Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carlos Hidalgo is active.

Publication


Featured researches published by Carlos Hidalgo.


Circulation Research | 2009

PKC Phosphorylation of Titin's PEVK Element. A Novel and Conserved Pathway for Modulating Myocardial Stiffness

Carlos Hidalgo; Bryan D. Hudson; Julius Bogomolovas; Yi Zhu; Brian Anderson; Marion L. Greaser; Siegfried Labeit; Henk Granzier

Rationale: Protein kinase C (PKC) regulates contractility of cardiac muscle cells by phosphorylating thin- and thick- filament–based proteins. Myocardial sarcomeres also contain a third myofilament, titin, and it is unknown whether titin can be phosphorylated by PKC and whether it affects passive tension. Objective: The purpose of this study was to examine the effect of PKC on titin phosphorylation and titin-based passive tension. Methods and Results: Phosphorylation assays with PKC&agr; revealed that titin is phosphorylated in skinned myocardial tissues; this effect is exacerbated by pretreating with protein phosphatase 1. In vitro phosphorylation of recombinant protein representing titin’s spring elements showed that PKC&agr; targets the proline – glutamate – valine – lysine (PEVK) spring element. Furthermore, mass spectrometry in combination with site-directed mutagenesis identified 2 highly conserved sites in the PEVK region that are phosphorylated by PKC&agr; (S11878 and S12022); when these 2 sites are mutated to alanine, phosphorylation is effectively abolished. Mechanical experiments with skinned left ventricular myocardium revealed that PKC&agr; significantly increases titin-based passive tension, an effect that is reversed by protein phosphatase 1. Single molecule force-extension curves show that PKC&agr; decreases the PEVK persistence length (from 1.20 nm to 0.55 nm), without altering the contour length, and using a serially-linked wormlike chain model we show that this increases titin-based passive force with a sarcomere length dependence that is similar to that measured in skinned myocardium after PKC&agr; phosphorylation. Conclusions: PKC phosphorylation of titin is a novel and conserved pathway that links myocardial signaling and myocardial stiffness.


Circulation | 2013

Right Ventricular Diastolic Impairment in Patients With Pulmonary Arterial Hypertension

Silvia Rain; M. Louis Handoko; Pia Trip; C. Tji-Joong Gan; Nico Westerhof; Ger J.M. Stienen; Walter J. Paulus; C. Ottenheijm; J. Tim Marcus; Peter Dorfmüller; Christophe Guignabert; Marc Humbert; P. Macdonald; Cris dos Remedios; Piet E. Postmus; Chandra Saripalli; Carlos Hidalgo; Henk Granzier; Anton Vonk-Noordegraaf; Jolanda van der Velden; Frances S. de Man

Background— The role of right ventricular (RV) diastolic stiffness in pulmonary arterial hypertension (PAH) is not well established. Therefore, we investigated the presence and possible underlying mechanisms of RV diastolic stiffness in PAH patients. Methods and Results— Single-beat RV pressure-volume analyses were performed in 21 PAH patients and 7 control subjects to study RV diastolic stiffness. Data are presented as mean±SEM. RV diastolic stiffness (&bgr;) was significantly increased in PAH patients (PAH, 0.050±0.005 versus control, 0.029±0.003; P<0.05) and was closely associated with disease severity. Subsequently, we searched for possible underlying mechanisms using RV tissue of PAH patients undergoing heart/lung transplantation and nonfailing donors. Histological analyses revealed increased cardiomyocyte cross-sectional areas (PAH, 453±31 &mgr;m2 versus control, 218±21 &mgr;m2; P<0.001), indicating RV hypertrophy. In addition, the amount of RV fibrosis was enhanced in PAH tissue (PAH, 9.6±0.7% versus control, 7.2±0.6%; P<0.01). To investigate the contribution of stiffening of the sarcomere (the contractile apparatus of RV cardiomyocytes) to RV diastolic stiffness, we isolated and membrane-permeabilized single RV cardiomyocytes. Passive tension at different sarcomere lengths was significantly higher in PAH patients compared with control subjects (>200%; Pinteraction<0.001), indicating stiffening of RV sarcomeres. An important regulator of sarcomeric stiffening is the sarcomeric protein titin. Therefore, we investigated titin isoform composition and phosphorylation. No alterations were observed in titin isoform composition (N2BA/N2B ratio: PAH, 0.78±0.07 versus control, 0.91±0.08), but titin phosphorylation in RV tissue of PAH patients was significantly reduced (PAH, 0.16±0.01 arbitrary units versus control, 0.20±0.01 arbitrary units; P<0.05). Conclusions— RV diastolic stiffness is significantly increased in PAH patients, with important contributions from increased collagen and intrinsic stiffening of the RV cardiomyocyte sarcomeres.


