Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Am J Physiol Heart Circ Physiol ; 326(3): H860-H869, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38276948

ABSTRACT

Targeting alternative exons for therapeutic gain has been achieved in a few instances and potentially could be applied more broadly. The myosin phosphatase (MP) enzyme is a critical hub upon which signals converge to regulate vessel tone. Alternative exon 24 of myosin phosphatase regulatory subunit (Mypt1 E24) is an ideal target as toggling between the two isoforms sets smooth muscle sensitivity to vasodilators such as nitric oxide (NO). This study aimed to develop a gene-based therapy to suppress splicing of Mypt1 E24 thereby switching MP enzyme to the NO-responsive isoform. CRISPR/Cas9 constructs were effective at editing of Mypt1 E24 in vitro; however, targeting of vascular smooth muscle in vivo with AAV9 was inefficient. In contrast, an octo-guanidine conjugated antisense oligonucleotide targeting the 5' splice site of Mypt1 E24 was highly efficient in vivo. It reduced the percent splicing inclusion of Mypt1 E24 from 80% to 10% in mesenteric arteries. The maximal and half-maximal effects occurred at 12.5 and 6.25 mg/kg, respectively. The effect persisted for at least 1 mo without toxicity. This highly effective splice-blocking antisense oligonucleotide could be developed as a novel therapy to reverse vascular dysfunction common to diseases such as hypertension and heart failure.NEW & NOTEWORTHY Alternative exon usage is a major driver of phenotypic diversity in all cell types including smooth muscle. However, the functional significance of most of the hundreds of thousands of alternative exons has not been defined, nor in most cases even tested. If their importance to vascular function were known these alternative exons could represent novel therapeutic targets. Here, we present injection of Vivo-morpholino splice-blocking antisense oligonucleotides as a simple, efficient, and cost-effective method for suppression of alternative exon usage in vascular smooth muscle in vivo.


Subject(s)
Muscle, Smooth, Vascular , Oligonucleotides, Antisense , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Phosphoprotein Phosphatases/metabolism , Exons , Protein Isoforms/metabolism , Alternative Splicing , Phosphorylation
2.
Physiol Rep ; 11(21): e15844, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37960982

ABSTRACT

Smoothelins are cytoskeletal proteins with a single C-terminal calponin homology domain type 2 (CHD2). Little is known about the significance of variation in SMTN CHD2 domains, addressed here through analysis of public databases. A conserved 152 nt penultimate constitutive exon present in all SMTNs encodes helices II-IV of CHD2 with high identity (nt/aa 63/65%). Variable CHD2s of SMTN (helices IV-VI) are generated by alternative splicing of 165 nt exon E20. E20 and the CHD2 it encodes have high homology with the terminal constitutive exon of SMTNL1 (E8; nt/aa 72/75% identity). Unique to these CHD2 variants are a conserved extended nine amino acid C-terminal tail containing KTKK ubiquitination motifs. When E20 of SMTN is skipped (SMTN E20-), constitutive terminal E21 codes for helices IV-VI of CHD2. SMTN E21 has high identity with the terminal exon of SMTNL2 (E8; nt/aa 75/81% identity of aligned sequences) except for coding for a unique extended C-terminus (24 nt; 8aa) conserved only in mammals. SMTN isoform expression is tissue-specific: SMTNE20- and SMTNE20+ are highly expressed in SMC and non-muscle cells, respectively, while SMTNL1 + 2 are highly expressed in skeletal muscle cells. Tissue-specific expression of SMTN CHD2s with unique helices IV-VI suggest tissue-specific functions that require further study.


Subject(s)
Microfilament Proteins , Muscle Proteins , Animals , Muscle Proteins/genetics , Muscle Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cytoskeletal Proteins/metabolism , Mammals/metabolism , Calponins
3.
Cell Mol Life Sci ; 79(8): 459, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35913515

ABSTRACT

Differentiation of smooth muscle cells (SMCs) depends on serum response factor (SRF) and its co-activator myocardin (MYOCD). The role of MYOCD for the SMC program of gene transcription is well established. In contrast, the role of MYOCD in control of SMC-specific alternative exon usage, including exon splicing, has not been explored. In the current work we identified four splicing factors (MBNL1, RBPMS, RBPMS2, and RBFOX2) that correlate with MYOCD across human SMC tissues. Forced expression of MYOCD family members in human coronary artery SMCs in vitro upregulated expression of these splicing factors. For global profiling of transcript diversity, we performed RNA-sequencing after MYOCD transduction. We analyzed alternative transcripts with three different methods. Exon-based analysis identified 1637 features with differential exon usage. For example, usage of 3´ exons in MYLK that encode telokin increased relative to 5´ exons, as did the 17 kDa telokin to 130 kDa MYLK protein ratio. Dedicated event-based analysis identified 239 MYOCD-driven splicing events. Events involving MBNL1, MCAM, and ACTN1 were among the most prominent, and this was confirmed using variant-specific PCR analyses. In support of a role for RBPMS and RBFOX2 in MYOCD-driven splicing we found enrichment of their binding motifs around differentially spliced exons. Moreover, knockdown of either RBPMS or RBFOX2 antagonized splicing events stimulated by MYOCD, including those involving ACTN1, VCL, and MBNL1. Supporting an in vivo role of MYOCD-SRF-driven splicing, we demonstrate altered Rbpms expression and splicing in inducible and SMC-specific Srf knockout mice. We conclude that MYOCD-SRF, in part via RBPMS and RBFOX2, induce a program of differential exon usage and alternative splicing as part of the broader program of SMC differentiation.


