong MC, Daugherty A. Role of metabolism and receptor responsiveness in the attenuated responses to Angiotensin II in mice compared to rats. ACTA ACUST UNITED AC 2004; 117:107-16. [PMID: 14700746 DOI: 10.1016/j.regpep.2003.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chronic infusion of Angiotensin II (AngII) to rats is a well-characterized model for determining AngII physiology. Genetic manipulations have strengthened knowledge of AngII; however, they do not permit an increase in AngII to be initiated at a selected age, duration and dose. Therefore, exogenous AngII administration remains an important technique to define its biological effect. We previously noted that infusion of AngII to mice had minimal effects compared to the same dose given to rats. In this study, we compared the effects of chronic infusion of the same dose of AngII to C57BL/6 mice and Sprague-Dawley rats, two commonly used rodent models. Rats administered AngII exhibited reductions (by 22%) in body weight, which were not evident in mice. AngII increased blood pressure by 54 mm Hg in rats, but had no effect in mice. Vascular histology demonstrated that AngII caused medial hypertrophy in rats, with adventitial expansion in mice. Plasma concentrations of AngII and its catabolic fragments were elevated (twofold) in mice compared to rats. Angiotensin receptor affinity, density and distribution were similar in rats and mice. Infusion of AngII decreased AngII receptor density in the kidney (by 78%) and spleen (by 29%) of mice, but had no effect in rats. AngII produced a sustained contractile response in rat aortic strips, but minimal responses in mouse aorta. These results demonstrate that differences in circulating angiotensin peptides, AngII receptor regulation, and vascular reactivity contribute to diminished responses to AngII infusion in mice compared to rats. Results from this study suggest that considerably higher doses of AngII may be required to elicit physiologic effects of AngII in mice.
Collapse
Affiliation(s)
- Lisa A Cassis
- Room 434, Division of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0082, USA.
| | | | | | | |
Collapse
|
44
|
Shimomura E, Shiraishi M, Iwanaga T, Seto M, Sasaki Y, Ikeda M, Ito K. Inhibition of protein kinase C-mediated contraction by Rho kinase inhibitor fasudil in rabbit aorta. Naunyn Schmiedebergs Arch Pharmacol 2004; 370:414-22. [PMID: 15459803 DOI: 10.1007/s00210-004-0975-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2004] [Accepted: 08/02/2004] [Indexed: 10/26/2022]
Abstract
Protein kinase C (PKC) activation by a phorbol ester increases myosin light chain (MLC(20)) phosphorylation through inhibition of MLC phosphatase (MLCP) and enhances contraction of vascular smooth muscle. We investigated whether Rho kinase, which is known to inhibit MLCP, is involved in the MLC(20) phosphorylation caused by a phorbol ester, 12-deoxyphorbol 13-isobutyrate (DPB), in rabbit aortas. DPB (1 microM) increased MLC(20) phosphorylation and tension. The Rho kinase inhibitor fasudil (10 microM) inhibited the DPB-induced contraction and decreased the MLC(20) phosphorylation at Ser19, a site phosphorylated by MLC kinase, although it did not affect the phosphorylation of total MLC(20). Rinsing a 65.4 mM KCl-contracted aorta with Ca(2+)-free, EGTA solution caused rapid dephosphorylation of MLC(20) and relaxation. When DPB was present in the rinsing solution, the MLC(20) dephosphorylation and the relaxation were inhibited. In this protocol, Ro31-8220 (10 microM), a PKC inhibitor, suppressed the phosphorylation of total MLC(20) and Ser19 induced by DPB. Fasudil also inhibited the Ser19 phosphorylation to a degree similar to Ro31-8220 and accelerated relaxation, which was less than the relaxation caused by Ro31-8220. The phospholipase A(2) inhibitor ONO-RS-082 (5 microM) inhibited the DPB-induced Ser19 phosphorylation but only transiently decreased the tension, suggesting the involvement of arachidonic acid in the phosphorylation and the existence of a MLC(20) phosphorylation-independent mechanism. When fasudil was combined with ONO-RS-082, fasudil exerted additional inhibition of the tension without further inhibition of the Ser19 phosphorylation. DPB phosphorylated the 130 kDa myosin binding subunit (MBS) of MLCP and fasudil inhibited the phosphorylation. These data suggest that the inhibition by fasudil of DPB-induced contraction and phosphorylation of MLC(20) at the MLC kinase-targeted site is a result of inhibition of Rho kinase. Thus, the PKC-dependent Ca(2+)-sensitization of vascular smooth muscle involves Rho kinase. A MLC(20) phosphorylation-independent mechanism is also involved in the Ca(2+)-sensitization.