Neurology | 2013

Recessive truncating titin gene, TTN, mutations presenting as centronuclear myopathy.

Ozge Ceyhan-Birsoy; Pankaj B. Agrawal; Carlos Hidalgo; Klaus Schmitz-Abe; Elizabeth T. DeChene; Lindsay C. Swanson; Rachel Soemedi; Nasim Vasli; Susan T. Iannaccone; Perry B. Shieh; Natasha Shur; Jane M. Dennison; Michael W. Lawlor; Jocelyn Laporte; Kyriacos Markianos; William G. Fairbrother; Henk Granzier; Alan H. Beggs

Objective: To identify causative genes for centronuclear myopathies (CNM), a heterogeneous group of rare inherited muscle disorders that often present in infancy or early life with weakness and hypotonia, using next-generation sequencing of whole exomes and genomes. Methods: Whole-exome or -genome sequencing was performed in a cohort of 29 unrelated patients with clinicopathologic diagnoses of CNM or related myopathy depleted for cases with mutations of MTM1, DNM2, and BIN1. Immunofluorescence analyses on muscle biopsies, splicing assays, and gel electrophoresis of patient muscle proteins were performed to determine the molecular consequences of mutations of interest. Results: Autosomal recessive compound heterozygous truncating mutations of the titin gene, TTN, were identified in 5 individuals. Biochemical analyses demonstrated increased titin degradation and truncated titin proteins in patient muscles, establishing the impact of the mutations. Conclusions: Our study identifies truncating TTN mutations as a cause of congenital myopathy that is reported as CNM. Unlike the classic CNM genes that are all involved in excitation-contraction coupling at the triad, TTN encodes the giant sarcomeric protein titin, which forms a myofibrillar backbone for the components of the contractile machinery. This study expands the phenotypic spectrum associated with TTN mutations and indicates that TTN mutation analysis should be considered in cases of possible CNM without mutations in the classic CNM genes.


Journal of Clinical Investigation | 2012

Mouse and computational models link Mlc2v dephosphorylation to altered myosin kinetics in early cardiac disease.

Farah Sheikh; Kunfu Ouyang; Stuart G. Campbell; Robert C. Lyon; Joyce Chuang; Dan Fitzsimons; Jared Tangney; Carlos Hidalgo; Charles S. Chung; Hongqiang Cheng; Nancy D. Dalton; Yusu Gu; Hideko Kasahara; Majid Ghassemian; Jeffrey H. Omens; Kirk L. Peterson; Henk Granzier; Richard L. Moss; Andrew D. McCulloch; Ju Chen

Actin-myosin interactions provide the driving force underlying each heartbeat. The current view is that actin-bound regulatory proteins play a dominant role in the activation of calcium-dependent cardiac muscle contraction. In contrast, the relevance and nature of regulation by myosin regulatory proteins (for example, myosin light chain-2 [MLC2]) in cardiac muscle remain poorly understood. By integrating gene-targeted mouse and computational models, we have identified an indispensable role for ventricular Mlc2 (Mlc2v) phosphorylation in regulating cardiac muscle contraction. Cardiac myosin cycling kinetics, which directly control actin-myosin interactions, were directly affected, but surprisingly, Mlc2v phosphorylation also fed back to cooperatively influence calcium-dependent activation of the thin filament. Loss of these mechanisms produced early defects in the rate of cardiac muscle twitch relaxation and ventricular torsion. Strikingly, these defects preceded the left ventricular dysfunction of heart disease and failure in a mouse model with nonphosphorylatable Mlc2v. Thus, there is a direct and early role for Mlc2 phosphorylation in regulating actin-myosin interactions in striated muscle contraction, and dephosphorylation of Mlc2 or loss of these mechanisms can play a critical role in heart failure.


Journal of Biological Chemistry | 2009

Nebulin Alters Cross-bridge Cycling Kinetics and Increases Thin Filament Activation A NOVEL MECHANISM FOR INCREASING TENSION AND REDUCING TENSION COST

Murali Chandra; Ranganath Mamidi; Steven J. Ford; Carlos Hidalgo; Christian Witt; C. Ottenheijm; Siegfried Labeit; Henk Granzier