Subject(s)
Alternative Splicing , Myocytes, Smooth Muscle , Alternative Splicing/genetics , Animals , Cell Differentiation/genetics , Exons/genetics , Humans , Mice , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Repressor Proteins/metabolism , Trans-Activators
4.
Am J Physiol Regul Integr Comp Physiol ; 322(4): R281-R291, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35107022

ABSTRACT

Alternative splicing of exon24 (E24) of myosin phosphatase targeting subunit 1 (Mypt1) by setting sensitivity to nitric oxide (NO)/cGMP-mediated relaxation is a key determinant of smooth muscle function. Here we defined expression of myosin phosphatase (MP) subunits and isoforms by creation of new genetic mouse models, assay of human and mouse tissues, and query of public databases. A Mypt1-LacZ reporter mouse revealed that Mypt1 transcription is turned on early in development during smooth muscle differentiation. Mypt1 is not as tightly restricted in its expression as smooth muscle myosin heavy chain (Myh11) and its E6 splice variant. Mypt1 is enriched in mature smooth versus nonmuscle cells. The E24 splice variant and leucine zipper minus protein isoform that it encodes is enriched in phasic versus tonic smooth muscle. In the vascular system, E24 splicing increases as vessel size decreases. In the gastrointestinal system, E24 splicing is most predominant in smooth muscle of the small intestine. Tissue-specific expression of MP subunits and Mypt1 E24 splicing is conserved in humans, whereas a splice variant of the inhibitory subunit (CPI-17) is unique to humans. A Mypt1 E24 mini-gene splicing reporter mouse generated to define patterns of E24 splicing in smooth muscle cells (SMCs) dispersed throughout the organ systems was unsuccessful. In summary, expression of Mypt1 and splicing of E24 is part of the program of smooth muscle differentiation, is further enhanced in phasic smooth muscle, and is conserved in humans. Its low-level expression in nonmuscle cells may confound its measurement in tissue samples.


Subject(s)
Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Myosin-Light-Chain Phosphatase , Animals , Cyclic GMP/metabolism , Humans , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin-Light-Chain Phosphatase/genetics , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
Pflugers Arch ; 473(4): 611-622, 2021 04.
Article in English | MEDLINE | ID: mdl-33145641

ABSTRACT

Alternative splicing of exon 24 (E24) of the myosin phosphatase regulatory subunit (Mypt1) tunes smooth muscle sensitivity to NO/cGMP-mediated vasorelaxation and thereby controls blood pressure (BP) in otherwise normal mice. This occurs via the toggling in or out of a C-terminal leucine zipper (LZ) motif required for hetero-dimerization with and activation by cGMP-dependent protein kinase cGK1α. Here we tested the hypothesis that editing (deletion) of E24, by shifting to the LZ positive isoform of Mypt1, would suppress the hypertensive response to angiotensin II (AngII). To test this, mice underwent tamoxifen-inducible and smooth muscle-specific deletion of E24 (E24 cKO) at age 6 weeks followed by a chronic slow-pressor dose of AngII (400 ng/kg/min) plus additional stressors. E24 cKO suppressed the hypertensive response to AngII alone or with the addition of a high salt diet. This effect was not a function of altered salt balance as there were no differences in intake or renal excretion of sodium. This effect was NO dependent as L-NAME in the drinking water caused an exaggerated hypertensive response in the E24cKO mice. E24cKO mouse mesenteric arteries were more sensitive to DEA/NO-induced vasorelaxation and less responsive to AngII- and α-adrenergic-induced vasoconstriction at baseline. Only the latter two effects were still present after 2 weeks of chronic AngII treatment. We conclude that editing of Mypt1 E24, by shifting the expression of naturally occurring isoforms and sensitizing to NO-mediated vasodilation, could be a novel approach to the treatment of human hypertension.