Collapse
Affiliation(s)
- Erika Shimomura
- Department of Veterinary Pharmacology, Faculty of Agriculture, Miyazaki University, 889-2192 Miyazaki, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Patil SB, Tsunoda Y, Pawar MD, Bitar KN. Translocation and association of ROCK-II with RhoA and HSP27 during contraction of rabbit colon smooth muscle cells. Biochem Biophys Res Commun 2004; 319:95-102. [PMID: 15158447 DOI: 10.1016/j.bbrc.2004.04.159] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Indexed: 11/15/2022]
Abstract
The focus of the paper is to understand the role of HSP27 in mediating the association of RhoA with ROCK-II in sustained contraction of smooth muscle cells from the rabbit colon. In circular smooth muscle cells; acetylcholine-induced contraction (10(-7)M) was associated with translocation of ROCK-II to the particulate fraction, which remained sustained at 4 min after stimulation (135.1+/-8.1% increase, P </= 0.05). There was also an increased association of ROCK-II with RhoA particulate fraction (147.46+/-9.31 and 148.22+/-9.41, n = 3, P </= 0.05) and with HSP27 (155.6+/-10.7% increase, P </= 0.05) in the particulate fraction. Pre-incubation of cells with Y27632 resulted in the inhibition of the association of ROCK-II with RhoA in the particulate fraction. Acetylcholine (10(-7)M) induced sustained phosphorylation of MLC (122.75+/-9.97%, P </= 0.05 and 174.65+/-28.36%, P </= 0.05 increase in the di phospho-MLC at 30s and 4 min, respectively), which was inhibited upon pre-incubation with Y27632. Results suggest that ROCK-II undergoes a translocation to the particulate fraction with RhoA and with HSP27, suggesting that translocation and association of ROCK-II with RhoA is mediated by HSP27. Maintenance of the functional association of RhoA with ROCK-II in the particulate fraction mediated by HSP27 appears to be important to retain MLC in the phosphorylated state and hence the sustained contraction.
Collapse
Affiliation(s)
- Suresh B Patil
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
46
|
Patil SB, Pawar MD, Bitar KN. Direct association and translocation of PKC-alpha with calponin. Am J Physiol Gastrointest Liver Physiol 2004; 286:G954-63. [PMID: 14726309 DOI: 10.1152/ajpgi.00477.2003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Calponin has been implicated in the regulation of smooth muscle contraction through its interaction with F-actin and inhibition of the actin-activated MgATPase activity of phosphorylated myosin. Calponin has also been shown to interact with PKC. We have studied the interaction of calponin with PKC-alpha and with the low molecular weight heat-shock protein (HSP)27 in contraction of colonic smooth muscle cells. Particulate fractions from isolated smooth muscle cells were immunoprecipitated with antibodies to calponin and Western blot analyzed with antibodies to HSP27 and to PKC-alpha. Acetylcholine induced a sustained increase in the immunocomplexing of calponin with HSP27 and of calponin with PKC-alpha in the particulate fraction, indicating an association of the translocated proteins in the membrane. To examine whether the observed interaction in vivo is due to a direct interaction of calponin with PKC-alpha, a cDNA of 1.3 kb of human calponin gene was PCR amplified. PCR product encoding 622 nt of calponin cDNA (nt 351-972 corresponding to amino acids 92-229) was expressed as fusion glutathione S-transferase (GST) protein in the vector pGEX-KT. We have studied the direct association of GST-calponin fusion protein with recombinant PKC-alpha in vitro. Western blot analysis of the fractions collected after elution with reduced glutathione buffer (pH 8.0) show a coelution of GST-calponin with PKC-alpha, indicating a direct association of GST-calponin with PKC-alpha. These data suggest that there is a direct association of translocated calponin and PKC-alpha in the membrane and a role for the complex calponin-PKC-alpha-HSP27, in contraction of colonic smooth muscle cells.
Collapse
Affiliation(s)
- Suresh B Patil
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
47
|
Hirano K, Derkach DN, Hirano M, Nishimura J, Takahashi S, Kanaide H. Transduction of the N-Terminal Fragments of MYPT1 Enhances Myofilament Ca2+Sensitivity in an Intact Coronary Artery. Arterioscler Thromb Vasc Biol 2004; 24:464-9. [PMID: 14707041 DOI: 10.1161/01.atv.0000116028.42230.4c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The region of the 110 kDa regulatory subunit (MYPT1) of smooth muscle myosin phosphatase involved in the regulation of contraction was determined under physiological conditions. METHODS AND RESULTS Using HIV Tat protein-mediated protein transduction, the N-terminal fragments of MYPT1 were introduced to the intact porcine coronary arterial strips. Pre-incubation with 3 micromol/L TAT-MYPT1(1-374), a construct containing the Tat peptide and the residues 1 to 374 of MYPT1, for 15 minutes augmented (2.4-fold) the subsequent contraction induced by adding 1.25 mmol/L of extracellular Ca2+ under 118 mmol/L K+ depolarization, with no augmentation of the [Ca2+]i elevation. The deletion of the Tat peptide, MYPT1(1-374), abolished the augmenting effect. TAT-MYPT1(1-296) demonstrated a weaker but significant augmentation (1.7-fold). However, TAT-MYPT1(1-171), TAT-MYPT1(39-374), TAT-MYPT1(39-296), and TAT-MYPT1(297-374) had no augmenting activity. The myosin light chain phosphorylation level as a function of extracellular Ca2+ concentrations was shifted to the left in the strips pretreated with TAT-MYPT1(1-374) compared with the control. CONCLUSIONS Region 1 to 296 was the minimal region involved in the enhancement of contraction, and region 297 to 374 played a supplemental role. These results suggested that the interaction mainly between catalytic subunit and MYPT1 play a critical role in the regulation of the endogenous myosin phosphatase in intact smooth muscle.
Collapse
Affiliation(s)
- Katsuya Hirano
- Division of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Regulation of vascular smooth muscle cell contractile state is critical for the maintenance of blood vessel tone. Abnormal vascular smooth muscle cell contractility plays an important role in the pathogenesis of hypertension, blood vessel spasm, and atherosclerosis. Myosin phosphatase, the key enzyme controlling myosin light chain dephosphorylation, regulates smooth muscle cell contraction. Vasoconstrictor and vasodilator pathways inhibit and activate myosin phosphatase, respectively. G-protein-coupled receptor agonists can inhibit myosin phosphatase and cause smooth muscle cell contraction by activating RhoA/Rho kinase, whereas NO/cGMP can activate myosin phosphatase and cause smooth muscle cell relaxation by activation of cGMP-dependent protein kinase. We have used yeast two-hybrid screening to identify a 116-kDa human protein that interacts with both myosin phosphatase and RhoA. This myosin phosphatase-RhoA interacting protein, or M-RIP, is highly homologous to murine p116RIP3, is expressed in vascular smooth muscle, and is localized to actin myofilaments. M-RIP binds directly to the myosin binding subunit of myosin phosphatase in vivo in vascular smooth muscle cells by an interaction between coiled-coil and leucine zipper domains in the two proteins. An adjacent domain of M-RIP directly binds RhoA in a nucleotide-independent manner. M-RIP copurifies with RhoA and Rho kinase, colocalizes on actin stress fibers with RhoA and MBS, and is associated with Rho kinase activity in vascular smooth muscle cells. M-RIP can assemble a complex containing both RhoA and MBS, suggesting that M-RIP may play a role in myosin phosphatase regulation by RhoA.
Collapse
Affiliation(s)
- Howard K Surks
- Molecular Cardiology Research Institute, Cardiology Division and Department of Medicine, Tufts-New England Medical Center, Boston, Massachusetts 02111, USA.
| | | | | |
Collapse
|
49
|
Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003; 83:1325-58. [PMID: 14506307 DOI: 10.1152/physrev.00023.2003] [Citation(s) in RCA: 1547] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Ca2+ sensitivity of smooth muscle and nonmuscle myosin II reflects the ratio of activities of myosin light-chain kinase (MLCK) to myosin light-chain phosphatase (MLCP) and is a major, regulated determinant of numerous cellular processes. We conclude that the majority of phenotypes attributed to the monomeric G protein RhoA and mediated by its effector, Rho-kinase (ROK), reflect Ca2+ sensitization: inhibition of myosin II dephosphorylation in the presence of basal (Ca2+ dependent or independent) or increased MLCK activity. We outline the pathway from receptors through trimeric G proteins (Galphaq, Galpha12, Galpha13) to activation, by guanine nucleotide exchange factors (GEFs), from GDP. RhoA. GDI to GTP. RhoA and hence to ROK through a mechanism involving association of GEF, RhoA, and ROK in multimolecular complexes at the lipid cell membrane. Specific domains of GEFs interact with trimeric G proteins, and some GEFs are activated by Tyr kinases whose inhibition can inhibit Rho signaling. Inhibition of MLCP, directly by ROK or by phosphorylation of the phosphatase inhibitor CPI-17, increases phosphorylation of the myosin II regulatory light chain and thus the activity of smooth muscle and nonmuscle actomyosin ATPase and motility. We summarize relevant effects of p21-activated kinase, LIM-kinase, and focal adhesion kinase. Mechanisms of Ca2+ desensitization are outlined with emphasis on the antagonism between cGMP-activated kinase and the RhoA/ROK pathway. We suggest that the RhoA/ROK pathway is constitutively active in a number of organs under physiological conditions; its aberrations play major roles in several disease states, particularly impacting on Ca2+ sensitization of smooth muscle in hypertension and possibly asthma and on cancer neoangiogenesis and cancer progression. It is a potentially important therapeutic target and a subject for translational research.
Collapse
Affiliation(s)
- Andrew P Somlyo
- Dept. of Molecular Physiology and Biological Physics, Univ. of Virginia, PO Box 800736, Charlottesville, VA 22908-0736.
| | | |
Collapse
|
50
|
Surks HK, Mendelsohn ME. Dimerization of cGMP-dependent protein kinase 1alpha and the myosin-binding subunit of myosin phosphatase: role of leucine zipper domains. Cell Signal 2003; 15:937-44. [PMID: 12873707 DOI: 10.1016/s0898-6568(03)00057-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nitric oxide (NO) and nitrovasodilators induce vascular smooth muscle cell relaxation in part by cGMP-dependent protein kinase (cGK)-mediated activation of myosin phosphatase, which dephosphorylates myosin light chains. We recently found that cGMP-dependent protein kinase 1alpha binds directly to the myosin-binding subunit (MBS) of myosin phosphatase via the leucine/isoleucine zipper of cGK. We have now studied the role of the leucine zipper domain of MBS in dimerization with cGK and the leucine/isoleucine zipper and leucine zipper domains of both proteins in homodimerization. Mutagenesis of the MBS leucine zipper domain disrupts cGKIalpha-MBS dimerization. Mutagenesis of the MBS leucine zipper eliminates MBS homodimerization, while similar disruption of the cGKIalpha leucine/isoleucine zipper does not prevent formation of cGK dimers. The MBS leucine zipper domain is phosphorylated by cGK, but this does not have any apparent effect on heterodimer formation between the two proteins. MBS LZ mutants that are unable to bind cGK were poor substrates for cGK. These data support the theory that the MBS leucine zipper domain is necessary and sufficient to mediate both MBS homodimerization and binding of the protein to cGK. In contrast, the leucine/isoleucine zipper of cGK is required for binding to MBS, but not for cGK homodimerization. These data support that the MBS and cGK leucine zipper domains mediate the interaction between these two proteins. The contribution of these domains to both homodimerization and their specific interaction with each other suggest that additional regulatory mechanisms involving these domains may exist.
Collapse
Affiliation(s)
- Howard K Surks
- Molecular Cardiology Research Institute, Cardiology Division, Department of Medicine, New England Medical Center Hospitals, Inc., Tufts University School of Medicine, 750 Washington Street, Box 80, Boston, MA 02111, USA.
| | | |
Collapse
|