Nebulin is a giant filamentous F-actin-binding protein (∼800 kDa) that binds along the thin filament of the skeletal muscle sarcomere. Nebulin is one of the least well understood major muscle proteins. Although nebulin is usually viewed as a structural protein, here we investigated whether nebulin plays a role in muscle contraction by using skinned muscle fiber bundles from a nebulin knock-out (NEB KO) mouse model. We measured force-pCa (−log[Ca2+]) and force-ATPase relations, as well as the rate of tension re-development (ktr) in tibialis cranialis muscle fibers. To rule out any alterations in troponin (Tn) isoform expression and/or status of Tn phosphorylation, we studied fiber bundles that had been reconstituted with bacterially expressed fast skeletal muscle recombinant Tn. We also performed a detailed analysis of myosin heavy chain, myosin light chain, and myosin light chain 2 phosphorylation, which showed no significant differences between wild type and NEB KO. Our mechanical studies revealed that NEB KO fibers had increased tension cost (5.9 versus 4.4 pmol millinewtons−1 mm−1 s−1) and reductions in ktr (4.7 versus 7.3 s−1), calcium sensitivity (pCa50 5.74 versus 5.90), and cooperativity of activation (nH 3.64 versus 4.38). Our findings indicate the following: 1) in skeletal muscle nebulin increases thin filament activation, and 2) through altering cross-bridge cycling kinetics, nebulin increases force and efficiency of contraction. These novel properties of nebulin add a new level of understanding of skeletal muscle function and provide a mechanism for the severe muscle weakness in patients with nebulin-based nemaline myopathy.


Circulation | 2014

Experimentally increasing titin compliance in a novel mouse model attenuates the Frank-Starling mechanism but has a beneficial effect on diastole.

Mei Methawasin; Kirk R. Hutchinson; Eun-Jeong Lee; John E. Smith; Chandra Saripalli; Carlos Hidalgo; C. Ottenheijm; Henk Granzier

Background— Experimentally upregulating compliant titins has been suggested as a therapeutic for lowering pathological diastolic stiffness levels. However, how increasing titin compliance impacts global cardiac function requires in-depth study. We investigate the effect of upregulating compliant titins in a novel mouse model with a genetically altered titin splicing factor; integrative approaches were used from intact cardiomyocyte mechanics to pressure-volume analysis and Doppler echocardiography. Methods and Results— Compliant titins were upregulated through deletion of the RNA Recognition Motif of the splicing factor RBM20 (Rbm20&Dgr;RRMmice). A genome-wide exon expression analysis and a candidate approach revealed that the phenotype is likely to be dominated by greatly increased lengths of titin’s spring elements. At both cardiomyocyte and left ventricular chamber levels, diastolic stiffness was reduced in heterozygous (+/−) Rbm20&Dgr;RRMmice with a further reduction in homozygous (−/−) mice at only the intact myocyte level. Fibrosis was present in only −/− Rbm20&Dgr;RRM hearts. The Frank-Starling Mechanism was reduced in a graded fashion in Rbm20&Dgr;RRM mice, at both the cardiomyocyte and left ventricular chamber levels. Exercise tests revealed an increase in exercise capacity in +/− mice. Conclusions— Titin is not only important in diastolic but also in systolic cardiac function. Upregulating compliant titins reduces diastolic chamber stiffness owing to the increased compliance of myocytes, but it depresses end-systolic elastance; under conditions of exercise, the beneficial effects on diastolic function dominate. Therapeutic manipulation of the RBM20-based splicing system might be able to minimize effects on fibrosis and systolic function while improving the diastolic function in patients with heart failure.


Circulation | 2013

Shortening of the Elastic Tandem Immunoglobulin Segment of Titin Leads to Diastolic Dysfunction

Charles S. Chung; Kirk R. Hutchinson; Mei Methawasin; Chandra Saripalli; John E. Smith; Carlos Hidalgo; Xiuju Luo; Siegfried Labeit; Caiying Guo; Henk Granzier

Background— Diastolic dysfunction is a poorly understood but clinically pervasive syndrome that is characterized by increased diastolic stiffness. Titin is the main determinant of cellular passive stiffness. However, the physiological role that the tandem immunoglobulin (Ig) segment of titin plays in stiffness generation and whether shortening this segment is sufficient to cause diastolic dysfunction need to be established. Methods and Results— We generated a mouse model in which 9 Ig-like domains (Ig3–Ig11) were deleted from the proximal tandem Ig segment of the spring region of titin (IG KO). Exon microarray analysis revealed no adaptations in titin splicing, whereas novel phospho-specific antibodies did not detect changes in titin phosphorylation. Passive myocyte stiffness was increased in the IG KO, and immunoelectron microscopy revealed increased extension of the remaining titin spring segments as the sole likely underlying mechanism. Diastolic stiffness was increased at the tissue and organ levels, with no consistent changes in extracellular matrix composition or extracellular matrix–based passive stiffness, supporting a titin-based mechanism for in vivo diastolic dysfunction. Additionally, IG KO mice have a reduced exercise tolerance, a phenotype often associated with diastolic dysfunction. Conclusions— Increased titin-based passive stiffness is sufficient to cause diastolic dysfunction with exercise intolerance.