Subject(s)
Angiotensin II/metabolism , Hypertension/metabolism , Myosin-Light-Chain Phosphatase/genetics , Nitric Oxide/metabolism , Vasodilation , Animals , Hypertension/genetics , Hypertension/physiopathology , Leucine Zippers , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Mutation , Myosin-Light-Chain Phosphatase/chemistry , Myosin-Light-Chain Phosphatase/metabolism
6.
Birth Defects Res ; 110(20): 1517-1530, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30576091

ABSTRACT

Low oxygen concentration (hypoxia) is part of normal embryonic development, yet the situation is complex. Oxygen (O2 ) is a janus gas with low levels signaling through hypoxia-inducible transcription factor (HIF) that are required for development of fetal and placental vasculature and fetal red blood cells. This results in coupling of fetus and mother around midgestation as a functional feto-placental unit (FPU) for O2 transport, which is required for continued growth and development of the fetus. Defects in these processes may leave the developing fetus vulnerable to O2 deprivation or other stressors during this critical midgestational transition when common septal and conotruncal heart defects (CHDs) are likely to arise. Recent human epidemiological and case-control studies support an association between placental dysfunction, manifest as early onset pre-eclampsia (PE) and increased serum bio-markers, and CHD. Animal studies support this association, in particular those using gene inactivation in the mouse. Sophisticated methods for gene inactivation, cell fate mapping, and a quantitative bio-reporter of O2 concentration support the premise that hypoxic stress at critical stages of development leads to CHD. The secondary heart field contributing to the cardiac outlet is a key target, with activation of the un-folded protein response and abrogation of FGF signaling or precocious activation of a cardiomyocyte transcriptional program for differentiation, suggested as mechanisms. These studies provide a strong foundation for further study of feto-placental coupling and hypoxic stress in the genesis of human CHD.


Subject(s)
Hypoxia/embryology , Maternal-Fetal Exchange/physiology , Oxygen/metabolism , Animals , Case-Control Studies , Disease Models, Animal , Female , Fetus/physiopathology , Gestational Age , Heart Defects, Congenital/etiology , Humans , Hypoxia/metabolism , Hypoxia/physiopathology , Mice , Oxygen/physiology , Placenta/metabolism , Placenta/physiopathology , Placentation/physiology , Pre-Eclampsia/etiology , Pregnancy , Prenatal Care , Rats , Signal Transduction
7.
Dev Biol ; 422(2): 171-185, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27940158

ABSTRACT

A critical transition occurs near mid-gestation of mammalian pregnancy. Prior to this transition, low concentrations of oxygen (hypoxia) signaling through Hypoxia Inducible Factor (HIF) functions as a morphogen for the placenta and fetal organs. Subsequently, functional coupling of the placenta and fetal cardiovascular system for oxygen (O2) transport is required to support the continued growth and development of the fetus. Here we tested the hypothesis that Hif-1α is required in maternal cells for placental morphogenesis and function. We used Tamoxifen-inducible Cre-Lox to inactivate Hif-1α in maternal tissues at E8.5 (MATcKO), and used ODD-Luciferase as a reporter of hypoxia in placenta and fetal tissues. MATcKO of Hif-1α reduced the number of uterine natural killer (uNK) cells and Tpbpa-positve trophoblast cells in the maternal decidua at E13.5 -15.5. There were dynamic changes in all three layers of E13.5-15.5 MATcKO placenta. Of note was the under-development of the labyrinth at E15.5 associated with reduced Ki67 and increased TUNEL staining consistent with reduced cell proliferation and increased apoptosis. Labyrinth defects were particularly evident in placentas connected to effectively HIF-1α heterozygous null embryos. MATcKO had no effect on basal ODD-Luciferase activity in fetal organs (heart, liver, brain) at any stage, but at E13.5-15.5 resulted in enhanced induction of the ODD-Luciferase hypoxia reporter when the dam's inspired O2 was reduced to 8% for 4 hours. MATcKO also slowed the growth after E13.5 of fetuses that were effectively heterozygous for Hif-1α, with most being non-viable at E15.5. The hearts of these E15.5 fetuses were abnormal with reduction in size, thickened epicardium and mesenchymal septum. We conclude that maternal HIF-1α is required for placentation including recruitment of uNK and trophoblast cells into the maternal decidua and other trophoblast cell behaviors. The placental defects render the fetus vulnerable to O2 deprivation after mid-gestation.


Subject(s)
Cell Hypoxia/physiology , Heart/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Placenta/embryology , Placentation/genetics , Animals , Apoptosis , Cell Proliferation , Female , Heart/growth & development , Heart Defects, Congenital/embryology , In Situ Nick-End Labeling , Killer Cells, Natural/immunology , Mice , Oxygen/metabolism , Placenta/abnormalities , Placenta/cytology , Placentation/physiology , Pregnancy , Pregnancy Proteins/metabolism , Trophoblasts/metabolism
8.
Am J Physiol Heart Circ Physiol ; 310(11): H1715-24, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27084390

ABSTRACT

The cGMP activated kinase cGK1α is targeted to its substrates via leucine zipper (LZ)-mediated heterodimerization and thereby mediates vascular smooth muscle (VSM) relaxation. One target is myosin phosphatase (MP), which when activated by cGK1α results in VSM relaxation even in the presence of activating calcium. Variants of MP regulatory subunit Mypt1 are generated by alternative splicing of the 31 nt exon 24 (E24), which, by changing the reading frame, codes for isoforms that contain or lack the COOH-terminal LZ motif (E24+/LZ-; E24-/LZ+). Expression of these isoforms is vessel specific and developmentally regulated, modulates in disease, and is proposed to confer sensitivity to nitric oxide (NO)/cGMP-mediated vasorelaxation. To test this, mice underwent Tamoxifen-inducible and smooth muscle-specific knockout of E24 (E24 cKO) after weaning. Deletion of a single allele of E24 (shift to Mypt1 LZ+) enhanced vasorelaxation of first-order mesenteric arteries (MA1) to diethylamine-NONOate (DEA/NO) and to cGMP in permeabilized and calcium-clamped arteries and lowered blood pressure. There was no further effect of deletion of both E24 alleles, indicating high sensitivity to shift of Mypt1 isoforms. However, a unique property of MA1s from homozygous E24 cKOs was significantly reduced force generation to α-adrenergic activation. Furthermore 2 wk of high-salt (4% NaCl) diet increased MA1 force generation to phenylephrine in control mice, a response that was markedly suppressed in the E24 cKO homozygotes. Thus Mypt1 E24 splice variants tune arterial reactivity and could be worthy targets for lowering vascular resistance in disease states.


Subject(s)
Mesenteric Arteries/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Vasodilation/drug effects , Alleles , Alternative Splicing , Animals , Mesenteric Arteries/drug effects , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Phosphatase/genetics , Protein Isoforms/metabolism , Sodium Chloride/pharmacology
9.
Am J Physiol Heart Circ Physiol ; 309(9): H1468-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26371173

ABSTRACT

We examined the effect of stress in the first 2 wk of life induced by brief periods of daily maternal separation on developmental programming of rat small resistance mesenteric arteries (MAs). In MAs of littermate controls, mRNAs encoding mediators of vasoconstriction, including the α1a-adrenergic receptor, smooth muscle myosin heavy chain, and CPI-17, the inhibitory subunit of myosin phosphatase, increased from after birth through sexual [postnatal day (PND) 35] and full maturity, up to ∼80-fold, as measured by quantitative PCR. This was commensurate with two- to fivefold increases in maximum force production to KCl depolarization, calcium, and the α-adrenergic agonist phenylephrine, and increasing systolic blood pressure. Rats exposed to maternal separation stress as neonates had markedly accelerated trajectories of maturation of arterial contractile gene expression and function measured at PND14 or PND21 (weaning), 1 wk after the end of the stress protocol. This was suppressed by the α-adrenergic receptor blocker terazosin (0.5 mg·kg ip(-1)·day(-1)), indicating dependence on stress activation of sympathetic signaling. Due to the continued maturation of MAs in control rats, by sexual maturity (PND35) and into adulthood, no differences were observed in arterial function or response to a second stressor in rats stressed as neonates. Thus early life stress misprograms resistance artery smooth muscle, increasing vasoconstrictor function and blood pressure. This effect wanes in later stages, suggesting plasticity during arterial maturation. Further studies are indicated to determine whether stress in different periods of arterial maturation may cause misprogramming persisting through maturity and the potential salutary effect of α-adrenergic blockade in suppression of this response.


Subject(s)
Blood Pressure/genetics , Gene Expression Regulation, Developmental , Maternal Deprivation , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism , Stress, Psychological/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Blood Pressure/drug effects , Mesenteric Arteries/growth & development , Muscle Proteins/genetics , Muscle, Smooth, Vascular/growth & development , Myosin Heavy Chains/genetics , Myosin-Light-Chain Kinase/genetics , Phenylephrine/pharmacology , Phosphoproteins/genetics , Prazosin/analogs & derivatives , Prazosin/pharmacology , Protein Phosphatase 1/genetics , Rats , Receptors, Adrenergic, alpha-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Psychological/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/genetics
10.
Circ Cardiovasc Genet ; 8(5): 665-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26232087

ABSTRACT

BACKGROUND: Oxidative stress is manifested in embryos exposed to maternal diabetes mellitus, yet specific mechanisms for diabetes mellitus-induced heart defects are not defined. Gene deletion of intermediates of Wingless-related integration (Wnt) signaling causes heart defects similar to those observed in embryos from diabetic pregnancies. We tested the hypothesis that diabetes mellitus-induced oxidative stress impairs Wnt signaling, thereby causing heart defects, and that these defects can be rescued by transgenic overexpression of the reactive oxygen species scavenger superoxide dismutase 1 (SOD1). METHODS AND RESULTS: Wild-type (WT) and SOD1-overexpressing embryos from nondiabetic WT control dams and nondiabetic/diabetic WT female mice mated with SOD1 transgenic male mice were analyzed. No heart defects were observed in WT and SOD1 embryos under nondiabetic conditions. WT embryos of diabetic dams had a 26% incidence of cardiac outlet defects that were suppressed by SOD1 overexpression. Insulin treatment reduced blood glucose levels and heart defects. Diabetes mellitus increased superoxide production, canonical Wnt antagonist expression, caspase activation, and apoptosis and suppressed cell proliferation. Diabetes mellitus suppressed Wnt signaling intermediates and Wnt target gene expression in the embryonic heart, each of which were reversed by SOD1 overexpression. Hydrogen peroxide and peroxynitrite mimicked the inhibitory effect of high glucose on Wnt signaling, which was abolished by the SOD1 mimetic, tempol. CONCLUSIONS: The oxidative stress of diabetes mellitus impairs Wnt signaling and causes cardiac outlet defects that are rescued by SOD1 overexpression. This suggests that targeting of components of the Wnt5a signaling pathway may be a viable strategy for suppression of congenital heart defects in fetuses of diabetic pregnancies.


Subject(s)
Heart Defects, Congenital/genetics , Oxidative Stress , Pregnancy in Diabetics , Superoxide Dismutase/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis , Diabetes Complications/genetics , Diabetes Mellitus, Experimental , Female , Gene Expression , Heart/embryology , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/metabolism , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Transgenic , Peroxynitrous Acid/pharmacology , Pregnancy , Signal Transduction/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Wnt Proteins/genetics , Wnt-5a Protein
11.
Physiol Rep ; 3(7)2015 Jul.
Article in English | MEDLINE | ID: mdl-26156969

ABSTRACT

Deep sequencing of RNA samples from rat small mesenteric arteries (MA) and aorta (AO) identified common and unique features of their gene programs. ~5% of mRNAs were quantitatively differentially expressed in MA versus AO. Unique transcriptional control in MA smooth muscle is suggested by the selective or enriched expression of transcription factors Nkx2-3, HAND2, and Tcf21 (Capsulin). Enrichment in AO of PPAR transcription factors and their target genes of mitochondrial function, lipid metabolism, and oxidative phosphorylation is consistent with slow (oxidative) tonic smooth muscle. In contrast MA was enriched in contractile and calcium channel mRNAs suggestive of components of fast (glycolytic) phasic smooth muscle. Myosin phosphatase regulatory subunit paralogs Mypt1 and p85 were expressed at similar levels, while smooth muscle MLCK was the only such kinase expressed, suggesting functional redundancy of the former but not the latter in accordance with mouse knockout studies. With regard to vaso-regulatory signals, purinergic receptors P2rx1 and P2rx5 were reciprocally expressed in MA versus AO, while the olfactory receptor Olr59 was enriched in MA. Alox15, which generates the EDHF HPETE, was enriched in MA while eNOS was equally expressed, consistent with the greater role of EDHF in the smaller arteries. mRNAs that were not expressed at a level consistent with impugned function include skeletal myogenic factors, IKK2, nonmuscle myosin, and Gnb3. This screening analysis of gene expression in the small mesenteric resistance arteries suggests testable hypotheses regarding unique aspects of small artery function in the regional control of blood flow.

12.
Am J Physiol Heart Circ Physiol ; 308(9): H1039-50, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25724497

ABSTRACT

Microcirculatory dysfunction may cause tissue malperfusion and progression to organ failure in the later stages of sepsis, but the role of smooth muscle contractile dysfunction is uncertain. Mice were given intraperitoneal LPS, and mesenteric arteries were harvested at 6-h intervals for analyses of gene expression and contractile function by wire myography. Contractile (myosin and actin) and regulatory [myosin light chain kinase and phosphatase subunits (Mypt1, CPI-17)] mRNAs and proteins were decreased in mesenteric arteries at 24 h concordant with reduced force generation to depolarization, Ca(2+), and phenylephrine. Vasodilator sensitivity to DEA/nitric oxide (NO) and cGMP under Ca(2+) clamp were increased at 24 h after LPS concordant with a switch to Mypt1 exon 24- splice variant coding for a leucine zipper (LZ) motif required for PKG-1α activation of myosin phosphatase. This was reproduced by smooth muscle-specific deletion of Mypt1 exon 24, causing a shift to the Mypt1 LZ+ isoform. These mice had significantly lower resting blood pressure than control mice but similar hypotensive responses to LPS. The vasodilator sensitivity of wild-type mice to DEA/NO, but not cGMP, was increased at 6 h after LPS. This was abrogated in mice with a redox dead version of PKG-1α (Cys42Ser). Enhanced vasorelaxation in early endotoxemia is mediated by redox signaling through PKG-1α but in later endotoxemia by myosin phosphatase isoform shifts enhancing sensitivity to NO/cGMP as well as smooth muscle atrophy. Muscle atrophy and modulation may be a novel target to suppress microcirculatory dysfunction; however, inactivation of inducible NO synthase, treatment with the IL-1 antagonist IL-1ra, or early activation of α-adrenergic signaling did not suppressed this response.


Subject(s)
Lipopolysaccharides , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Sepsis/enzymology , Signal Transduction , Vasodilation , Animals , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/deficiency , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Genotype , Intracellular Signaling Peptides and Proteins , Isoenzymes , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Muscle Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Muscular Atrophy/chemically induced , Muscular Atrophy/enzymology , Muscular Atrophy/physiopathology , Myosin-Light-Chain Kinase/deficiency , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase/genetics , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Oxidation-Reduction , Phenotype , Phosphoproteins/genetics , RNA, Messenger/metabolism , Sepsis/chemically induced , Sepsis/genetics , Sepsis/physiopathology , Signal Transduction/drug effects , Time Factors , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
13.
Am J Physiol Cell Physiol ; 308(4): C289-96, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25428883

ABSTRACT

Diversity of smooth muscle within the vascular system is generated by alternative splicing of exons, yet there is limited understanding of its timing or control mechanisms. We examined splicing of myosin phosphatase regulatory subunit (Mypt1) exon 24 (E24) in relation to smooth muscle myosin heavy chain (Smmhc) and smoothelin (Smtn) alternative exons (Smmhc E6 and Smtn E20) during maturation of mouse mesenteric artery (MA) smooth muscle. The role of transformer 2ß (Tra2ß), a master regulator of splicing in flies, in maturation of arterial smooth muscle was tested through gene inactivation. Splicing of alternative exons in bladder smooth muscle was examined for comparative purposes. MA smooth muscle maturation began after postnatal week 2 and was complete at maturity, as indicated by switching to Mypt1 E24+ and Smtn E20- splice variants and 11-fold induction of Smmhc. Similar changes in bladder were complete by postnatal day 3. Splicing of Smmhc E6 was temporally dissociated from Mypt1 E24 and Smtn E20 and discordant between arteries and bladder. Tamoxifen-induced smooth muscle-specific inactivation of Tra2ß within the first week of life but not in maturity reduced splicing of Mypt1 E24 in MAs. Inactivation of Tra2ß causing a switch to the isoform of MYPT1 containing the COOH-terminal leucine zipper motif (E24-) increased arterial sensitivity to cGMP-mediated relaxation. In conclusion, maturation of mouse MA smooth muscle begins postnatally and continues until sexual maturity. TRA2ß is required for specification during this period of maturation, and its inactivation alters the contractile properties of mature arterial smooth muscle.


Subject(s)
Alternative Splicing , Cell Differentiation , Exons , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Myosin-Light-Chain Kinase/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Age Factors , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental , Genotype , In Vitro Techniques , Male , Mesenteric Arteries/enzymology , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Smooth Muscle Myosins/genetics , Smooth Muscle Myosins/metabolism , Vasodilation , Vasodilator Agents/pharmacology
14.
Microvasc Res ; 98: 166-71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24534069

ABSTRACT

Myosin phosphatase (MP) is a key target of signaling pathways that regulate smooth muscle tone and blood flow. Alternative splicing of MP targeting subunit (MYPT1) exon 24 (E24) generates isoforms with variable presence of a C-terminal leucine zipper (LZ) required for activation of MP by NO/cGMP. Here we examined the expression of MP and associated genes in a disease model in the coronary circulation. Female Yucatan miniature swine remained sedentary or were exercise-trained beginning eight weeks after placement of an ameroid constrictor around the left circumflex (LCX) artery. Fourteen weeks later epicardial arteries (~1mm) and resistance arterioles (~125 µm) were harvested and assayed for gene expression. MYPT1 isoforms were distinct in the epicardial arteries (E24-/LZ+) and resistance arterioles (E24+/LZ-) and unchanged by exercise training or coronary occlusion. MYPT1, CPI-17 and PDE5 mRNA levels were not different between arteries and arterioles while Kir2.1 and eNOS were 6.6-fold and 3.9-fold higher in the arterioles. There were no significant changes in transcript abundance in epicardial arteries of the collateralized (LCX) vs. non-occluded left anterior descending (LAD) territories, or in exercise-trained vs. sedentary pigs. There was a significant 1.2 fold increase in CPI-17 in collateral-dependent arterioles, independent of exercise, and a significant 1.7 fold increase in PDE5 in arterioles from exercise-trained pigs, independent of occlusion. We conclude that differences in MYPT1 E24 (LZ) isoforms, eNOS, and Kir2.1 distinguish epicardial arteries and resistance coronary arterioles. Up-regulation of coronary arteriolar PDE5 by exercise and CPI-17 by chronic occlusion could contribute to altered vasomotor responses and requires further study.


Subject(s)
Coronary Occlusion/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Physical Conditioning, Animal , Alternative Splicing , Animals , Arterioles/metabolism , Base Sequence , Coronary Circulation , Disease Models, Animal , Female , Humans , Isoenzymes/metabolism , Sequence Homology, Nucleic Acid , Species Specificity , Swine , Swine, Miniature
15.
Am J Physiol Regul Integr Comp Physiol ; 307(3): R256-70, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24898838

ABSTRACT

Variability in myosin phosphatase (MP) subunits may provide specificity in signaling pathways that regulate muscle tone. We utilized public databases and computational algorithms to investigate the phylogenetic diversity of MP regulatory (PPP1R12A-C) and inhibitory (PPP1R14A-D) subunits. The comparison of exonic coding sequences and expression data confirmed or refuted the existence of isoforms and their tissue-specific expression in different model organisms. The comparison of intronic and exonic sequences identified potential expressional regulatory elements. As examples, smooth muscle MP regulatory subunit (PPP1R12A) is highly conserved through evolution. Its alternative exon E24 is present in fish through mammals with two invariant features: 1) a reading frame shift generating a premature termination codon and 2) a hexanucleotide sequence adjacent to the 3' splice site hypothesized to be a novel suppressor of exon splicing. A characteristic of the striated muscle MP regulatory subunit (PPP1R12B) locus is numerous and phylogenetically variable transcriptional start sites. In fish this locus only codes for the small (M21) subunit, suggesting the primordial function of this gene. Inhibitory subunits show little intragenic variability; their diversity is thought to have arisen by expansion and tissue-specific expression of different gene family members. We demonstrate differences in the regulatory landscape between smooth muscle enriched (PPP1R14A) and more ubiquitously expressed (PPP1R14B) family members and identify deeply conserved intronic sequence and predicted transcriptional cis-regulatory elements. This bioinformatic and computational study has uncovered a number of attributes of MP subunits that supports selection of ideal model organisms and testing of hypotheses regarding their physiological significance and regulated expression.


Subject(s)
Biodiversity , Computational Biology , Computer Simulation , Myosin-Light-Chain Phosphatase/analysis , Myosin-Light-Chain Phosphatase/genetics , Protein Subunits/analysis , Protein Subunits/genetics , Amino Acid Sequence , Animals , Base Sequence , Chickens , Databases, Protein , Diptera , Humans , Mice , Models, Biological , Models, Genetic , Molecular Sequence Data , Myosin-Light-Chain Phosphatase/chemistry , Oligochaeta , Phylogeny , Protein Subunits/chemistry , Zebrafish
16.
Am J Physiol Heart Circ Physiol ; 307(4): H563-73, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24929853

ABSTRACT

There is evidence for developmental origins of vascular dysfunction yet little understanding of maturation of vascular smooth muscle (VSM) of regional circulations. We measured maturational changes in expression of myosin phosphatase (MP) and the broader VSM gene program in relation to mesenteric small resistance artery (SRA) function. We then tested the role of the sympathetic nervous system (SNS) in programming of SRAs and used genetically engineered mice to define the role of MP isoforms in the functional maturation of the mesenteric circulation. Maturation of rat mesenteric SRAs as measured by qPCR and immunoblotting begins after the second postnatal week and is not complete until maturity. It is characterized by induction of markers of VSM differentiation (smMHC, γ-, α-actin), CPI-17, an inhibitory subunit of MP and a key target of α-adrenergic vasoconstriction, α1-adrenergic, purinergic X1, and neuropeptide Y1 receptors of sympathetic signaling. Functional correlates include maturational increases in α-adrenergic-mediated force and calcium sensitization of force production (MP inhibition) measured in first-order mesenteric arteries ex vivo. The MP regulatory subunit Mypt1 E24+/LZ- isoform is specifically upregulated in SRAs during maturation. Conditional deletion of mouse Mypt1 E24 demonstrates that splicing of E24 causes the maturational reduction in sensitivity to cGMP-mediated vasorelaxation (MP activation). Neonatal chemical sympathectomy (6-hydroxydopamine) suppresses maturation of SRAs with minimal effect on a conduit artery. Mechanical denervation of the mature rat renal artery causes a reversion to the immature gene program. We conclude that the SNS captures control of the mesenteric circulation by programming maturation of the SRA smooth muscle.


Subject(s)
Gene Expression Regulation, Developmental , Mesenteric Arteries/metabolism , Renal Artery/metabolism , Sympathetic Nervous System/physiology , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation , Cyclic GMP/metabolism , Male , Mesenteric Arteries/growth & development , Mesenteric Arteries/innervation , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase/genetics , Myosin-Light-Chain Phosphatase/metabolism , Rats , Rats, Sprague-Dawley , Renal Artery/growth & development , Renal Artery/innervation , Sympathetic Nervous System/growth & development , Vasoconstriction , Vasodilator Agents/pharmacology
17.
J Am Heart Assoc ; 3(3): e000841, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855117

ABSTRACT

BACKGROUND: The heart develops under reduced and varying oxygen concentrations, yet there is little understanding of oxygen metabolism in the normal and mal-development of the heart. Here we used a novel reagent, the ODD-Luc hypoxia reporter mouse (oxygen degradation domain, ODD) of Hif-1α fused to Luciferase (Luc), to assay the activity of the oxygen sensor, prolyl hydroxylase, and oxygen reserve, in the developing heart. We tested the role of hypoxia-dependent responses in heart development by targeted inactivation of Hif-1α. METHODS AND RESULTS: ODD-Luciferase activity was 14-fold higher in mouse embryonic day 10.5 (E10.5) versus adult heart and liver tissue lysates. ODD-Luc activity decreased in 2 stages, the first corresponding with the formation of a functional cardiovascular system for oxygen delivery at E15.5, and the second after birth consistent with complete oxygenation of the blood and tissues. Reduction of maternal inspired oxygen to 8% for 4 hours caused minimal induction of luciferase activity in the maternal tissues but robust induction in the embryonic tissues in proportion to the basal activity, indicating a lack of oxygen reserve, and corresponding induction of a hypoxia-dependent gene program. Bioluminescent imaging of intact embryos demonstrated highest activity in the outflow portion of the E13.5 heart. Hif-1α inactivation or prolonged hypoxia caused outflow and septation defects only when targeted to this specific developmental window. CONCLUSIONS: Low oxygen concentrations and lack of oxygen reserve during a critical phase of heart organogenesis may provide a basis for vulnerability to the development of common septation and conotruncal heart defects.


Subject(s)
Heart Defects, Congenital/etiology , Hypoxia/embryology , Animals , Heart/embryology , Heart Defects, Congenital/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myocardium/chemistry , Myocardium/pathology , Oxygen/analysis , Real-Time Polymerase Chain Reaction
19.
Cell Rep ; 7(3): 634-44, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24767998

ABSTRACT

Autosomal dominant polycystic kidney disease is a common form of inherited kidney disease that is caused by mutations in two genes, PKD1 (polycystin-1) and PKD2 (polycystin-2). Mice with germline deletion of either gene die in midgestation with a vascular phenotype that includes profound edema. Although an endothelial cell defect has been suspected, the basis of this phenotype remains poorly understood. Here, we demonstrate that edema in Pkd1- and Pkd2-null mice is likely to be caused by defects in lymphatic development. Pkd1 and Pkd2 mutant embryos exhibit reduced lymphatic vessel density and vascular branching along with aberrant migration of early lymphatic endothelial cell precursors. We used cell-based assays to confirm that PKD1- and PKD2-depleted endothelial cells have an intrinsic defect in directional migration that is associated with a failure to establish front-rear polarity. Our studies reveal a role for polycystin signaling in lymphatic development.


Subject(s)
Endothelial Cells/cytology , Lymph Nodes/embryology , Signal Transduction , TRPP Cation Channels/metabolism , Animals , Cell Movement , Cell Polarity , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymph Nodes/metabolism , Lymphatic Vessels/embryology , Lymphatic Vessels/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , RNA Interference , RNA, Small Interfering/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
20.
Microcirculation ; 21(3): 239-48, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24112301

ABSTRACT

The dephosphorylation of myosin by the MP causes smooth muscle relaxation. MP is also a key target of signals that regulate vascular tone and thus blood flow and pressure. Here, we review studies from the past two decades that support the hypothesis that the regulated expression of MP subunits is a critical determinant of smooth muscle responses to constrictor and dilator signals. In particular, the highly regulated splicing of the regulatory subunit Mypt1 Exon 24 is proposed to tune sensitivity to NO/cGMP-mediated relaxation. The regulated transcription of the MP inhibitory subunit CPI-17 is proposed to determine sensitivity to agonist-mediated constriction. The expression of these subunits is specific in the microcirculation and varies in developmental and disease contexts. To date, the relationship between MP subunit expression and vascular function in these different contexts is correlative; confirmation of the hypothesis will require the generation of genetically engineered mice to test the role of MP subunits and their isoforms in the specificity of vascular smooth muscle responses to constrictor and dilator signals.


Subject(s)
Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Phosphatase/metabolism , Animals , Calcium Signaling , Humans , Intracellular Signaling Peptides and Proteins , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Microvessels/enzymology , Microvessels/physiology , Models, Cardiovascular , Muscle Contraction/physiology , Muscle Proteins , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase/chemistry , Myosin-Light-Chain Phosphatase/genetics , Phosphoproteins , Protein Subunits , Signal Transduction , Vasoconstriction/physiology , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...