Circulation | 2013

Shortening of Titin's Elastic Tandem Ig Segment Leads to Diastolic Dysfunction

Charles S. Chung; Kirk R. Hutchinson; Mei Methawasin; Chandra Saripalli; John E. Smith; Carlos Hidalgo; Xiuju Luo; Siegfried Labeit; Caiying Guo; Henk Granzier

Background— Diastolic dysfunction is a poorly understood but clinically pervasive syndrome that is characterized by increased diastolic stiffness. Titin is the main determinant of cellular passive stiffness. However, the physiological role that the tandem immunoglobulin (Ig) segment of titin plays in stiffness generation and whether shortening this segment is sufficient to cause diastolic dysfunction need to be established. Methods and Results— We generated a mouse model in which 9 Ig-like domains (Ig3–Ig11) were deleted from the proximal tandem Ig segment of the spring region of titin (IG KO). Exon microarray analysis revealed no adaptations in titin splicing, whereas novel phospho-specific antibodies did not detect changes in titin phosphorylation. Passive myocyte stiffness was increased in the IG KO, and immunoelectron microscopy revealed increased extension of the remaining titin spring segments as the sole likely underlying mechanism. Diastolic stiffness was increased at the tissue and organ levels, with no consistent changes in extracellular matrix composition or extracellular matrix–based passive stiffness, supporting a titin-based mechanism for in vivo diastolic dysfunction. Additionally, IG KO mice have a reduced exercise tolerance, a phenotype often associated with diastolic dysfunction. Conclusions— Increased titin-based passive stiffness is sufficient to cause diastolic dysfunction with exercise intolerance.


Trends in Cardiovascular Medicine | 2013

Tuning the molecular giant titin through phosphorylation: Role in health and disease

Carlos Hidalgo; Henk Granzier

Titin is a giant multi-functional filament that spans half of the sarcomere. Titins extensible I-band region functions as a molecular spring that provides passive stiffness to cardiac myocytes. Elevated diastolic stiffness is found in a large fraction of heart failure patients and thus understanding the normal mechanisms and pathophysiology of passive stiffness modulation is clinically important. Here we provide first a brief general background on titin including what is known about titin isoforms and then focus on recently discovered post-translational modifications of titin that alter passive stiffness. We discuss the various kinases that have been shown to phosphorylate titin and address the possible roles of titin phosphorylation in cardiac disease, including heart failure with preserved ejection fraction (HFpEF).


Journal of Molecular and Cellular Cardiology | 2013

The multifunctional Ca2+/calmodulin-dependent protein kinase II delta (CaMKIIδ) phosphorylates cardiac titin’s spring elements

Carlos Hidalgo; Charles S. Chung; Chandra Saripalli; Mei Methawasin; Kirk R. Hutchinson; George Tsaprailis; Siegfried Labeit; Alicia Mattiazzi; Henk Granzier

Titin-based passive stiffness is post-translationally regulated by several kinases that phosphorylate specific spring elements located within titins elastic I-band region. Whether titin is phosphorylated by calcium/calmodulin dependent protein kinase II (CaMKII), an important regulator of cardiac function and disease, has not been addressed. The aim of this work was to determine whether CaMKIIδ, the predominant CaMKII isoform in the heart, phosphorylates titin, and to use phosphorylation assays and mass spectrometry to study which of titins spring elements might be targeted by CaMKIIδ. It was found that CaMKIIδ phosphorylates titin in mouse LV skinned fibers, that the CaMKIIδ sites can be dephosphorylated by protein phosphatase 1 (PP1), and that under baseline conditions, in both intact isolated hearts and skinned myocardium, about half of the CaMKIIδ sites are phosphorylated. Mass spectrometry revealed that both the N2B and PEVK segments are targeted by CaMKIIδ at several conserved serine residues. Whether phosphorylation of titin by CaMKIIδ occurs in vivo, was tested in several conditions using back phosphorylation assays and phospho-specific antibodies to CaMKIIδ sites. Reperfusion following global ischemia increased the phosphorylation level of CaMKIIδ sites on titin and this effect was abolished by the CaMKII inhibitor KN-93. No changes in the phosphorylation level of the PEVK element were found suggesting that the increased phosphorylation level of titin in IR (ischemia reperfusion) might be due to phosphorylation of the N2B element. The findings of these studies show for the first time that titin can be phosphoryalated by CaMKIIδ, both in vitro and in vivo, and that titins molecular spring region that determines diastolic stiffness is a target of CaMKIIδ.

Collaboration


Dive into the Carlos Hidalgo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Ottenheijm

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael Gotthardt

Max Delbrück Center for Molecular Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge