1
|
Gentile F, Orlando G, Montuoro S, Ferrari Chen YF, Macefield V, Passino C, Giannoni A, Emdin M. Treating heart failure by targeting the vagus nerve. Heart Fail Rev 2024; 29:1201-1215. [PMID: 39117958 PMCID: PMC11455679 DOI: 10.1007/s10741-024-10430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Increased sympathetic and reduced parasympathetic nerve activity is associated with disease progression and poor outcomes in patients with chronic heart failure. The demonstration that markers of autonomic imbalance and vagal dysfunction, such as reduced heart rate variability and baroreflex sensitivity, hold prognostic value in patients with chronic heart failure despite modern therapies encourages the research for neuromodulation strategies targeting the vagus nerve. However, the approaches tested so far have yielded inconclusive results. This review aims to summarize the current knowledge about the role of the parasympathetic nervous system in chronic heart failure, describing the pathophysiological background, the methods of assessment, and the rationale, limits, and future perspectives of parasympathetic stimulation either by drugs or bioelectronic devices.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy.
| | - Giulia Orlando
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Yu Fu Ferrari Chen
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | | | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
2
|
Ferreira M, Cunha PS, Felix AC, Fonseca H, Oliveira M, Laranjo S, Rocha I. AFTER-CA: Autonomic Function Transformation and Evaluation Following Catheter Ablation in Atrial Fibrillation. J Clin Med 2024; 13:5796. [PMID: 39407858 PMCID: PMC11476626 DOI: 10.3390/jcm13195796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Catheter ablation (CA) is a well-established treatment for atrial fibrillation (AF). However, its effects on autonomic function and underlying mechanisms remain poorly understood. This study investigated autonomic and haemodynamic changes following CA and explored their potential implications for patient outcomes. Methods: Seventy-eight patients with AF underwent CA and were followed up at one, three, and six months. Autonomic function was assessed using a combination of head-up tilt (HUT), handgrip (HG), and deep breathing (DB) manoeuvres along with baroreflex sensitivity (BRS) and baroreflex effectiveness index (BEI) evaluation. Heart rate (HR), blood pressure (BP), and their variability were measured at each time point. Results: Significant autonomic alterations were observed after ablation, particularly at one month, with reductions in parasympathetic tone and baroreflex function. These changes gradually normalised by six months. Both pulmonary vein isolation (PVI) and cryoablation (CryO) had similar effects on autonomic regulation. Improvements in quality of life, measured by the AFEQT scores, were consistent with these physiological changes. Conclusions: CA for AF induces significant time-dependent autonomic and haemodynamic changes with recovery over six months. These findings underscore the need for ongoing monitoring and personalised post-ablation management. Further research is required to explore the mechanisms driving these alterations and their long-term impacts on patient outcomes.
Collapse
Affiliation(s)
- Monica Ferreira
- Faculdade de Medicina and Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (M.O.)
| | - Pedro Silva Cunha
- Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (P.S.C.); (H.F.)
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
| | - Ana Clara Felix
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
| | - Helena Fonseca
- Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (P.S.C.); (H.F.)
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
| | - Mario Oliveira
- Faculdade de Medicina and Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (M.O.)
- Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (P.S.C.); (H.F.)
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
| | - Sergio Laranjo
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Isabel Rocha
- Faculdade de Medicina and Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (M.O.)
| |
Collapse
|
3
|
Skeete J, Gordon JS, Kavinksy L, Huang HD, Aksu T. Cardioneuroablation for the management of neurally mediated syncope, sinus bradycardia, and atrioventricular block. J Interv Card Electrophysiol 2024:10.1007/s10840-024-01923-7. [PMID: 39327368 DOI: 10.1007/s10840-024-01923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Through several decades of medical advances, we have improved our understanding of the role of the autonomic nervous system in the production of a myriad of clinical cardiac conditions such as vasovagal syncope, situational syncope, carotid sinus hypersensitivity, vagally mediated sinus bradycardia, and atrioventricular block. While typically not associated with mortality, these common clinical entities may result in significant patient symptoms and morbidity and are often characterized by a frustrating treatment course with a paucity of effective strategies. In recent years, there has been increased interest in the management of these conditions via direct modulation of the parasympathetic component of the autonomic nervous system. This is achieved by targeting the ganglionated plexus central to the pathogenesis of these conditions via cardioneuroablation. The primary role of this strategy is evolving and serves to augment traditional treatment strategies such as lifestyle modification and pharmacotherapy. In this review, we examine the principles governing the role of cardioneuroablation in select populations with vasovagal syncope, sinus dysfunction, and atrioventricular block including the evolving evidence in this exciting field while keeping in mind the need for robust clinical studies examining the long-term effectiveness and safety.
Collapse
Affiliation(s)
- Jamario Skeete
- Division of Cardiology, Rush University Medical Center, 1717 West Congress Parkway, Chicago, IL, 60612, USA
| | - Jonathan S Gordon
- Division of Cardiology, Rush University Medical Center, 1717 West Congress Parkway, Chicago, IL, 60612, USA
| | - Lincoln Kavinksy
- Division of Cardiology, Rush University Medical Center, 1717 West Congress Parkway, Chicago, IL, 60612, USA
| | - Henry D Huang
- Division of Cardiology, Rush University Medical Center, 1717 West Congress Parkway, Chicago, IL, 60612, USA
| | - Tolga Aksu
- Department of Cardiology, Yeditepe University Hospital, Istanbul, 34100, Turkey.
| |
Collapse
|
4
|
Quigley KS, Gianaros PJ, Norman GJ, Jennings JR, Berntson GG, de Geus EJC. Publication guidelines for human heart rate and heart rate variability studies in psychophysiology-Part 1: Physiological underpinnings and foundations of measurement. Psychophysiology 2024; 61:e14604. [PMID: 38873876 DOI: 10.1111/psyp.14604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 12/22/2023] [Accepted: 04/04/2024] [Indexed: 06/15/2024]
Abstract
This Committee Report provides methodological, interpretive, and reporting guidance for researchers who use measures of heart rate (HR) and heart rate variability (HRV) in psychophysiological research. We provide brief summaries of best practices in measuring HR and HRV via electrocardiographic and photoplethysmographic signals in laboratory, field (ambulatory), and brain-imaging contexts to address research questions incorporating measures of HR and HRV. The Report emphasizes evidence for the strengths and weaknesses of different recording and derivation methods for measures of HR and HRV. Along with this guidance, the Report reviews what is known about the origin of the heartbeat and its neural control, including factors that produce and influence HRV metrics. The Report concludes with checklists to guide authors in study design and analysis considerations, as well as guidance on the reporting of key methodological details and characteristics of the samples under study. It is expected that rigorous and transparent recording and reporting of HR and HRV measures will strengthen inferences across the many applications of these metrics in psychophysiology. The prior Committee Reports on HR and HRV are several decades old. Since their appearance, technologies for human cardiac and vascular monitoring in laboratory and daily life (i.e., ambulatory) contexts have greatly expanded. This Committee Report was prepared for the Society for Psychophysiological Research to provide updated methodological and interpretive guidance, as well as to summarize best practices for reporting HR and HRV studies in humans.
Collapse
Affiliation(s)
- Karen S Quigley
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA
| | - Peter J Gianaros
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Greg J Norman
- Department of Psychology, The University of Chicago, Chicago, Illinois, USA
| | - J Richard Jennings
- Department of Psychiatry & Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gary G Berntson
- Department of Psychology & Psychiatry, The Ohio State University, Columbus, Ohio, USA
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Read DF, Booth GT, Daza RM, Jackson DL, Gladden RG, Srivatsan SR, Ewing B, Franks JM, Spurrell CH, Gomes AR, O'Day D, Gogate AA, Martin BK, Larson H, Pfleger C, Starita L, Lin Y, Shendure J, Lin S, Trapnell C. Single-cell analysis of chromatin and expression reveals age- and sex-associated alterations in the human heart. Commun Biol 2024; 7:1052. [PMID: 39187646 PMCID: PMC11347658 DOI: 10.1038/s42003-024-06582-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2024] [Indexed: 08/28/2024] Open
Abstract
Sex differences and age-related changes in the human heart at the tissue, cell, and molecular level have been well-documented and many may be relevant for cardiovascular disease. However, how molecular programs within individual cell types vary across individuals by age and sex remains poorly characterized. To better understand this variation, we performed single-nucleus combinatorial indexing (sci) ATAC- and RNA-Seq in human heart samples from nine donors. We identify hundreds of differentially expressed genes by age and sex and find epigenetic signatures of variation in ATAC-Seq data in this discovery cohort. We then scale up our single-cell RNA-Seq analysis by combining our data with five recently published single nucleus RNA-Seq datasets of healthy adult hearts. We find variation such as metabolic alterations by sex and immune changes by age in differential expression tests, as well as alterations in abundance of cardiomyocytes by sex and neurons with age. In addition, we compare our adult-derived ATAC-Seq profiles to analogous fetal cell types to identify putative developmental-stage-specific regulatory factors. Finally, we train predictive models of cell-type-specific RNA expression levels utilizing ATAC-Seq profiles to link distal regulatory sequences to promoters, quantifying the predictive value of a simple TF-to-expression regulatory grammar and identifying cell-type-specific TFs. Our analysis represents the largest single-cell analysis of cardiac variation by age and sex to date and provides a resource for further study of healthy cardiac variation and transcriptional regulation at single-cell resolution.
Collapse
Affiliation(s)
- David F Read
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Gregory T Booth
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rula Green Gladden
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sanjay R Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brent Ewing
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Jennifer M Franks
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | | | - Diana O'Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Aishwarya A Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Haleigh Larson
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Christian Pfleger
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA
| | - Lea Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Yiing Lin
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Children's Research Institute, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Shin Lin
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA.
| | - Cole Trapnell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
6
|
Abu Nahia K, Sulej A, Migdał M, Ochocka N, Ho R, Kamińska B, Zagorski M, Winata CL. scRNA-seq reveals the diversity of the developing cardiac cell lineage and molecular players in heart rhythm regulation. iScience 2024; 27:110083. [PMID: 38872974 PMCID: PMC11170199 DOI: 10.1016/j.isci.2024.110083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/26/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
We utilized scRNA-seq to delineate the diversity of cell types in the zebrafish heart. Transcriptome profiling of over 50,000 cells at 48 and 72 hpf defined at least 18 discrete cell lineages of the developing heart. Utilizing well-established gene signatures, we identified a population of cells likely to be the primary pacemaker and characterized the transcriptome profile defining this critical cell type. Two previously uncharacterized genes, atp1b3b and colec10, were found to be enriched in the sinoatrial cardiomyocytes. CRISPR/Cas9-mediated knockout of these two genes significantly reduced heart rate, implicating their role in cardiac development and conduction. Additionally, we describe other cardiac cell lineages, including the endothelial and neural cells, providing their expression profiles as a resource. Our results established a detailed atlas of the developing heart, providing valuable insights into cellular and molecular mechanisms, and pinpointed potential new players in heart rhythm regulation.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Natalia Ochocka
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Richard Ho
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
- The Njord Centre, Department of Physics, University of Oslo, Oslo, Norway
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marcin Zagorski
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
| | | |
Collapse
|
7
|
Aydin MD, Kanat A, Sahin B, Sahin MH, Ergene S, Demirtas R. New experimental finding of dangerous autonomic ganglia changes in cardiac injury following subarachnoid hemorrhage; a reciprocal culprit-victim relationship between the brain and heart. Int J Neurosci 2024; 134:91-102. [PMID: 35658782 DOI: 10.1080/00207454.2022.2086128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The vagal, stellate, and cardiac ganglia cells changes following subarachnoid hemorrhage (SAH) may occur. This study aimed to investigate if there is any relation between vagal network/stellate ganglion and intrinsic cardiac ganglia insult following SAH. MATERIALS AND METHODS Twenty-six rabbits were used in this study. Animals were randomly divided as control (GI, n = 5); SHAM 0.75 cc of saline-injected (n = 5) and study with autologous 1.5 cc blood injection into their cisterna magna(GIII, n = 15). All animals were followed for three weeks and then decapitated. Their motor vagal nucleus, nodose, stellate, and intracardiac ganglion cells were estimated by stereological methods and compared statistically. RESULTS Numerical documents of heart-respiratory rates, vagal nerve- ICG, and stellate neuron densities as follows: 276 ± 32/min-22 ± 3/min-10.643 ± 1.129/mm3-4 ± 1/mm3-12 ± 3/mm3 and 2 ± 1/cm3 in the control group; 221 ± 22/min-16 ± 4/min-8.699 ± 976/mm3-24 ± 9/mm3-103 ± 32/mm3 and 11 ± 3/cm3 in the SHAM group; and 191 ± 23/min-17 ± 4/min-9.719 ± 932/mm3-124 ± 31/mm3-1.542 ± 162/mm3 and 32 ± 9/cm3 in the SAH (study) group. The animals with burned neuro-cardiac web had more neurons of stellate ganglia and a less normal neuron density of nodose ganglia (p < 0.005). CONCLUSION Sypathico-parasympathetic imbalance induced vagal nerve-ICG disruption following SAH could be named as Burned Neurocardiac Web syndrome in contrast to broken heart because ICG/parasympathetic network degeneration could not be detected in classic broken heart syndrome. It was noted that cardiac ganglion degeneration is more prominent in animals' severe degenerated neuron density of nodose ganglia. We concluded that the cardiac ganglia network knitted with vagal-sympathetic-somatosensitive fibers has an important in heart function following SAH. The neurodegeneration of the cardiac may occur in SAH, and cause sudden death.Graphical abstract[Formula: see text].
Collapse
Affiliation(s)
- Mehmet Dumlu Aydin
- Department of Neurosurgery, Medical Faculty, of Ataturk University, Erzurum, Turkey
| | - Ayhan Kanat
- Department of Neurosurgery, Medical Faculty of Recep Tayyip, Erdogan University, Rize, Turkey
| | - Balkan Sahin
- Department of Neurosurgery, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Hakan Sahin
- Department of Neurosurgery, Medical Faculty, of Ataturk University, Erzurum, Turkey
| | - Saban Ergene
- Department of Cardiovascular Surgery, Medical Faculty of Recep Tayyip, Erdogan University, Rize, Turkey
| | - Rabia Demirtas
- Department of Pathology, Medical Faculty, of Ataturk University, Erzurum, Turkey
| |
Collapse
|
8
|
Lee K, Vanin S, Nashed M, Sarikahya MH, Laviolette SR, Natale DRC, Hardy DB. Cannabidiol Exposure During Gestation Leads to Adverse Cardiac Outcomes Early in Postnatal Life in Male Rat Offspring. Cannabis Cannabinoid Res 2024; 9:781-796. [PMID: 38358335 DOI: 10.1089/can.2023.0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Introduction: Studies indicate that ∼7% of pregnant individuals in North America consume cannabis in pregnancy. Pre-clinical studies have established that maternal exposure to Δ9-tetrahydrocannabinol (THC; major psychoactive component in cannabis) leads to fetal growth restriction and impaired cardiac function in offspring. However, the effects of maternal exposure to cannabidiol (CBD; major non-euphoric constituent) on cardiac outcomes in offspring remain unknown. Therefore, our objective is to investigate the functional and underlying molecular impacts in the hearts of offspring exposed to CBD in pregnancy. Methods: Pregnant Wistar rats were exposed to either 3 or 30 mg/kg CBD or vehicle control i.p. daily from gestational day 6 to term. Echocardiography was used to assess cardiac function in male and female offspring at postnatal day (PND) 21. Furthermore, quantitative polymerase chain reaction (qPCR), immunoblotting, and bulk RNA-sequencing (RNA-seq) were performed on PND21 offspring hearts. Results: Despite no differences in the heart-to-body weight ratio, both doses of CBD led to reduced cardiac function exclusively in male offspring at 3 weeks of age. Underlying this, significant alterations in the expression of the endocannabinoid system (ECS; e.g., decreased cannabinoid receptor 2) were observed. In addition, bulk RNA-seq data demonstrated transcriptional pathways significantly enriched in mitochondrial function/metabolism as well as development. Conclusion: Collectively, we demonstrated for the first time that gestational exposure to CBD, a constituent perceived as safe, leads to early sex-specific postnatal cardiac deficits and alterations in the cardiac ECS in offspring.
Collapse
Affiliation(s)
- Kendrick Lee
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sebastian Vanin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Mina Nashed
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Mohammed Halit Sarikahya
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - David R C Natale
- Departments of Biomedical and Molecular Sciences and Obstetrics and Gynaecology, Queen's University, Kingston, Canada
| | - Daniel B Hardy
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Obstetrics and Gynecology, Children's Health Research Institute, Lawson Health Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
9
|
Garbilis A, Mednieks J. Differences in Heart Rate Variability in the Frequency Domain between Different Groups of Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:900. [PMID: 38929516 PMCID: PMC11205485 DOI: 10.3390/medicina60060900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Heart rate variability (HRV) is defined as a physiological variation in duration between sinus beats. The aim of this study was to research and analyze the HRV between various groups of patients. Materials and Methods: A retrospective study was conducted in an outpatient setting. Patients who had undergone a tilt-table test were selected for this study and were divided into three groups based on their self-reported health anamnesis: group 1 (n = 84, mean age 45.8 ± 17.8) consisted of patients with no known orthostatic intolerance or neurodegenerative disease, group 2 consisted of patients with a known or suspected orthostatic intolerance (n = 50, mean age 46.5 ± 18.6), and group 3 consisted of patients with a known or suspected neurodegenerative disorder (n = 29, mean age 55.6 ± 20.4). During the tilt-table test, HRV frequency-domain parameters-normalized low frequency (LFnu) and high frequency (HFnu), absolute powers-absolute low frequency (LF-RRI), absolute high frequency (HF-RRI), and LF/HF ratio-were recorded during 5 min rest in the supine position. Results: Group 1 had a reduced LFnu at 52.93% (SD: 18.00) compared to group 2 at 58.57% (18.06) and group 3 at 61.80% (SD: 17.74), and group 1 had increased HFnu: group 1-47.08% (SD: 17.97), group 2-41.41% (SD: 18.03), and group 3-38.16% (SD: 14.7). LFnu and HFnu differences were statistically significant (p < 0.05). LF-RRI was reported as follows: group 1-531.32 ms2 (SD: 578.57), group 2-346.2 ms2 (SD: 447.96), and group 3-143.21 ms2 (SD: 166.96). HF-RRI was reported as follows: group 1-835.87 ms2 (SD: 1625.42), group 2-297.46 ms2 (SD: 507.15), and group 3-70.83 ms2 (SD: 75.67). LF-RRI and HF-RRI comparisons between groups were statistically significant (p < 0.001). LF/HF ratios were reported as follows: group 1-1.91 (SD: 2.29), group 2-2.43 (SD: 2.33), and group 3-2.54 (SD: 2.17). LF/HF ratio comparisons between groups were statistically significant at p < 0.05. Conclusions: This study shows that patients with known or suspected orthostatic intolerance and neurodegenerative disorders have reduced HRV, possibly caused by reduced parasympathetic modulation. HRV in patients with known or suspected neurodegenerative disorders is reduced more severely than in patients with orthostatic disorders. Other studies in HRV have indicated a possible increase of risk in cardiovascular disorders in patients with reduced HRV, and therefore, HRV analysis could be a potential clinical diagnostic tool. However, the lack of universally agreed upon methodology, reference values, and possible external and internal factor influence hinders the introduction of HRV examinations into wider clinical practice.
Collapse
Affiliation(s)
- Artūrs Garbilis
- Department of Residency, Rīga Stradiņš University, Dzirciema 16, LV-1007 Rīga, Latvia
- Department of Neurology and Neurosurgery, Rīga Stradiņš University, Pilsoņu 13, LV-1002 Rīga, Latvia;
| | - Jānis Mednieks
- Department of Neurology and Neurosurgery, Rīga Stradiņš University, Pilsoņu 13, LV-1002 Rīga, Latvia;
| |
Collapse
|
10
|
Vdovichenko ND, Nikonorova PA. Effect of Direct and Mediated through Maternal Organism Activation of the Catecholaminergic System on Heart Rate Parameters in Rat Fetuses. Bull Exp Biol Med 2024; 176:533-538. [PMID: 38722506 DOI: 10.1007/s10517-024-06062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Indexed: 05/18/2024]
Abstract
We performed a comparative analysis of direct and mediated through the maternal organism effects of elevated catecholamine concentration on changes in the cardiac activity parameters in female rats and their fetuses on gestation days 18 and 20 under in vivo conditions. Administration of L-DOPA, a precursor of catecholaminergic transmitters, did not cause chronotropic effects in fetuses. Analysis of HR variability showed that in fetuses, irrespective of the administration route, there was an increase in nervous influences while the leading role of humoral-metabolic factors in the regulation of HR was preserved. In females receiving L-DOPA injection on day 18 of gestation, a decrease in humoral-metabolic and an increase in nerve effects were observed; in rats injected with L-DOPA on day 20 of gestation, an increase in sympathetic influences was found. Administration of L-DOPA to fetuses provoked a slight increase in the power of all components of the heart rhythm periodogram spectrum in females on day 18 of gestation and their decrease on day 20. Changes in the parameters of HR variability in females can confirm the hypothesis that in the "mother-fetus" system, the heart rhythm in the mother can be affected by both maternal and fetal influences presumably through the humoral-metabolic regulation.
Collapse
Affiliation(s)
- N D Vdovichenko
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.
| | - P A Nikonorova
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
11
|
Giannino G, Braia V, Griffith Brookles C, Giacobbe F, D'Ascenzo F, Angelini F, Saglietto A, De Ferrari GM, Dusi V. The Intrinsic Cardiac Nervous System: From Pathophysiology to Therapeutic Implications. BIOLOGY 2024; 13:105. [PMID: 38392323 PMCID: PMC10887082 DOI: 10.3390/biology13020105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Abstract
The cardiac autonomic nervous system (CANS) plays a pivotal role in cardiac homeostasis as well as in cardiac pathology. The first level of cardiac autonomic control, the intrinsic cardiac nervous system (ICNS), is located within the epicardial fat pads and is physically organized in ganglionated plexi (GPs). The ICNS system does not only contain parasympathetic cardiac efferent neurons, as long believed, but also afferent neurons and local circuit neurons. Thanks to its high degree of connectivity, combined with neuronal plasticity and memory capacity, the ICNS allows for a beat-to-beat control of all cardiac functions and responses as well as integration with extracardiac and higher centers for longer-term cardiovascular reflexes. The present review provides a detailed overview of the current knowledge of the bidirectional connection between the ICNS and the most studied cardiac pathologies/conditions (myocardial infarction, heart failure, arrhythmias and heart transplant) and the potential therapeutic implications. Indeed, GP modulation with efferent activity inhibition, differently achieved, has been studied for atrial fibrillation and functional bradyarrhythmias, while GP modulation with efferent activity stimulation has been evaluated for myocardial infarction, heart failure and ventricular arrhythmias. Electrical therapy has the unique potential to allow for both kinds of ICNS modulation while preserving the anatomical integrity of the system.
Collapse
Affiliation(s)
- Giuseppe Giannino
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Valentina Braia
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Carola Griffith Brookles
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Federico Giacobbe
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Fabrizio D'Ascenzo
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Filippo Angelini
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Andrea Saglietto
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Gaetano Maria De Ferrari
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| | - Veronica Dusi
- Cardiology, Department of Medical Sciences, University of Turin, 10124 Torino, Italy
- Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, 10126 Torino, Italy
| |
Collapse
|
12
|
Celotto C, Sánchez C, Abdollahpur M, Sandberg F, Rodriguez Mstas JF, Laguna P, Pueyo E. The frequency of atrial fibrillatory waves is modulated by the spatiotemporal pattern of acetylcholine release: a 3D computational study. Front Physiol 2024; 14:1189464. [PMID: 38235381 PMCID: PMC10791938 DOI: 10.3389/fphys.2023.1189464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 10/10/2023] [Indexed: 01/19/2024] Open
Abstract
In atrial fibrillation (AF), the ECG P-wave, which represents atrial depolarization, is replaced with chaotic and irregular fibrillation waves (f waves). The f-wave frequency, F f, shows significant variations over time. Cardiorespiratory interactions regulated by the autonomic nervous system have been suggested to play a role in such variations. We conducted a simulation study to test whether the spatiotemporal release pattern of the parasympathetic neurotransmitter acetylcholine (ACh) modulates the frequency of atrial reentrant circuits. Understanding parasympathetic involvement in AF may guide more effective treatment approaches and could help to design autonomic markers alternative to heart rate variability (HRV), which is not available in AF patients. 2D tissue and 3D whole-atria models of human atrial electrophysiology in persistent AF were built. Different ACh release percentages (8% and 30%) and spatial ACh release patterns, including spatially random release and release from ganglionated plexi (GPs) and associated nerves, were considered. The temporal pattern of ACh release, ACh(t), was simulated following a sinusoidal waveform of frequency 0.125 Hz to represent the respiratory frequency. Different mean concentrations ( A C h ¯ ) and peak-to-peak ranges of ACh (ΔACh) were tested. We found that temporal variations in F f, F f(t), followed the simulated temporal ACh(t) pattern in all cases. The temporal mean of F f(t), F ¯ f , depended on the fibrillatory pattern (number and location of rotors), the percentage of ACh release nodes and A C h ¯ . The magnitude of F f(t) modulation, ΔF f, depended on the percentage of ACh release nodes and ΔACh. The spatial pattern of ACh release did not have an impact on F ¯ f and only a mild impact on ΔF f. The f-wave frequency, being indicative of vagal activity, has the potential to drive autonomic-based therapeutic actions and could replace HRV markers not quantifiable from AF patients.
Collapse
Affiliation(s)
- Chiara Celotto
- BSICoS Group, I3A and IIS-Aragón, University of Zaragoza, Zaragoza, Spain
- CIBER - Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Carlos Sánchez
- BSICoS Group, I3A and IIS-Aragón, University of Zaragoza, Zaragoza, Spain
- CIBER - Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | | | - Frida Sandberg
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | | | - Pablo Laguna
- BSICoS Group, I3A and IIS-Aragón, University of Zaragoza, Zaragoza, Spain
- CIBER - Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Esther Pueyo
- BSICoS Group, I3A and IIS-Aragón, University of Zaragoza, Zaragoza, Spain
- CIBER - Bioingeniería, Biomateriales, y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
13
|
Olmsted ZT, Paredes-Espinosa MB, Paluh JL. Embryonic Spinal Cord Innervation in Human Trunk Organogenesis Gastruloids: Cardiac Versus Enteric Customization and Beyond. Methods Mol Biol 2024; 2767:135-159. [PMID: 37284941 DOI: 10.1007/7651_2023_491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Trunk-biased human gastruloids provide the ability to couple developmentally relevant spinal neurogenesis and organ morphogenesis via spatiotemporal self-organization events from derivatives of the three germ layers. The multi-lineage nature of gastruloids provides the full complexity of regulatory signaling cues that surpasses directed organoids and lays the foundation for an ex vivo self-evolving system. Here we detail two distinct protocols for trunk-biased gastruloids from an elongated, polarized structure with coordinated organ-specific neural patterning. Following an induction phase to caudalize iPSCs to trunk phenotype, divergent features of organogenesis and end-organ innervation yield separate models of enteric and cardiac nervous system formation. Both protocols are permissive to multi-lineage development and allow the study of neural integration events within a native, embryo-like context. We discuss the customizability of human gastruloids and the optimization of initial and extended conditions that maintain a permissive environment for multi-lineage differentiation and integration.
Collapse
Affiliation(s)
- Zachary T Olmsted
- State University of New York Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience, Albany, NY, USA
- University of California Los Angeles, Ronald Reagan UCLA Medical Center, Department of Neurosurgery, Los Angeles, CA, USA
| | - Maria Belen Paredes-Espinosa
- State University of New York Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience, Albany, NY, USA
| | - Janet L Paluh
- State University of New York Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience, Albany, NY, USA
| |
Collapse
|
14
|
Hsu IU, Lin Y, Guo Y, Xu QJ, Shao Y, Wang RL, Yin D, Zhao J, Young LH, Zhao H, Zhang L, Chang RB. Differential developmental blueprints of organ-intrinsic nervous systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571306. [PMID: 38168446 PMCID: PMC10759999 DOI: 10.1101/2023.12.12.571306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The organ-intrinsic nervous system is a major interface between visceral organs and the brain, mediating important sensory and regulatory functions in the body-brain axis and serving as critical local processors for organ homeostasis. Molecularly, anatomically, and functionally, organ-intrinsic neurons are highly specialized for their host organs. However, the underlying mechanism that drives this specialization is largely unknown. Here, we describe the differential strategies utilized to achieve organ-specific organization between the enteric nervous system (ENS) 1 and the intrinsic cardiac nervous system (ICNS) 2 , a neuronal network essential for heart performance but poorly characterized. Integrating high-resolution whole-embryo imaging, single-cell genomics, spatial transcriptomics, proteomics, and bioinformatics, we uncover that unlike the ENS which is highly mobile and colonizes the entire gastrointestinal (GI) tract, the ICNS uses a rich set of extracellular matrix (ECM) genes that match with surrounding heart cells and an intermediate dedicated neuronal progenitor state to stabilize itself for a 'beads-on-the-necklace' organization on heart atria. While ICNS- and ENS-precursors are genetically similar, their differentiation paths are influenced by their host-organs, leading to distinct mature neuron types. Co-culturing ENS-precursors with heart cells shifts their identity towards the ICNS and induces the expression of heart-matching ECM genes. Our cross-organ study thus reveals fundamental principles for the maturation and specialization of organ-intrinsic neurons.
Collapse
|
15
|
Mousavi MS, Meknatkhah S, Imani A, Geramifar P, Riazi G. Comparable assessment of adolescent repeated physical or psychological stress effects on adult cardiac performance in female rats. Sci Rep 2023; 13:16401. [PMID: 37775558 PMCID: PMC10541905 DOI: 10.1038/s41598-023-43721-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023] Open
Abstract
Extensive evidence highlights a robust connection between various forms of chronic stress and cardiovascular disease (CVD). In today's fast-paced world, with chronic stressors abound, CVD has emerged as a leading global cause of mortality. The intricate interplay of physical and psychological stressors triggers distinct neural networks within the brain, culminating in diverse health challenges. This study aims to discern the unique impacts of chronic physical and psychological stress on the cardiovascular system, unveiling their varying potencies in precipitating CVD. Twenty-one adolescent female rats were methodically assigned to three groups: (1) control (n = 7), (2) physical stress (n = 7), and (3) psychological stress (n = 7). Employing a two-compartment enclosure, stressors were administered to the experimental rats over five consecutive days, each session lasting 10 min. After a 1.5-month recovery period post-stress exposure, a trio of complementary techniques characterized by high specificity or high sensitivity were employed to meticulously evaluate CVD. Echocardiography and single-photon emission computed tomography (SPECT) were harnessed to scrutinize left ventricular architecture and myocardial viability, respectively. Subsequently, the rats were ethically sacrificed to facilitate heart removal, followed by immunohistochemistry staining targeting glial fibrillary acidic protein (GFAP). Rats subjected to psychological stress showed a wider range of significant cardiac issues compared to control rats. This included left ventricular hypertrophy [IVSd: 0.1968 ± 0.0163 vs. 0.1520 ± 0.0076, P < 0.05; LVPWd: 0.2877 ± 0.0333 vs. 0.1689 ± 0.0057, P < 0.01; LVPWs: 0.3180 ± 0.0382 vs. 0.2226 ± 0.0121, P < 0.05; LV-mass: 1.283 ± 0.0836 vs. 1.000 ± 0.0241, P < 0.01], myocardial ischemia [21.30% vs. 32.97%, P < 0.001], and neuroinflammation. This outcome underscores the imperative of prioritizing psychological well-being during adolescence, presenting a compelling avenue to curtail the prevalence of CVD in adulthood. Furthermore, extending such considerations to individuals grappling with CVD might prospectively enhance their overall quality of life.
Collapse
Affiliation(s)
- Monireh-Sadat Mousavi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Sogol Meknatkhah
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Alireza Imani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamhossein Riazi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
16
|
Sanchez-Alonso JL, Fedele L, Copier JS, Lucarelli C, Mansfield C, Judina A, Houser SR, Brand T, Gorelik J. Functional LTCC-β 2AR Complex Needs Caveolin-3 and Is Disrupted in Heart Failure. Circ Res 2023; 133:120-137. [PMID: 37313722 PMCID: PMC10321517 DOI: 10.1161/circresaha.123.322508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Beta-2 adrenergic receptors (β2ARs) but not beta-2 adrenergic receptors (β1ARs) form a functional complex with L-type Ca2+ channels (LTCCs) on the cardiomyocyte membrane. However, how microdomain localization in the plasma membrane affects the function of these complexes is unknown. We aim to study the coupling between LTCC and β adrenergic receptors in different cardiomyocyte microdomains, the distinct involvement of PKA and CAMKII (Ca2+/calmodulin-dependent protein kinase II) and explore how this functional complex is disrupted in heart failure. METHODS Global signaling between LTCCs and β adrenergic receptors was assessed with whole-cell current recordings and western blot analysis. Super-resolution scanning patch-clamp was used to explore the local coupling between single LTCCs and β1AR or β2AR in different membrane microdomains in control and failing cardiomyocytes. RESULTS LTCC open probability (Po) showed an increase from 0.054±0.003 to 0.092±0.008 when β2AR was locally stimulated in the proximity of the channel (<350 nm) in the transverse tubule microdomain. In failing cardiomyocytes, from both rodents and humans, this transverse tubule coupling between LTCC and β2AR was lost. Interestingly, local stimulation of β1AR did not elicit any change in the Po of LTCCs, indicating a lack of proximal functional interaction between the two, but we confirmed a general activation of LTCC via β1AR. By using blockers of PKA and CaMKII and a Caveolin-3-knockout mouse model, we conclude that the β2AR-LTCC regulation requires the presence of caveolin-3 and the activation of the CaMKII pathway. By contrast, at a cellular "global" level PKA plays a major role downstream β1AR and results in an increase in LTCC current. CONCLUSIONS Regulation of the LTCC activity by proximity coupling mechanisms occurs only via β2AR, but not β1AR. This may explain how β2ARs tune the response of LTCCs to adrenergic stimulation in healthy conditions. This coupling is lost in heart failure; restoring it could improve the adrenergic response of failing cardiomyocytes.
Collapse
Affiliation(s)
- Jose L. Sanchez-Alonso
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Laura Fedele
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Jaël S. Copier
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Carla Lucarelli
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Catherine Mansfield
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Aleksandra Judina
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Steven R. Houser
- Department of Physiology, Cardiovascular Research Center, Lewis Katz Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, United Kingdom (J.L.S.-A., L.F., J.S.C., C.L., C.M., A.J., T.B., J.G.)
| |
Collapse
|
17
|
Abstract
The cardiovascular system is hardwired to the brain via multilayered afferent and efferent polysynaptic axonal connections. Two major anatomically and functionally distinct though closely interacting subcircuits within the cardiovascular system have recently been defined: The artery-brain circuit and the heart-brain circuit. However, how the nervous system impacts cardiovascular disease progression remains poorly understood. Here, we review recent findings on the anatomy, structures, and inner workings of the lesser-known artery-brain circuit and the better-established heart-brain circuit. We explore the evidence that signals from arteries or the heart form a systemic and finely tuned cardiovascular brain circuit: afferent inputs originating in the arterial tree or the heart are conveyed to distinct sensory neurons in the brain. There, primary integration centers act as hubs that receive and integrate artery-brain circuit-derived and heart-brain circuit-derived signals and process them together with axonal connections and humoral cues from distant brain regions. To conclude the cardiovascular brain circuit, integration centers transmit the constantly modified signals to efferent neurons which transfer them back to the cardiovascular system. Importantly, primary integration centers are wired to and receive information from secondary brain centers that control a wide variety of brain traits encoded in engrams including immune memory, stress-regulating hormone release, pain, reward, emotions, and even motivated types of behavior. Finally, we explore the important possibility that brain effector neurons in the cardiovascular brain circuit network connect efferent signals to other peripheral organs including the immune system, the gut, the liver, and adipose tissue. The enormous recent progress vis-à-vis the cardiovascular brain circuit allows us to propose a novel neurobiology-centered cardiovascular disease hypothesis that we term the neuroimmune cardiovascular circuit hypothesis.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (C.Y.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| | - Cristina Godinho-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal (C.G.-S., H.V.-F.)
| | | | - Qian J Xu
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT (Q.J.X., R.B.C.)
| | - Rui B Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT (Q.J.X., R.B.C.)
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), Munich, Germany (S.K.M., C.Y., C.W., A.J.R.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (S.K.M., C.W., A.J.R.H.)
| |
Collapse
|
18
|
Gee MM, Lenhoff AM, Schwaber JS, Ogunnaike BA, Vadigepalli R. Closed-loop modeling of central and intrinsic cardiac nervous system circuits underlying cardiovascular control. AIChE J 2023; 69:e18033. [PMID: 37250861 PMCID: PMC10211393 DOI: 10.1002/aic.18033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/02/2023] [Indexed: 01/16/2023]
Abstract
The baroreflex is a multi-input, multi-output control physiological system that regulates blood pressure by modulating nerve activity between the brainstem and the heart. Existing computational models of the baroreflex do not explictly incorporate the intrinsic cardiac nervous system (ICN), which mediates central control of the heart function. We developed a computational model of closed-loop cardiovascular control by integrating a network representation of the ICN within central control reflex circuits. We examined central and local contributions to the control of heart rate, ventricular functions, and respiratory sinus arrhythmia (RSA). Our simulations match the experimentally observed relationship between RSA and lung tidal volume. Our simulations predicted the relative contributions of the sensory and the motor neuron pathways to the experimentally observed changes in the heart rate. Our closed-loop cardiovascular control model is primed for evaluating bioelectronic interventions to treat heart failure and renormalize cardiovascular physiology.
Collapse
Affiliation(s)
- Michelle M Gee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Abraham M Lenhoff
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - James S Schwaber
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Babatunde A Ogunnaike
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
| | - Rajanikanth Vadigepalli
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
19
|
Bizanti A, Zhang Y, Harden SW, Chen J, Hoover DB, Gozal D, Shivkumar K, Cheng ZJ. Catecholaminergic axon innervation and morphology in flat-mounts of atria and ventricles of mice. J Comp Neurol 2023; 531:596-617. [PMID: 36591925 PMCID: PMC10499115 DOI: 10.1002/cne.25444] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 01/03/2023]
Abstract
Sympathetic efferent axons regulate cardiac functions. However, the topographical distribution and morphology of cardiac sympathetic efferent axons remain insufficiently characterized due to the technical challenges involved in immunohistochemical labeling of the thick walls of the whole heart. In this study, flat-mounts of the left and right atria and ventricles of FVB mice were immunolabeled for tyrosine hydroxylase (TH), a marker of sympathetic nerves. Atrial and ventricular flat-mounts were scanned using a confocal microscope to construct montages. We found (1) In the atria: A few large TH-immunoreactive (IR) axon bundles entered both atria, branched into small bundles and then single axons that eventually formed very dense terminal networks in the epicardium, myocardium and inlet regions of great vessels to the atria. Varicose TH-IR axons formed close contact with cardiomyocytes, vessels, and adipocytes. Multiple intrinsic cardiac ganglia (ICG) were identified in the epicardium of both atria, and a subpopulation of the neurons in the ICG were TH-IR. Most TH-IR axons in bundles traveled through ICG before forming dense varicose terminal networks in cardiomyocytes. We did not observe varicose TH-IR terminals encircling ICG neurons. (2) In the left and right ventricles and interventricular septum: TH-IR axons formed dense terminal networks in the epicardium, myocardium, and vasculature. Collectively, TH labeling is achievable in flat-mounts of thick cardiac walls, enabling detailed mapping of catecholaminergic axons and terminal structures in the whole heart at single-cell/axon/varicosity scale. This approach provides a foundation for future quantification of the topographical organization of the cardiac sympathetic innervation in different pathological conditions.
Collapse
Affiliation(s)
- Ariege Bizanti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Yuanyuan Zhang
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Scott W Harden
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - David Gozal
- Department of Child Health and Child Health Research Institute, and Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Kalyanam Shivkumar
- Department of Medicine, Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, University of California, Los Angeles, California, USA
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
20
|
Sahoglu SG, Kazci YE, Karadogan B, Aydin MS, Nebol A, Turhan MU, Ozturk G, Cagavi E. High-resolution mapping of sensory fibers at the healthy and post-myocardial infarct whole transgenic hearts. J Neurosci Res 2023; 101:338-353. [PMID: 36517461 DOI: 10.1002/jnr.25150] [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: 07/07/2022] [Revised: 10/15/2022] [Accepted: 11/19/2022] [Indexed: 12/23/2022]
Abstract
The sensory nervous system is critical to maintain cardiac function. As opposed to efferent innervation, less is known about cardiac afferents. For this, we mapped the VGLUT2-expressing cardiac afferent fibers of spinal and vagal origin by using the VGLUT2::tdTomato double transgenic mouse as an approach to visualize the whole hearts both at the dorsal and ventral sides. For comparison, we colabeled mixed-sex transgenic hearts with either TUJ1 protein for global cardiac innervation or tyrosine hydroxylase for the sympathetic network at the healthy state or following ischemic injury. Interestingly, the nerve density for global and VGLUT2-expressing afferents was found significantly higher on the dorsal side compared to the ventral side. From the global nerve innervation detected by TUJ1 immunoreactivity, VGLUT2 afferent innervation was detected to be 15-25% of the total network. The detailed characterization of both the atria and the ventricles revealed a remarkable diversity of spinal afferent nerve ending morphologies of flower sprays, intramuscular endings, and end-net branches that innervate distinct anatomical parts of the heart. Using this integrative approach in a chronic myocardial infarct model, we showed a significant increase in hyperinnervation in the form of axonal sprouts for cardiac afferents at the infarct border zone, as well as denervation at distal sites of the ischemic area. The functional and physiological consequences of the abnormal sensory innervation remodeling post-ischemic injury should be further evaluated in future studies regarding their potential contribution to cardiac dysfunction.
Collapse
Affiliation(s)
- Sevilay Goktas Sahoglu
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Neuroscience Program, Institute of Health Sciences, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Yusuf Enes Kazci
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Neuroscience Program, Institute of Health Sciences, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Behnaz Karadogan
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Serif Aydin
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Aylin Nebol
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Medical Biology and Genetics Program, Institute of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Ugurcan Turhan
- Department of Cardiovascular Surgery, Cerrahpasa School of Medicine, Istanbul University, Istanbul, Turkey
| | - Gurkan Ozturk
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, İstanbul, Turkey
| | - Esra Cagavi
- Regenerative and Restorative Medical Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.,Medical Biology and Genetics Program, Institute of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
21
|
Huffman WJ, Musselman ED, Pelot NA, Grill WM. Measuring and modeling the effects of vagus nerve stimulation on heart rate and laryngeal muscles. Bioelectron Med 2023; 9:3. [PMID: 36797733 PMCID: PMC9936668 DOI: 10.1186/s42234-023-00107-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Reduced heart rate (HR) during vagus nerve stimulation (VNS) is associated with therapy for heart failure, but stimulation frequency and amplitude are limited by patient tolerance. An understanding of physiological responses to parameter adjustments would allow differential control of therapeutic and side effects. To investigate selective modulation of the physiological responses to VNS, we quantified the effects and interactions of parameter selection on two physiological outcomes: one related to therapy (reduced HR) and one related to side effects (laryngeal muscle EMG). METHODS We applied a broad range of stimulation parameters (mean pulse rates (MPR), intra-burst frequencies, and amplitudes) to the vagus nerve of anesthetized mice. We leveraged the in vivo recordings to parameterize and validate computational models of HR and laryngeal muscle activity across amplitudes and temporal patterns of VNS. We constructed a finite element model of excitation of fibers within the mouse cervical vagus nerve. RESULTS HR decreased with increased amplitude, increased MPR, and decreased intra-burst frequency. EMG increased with increased MPR. Preferential HR effects over laryngeal EMG effects required combined adjustments of amplitude and MPR. The model of HR responses highlighted contributions of ganglionic filtering to VNS-evoked changes in HR at high stimulation frequencies. Overlap in activation thresholds between small and large modeled fibers was consistent with the overlap in dynamic ranges of related physiological measures (HR and EMG). CONCLUSION The present study provides insights into physiological responses to VNS required for informed parameter adjustment to modulate selectively therapeutic effects and side effects.
Collapse
Affiliation(s)
- William J. Huffman
- Department of Biomedical Engineering, Duke University, Fitzpatrick CIEMAS, Box 90281, Room 1427, 101 Science Drive, Durham, NC 27708-0281 USA
| | - Eric D. Musselman
- Department of Biomedical Engineering, Duke University, Fitzpatrick CIEMAS, Box 90281, Room 1427, 101 Science Drive, Durham, NC 27708-0281 USA
| | - Nicole A. Pelot
- Department of Biomedical Engineering, Duke University, Fitzpatrick CIEMAS, Box 90281, Room 1427, 101 Science Drive, Durham, NC 27708-0281 USA
| | - Warren M. Grill
- Department of Biomedical Engineering, Duke University, Fitzpatrick CIEMAS, Box 90281, Room 1427, 101 Science Drive, Durham, NC 27708-0281 USA
- Department of Electrical and Computer Engineering, Duke University, Durham, USA
- Department of Neurobiology Engineering, Duke University, Durham, USA
- Department of Neurosurgery Engineering, Duke University, Durham, USA
| |
Collapse
|
22
|
Sykora M, Andelova K, Szeiffova Bacova B, Egan Benova T, Martiskova A, Knezl V, Tribulova N. Hypertension Induces Pro-arrhythmic Cardiac Connexome Disorders: Protective Effects of Treatment. Biomolecules 2023; 13:biom13020330. [PMID: 36830700 PMCID: PMC9953310 DOI: 10.3390/biom13020330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
Prolonged population aging and unhealthy lifestyles contribute to the progressive prevalence of arterial hypertension. This is accompanied by low-grade inflammation and over time results in heart dysfunction and failure. Hypertension-induced myocardial structural and ion channel remodeling facilitates the development of both atrial and ventricular fibrillation, and these increase the risk of stroke and sudden death. Herein, we elucidate hypertension-induced impairment of "connexome" cardiomyocyte junctions. This complex ensures cell-to-cell adhesion and coupling for electrical and molecular signal propagation. Connexome dysfunction can be a key factor in promoting the occurrence of both cardiac arrhythmias and heart failure. However, the available literature indicates that arterial hypertension treatment can hamper myocardial structural remodeling, hypertrophy and/or fibrosis, and preserve connexome function. This suggests the pleiotropic effects of antihypertensive agents, including anti-inflammatory. Therefore, further research is required to identify specific molecular targets and pathways that will protect connexomes, and it is also necessary to develop new approaches to maintain heart function in patients suffering from primary or pulmonary arterial hypertension.
Collapse
|
23
|
Mircea AA, Mori S, Ajijola OA, Khakpour H. Sinus Arrest during Radiofrequency Ablation from the Infero-septal Process of the Left Ventricle: Proposed Mechanisms of an Uncommon Finding. HeartRhythm Case Rep 2023. [DOI: 10.1016/j.hrcr.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
24
|
Filosa A, Sawamiphak S. Heart development and regeneration-a multi-organ effort. FEBS J 2023; 290:913-930. [PMID: 34894086 DOI: 10.1111/febs.16319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Development of the heart, from early morphogenesis to functional maturation, as well as maintenance of its homeostasis are tasks requiring collaborative efforts of cardiac tissue and different extra-cardiac organ systems. The brain, lymphoid organs, and gut are among the interaction partners that can communicate with the heart through a wide array of paracrine signals acting at local or systemic level. Disturbance of cardiac homeostasis following ischemic injury also needs immediate response from these distant organs. Our hearts replace dead muscles with non-contractile fibrotic scars. We have learned from animal models capable of scarless repair that regenerative capability of the heart does not depend only on competency of the myocardium and cardiac-intrinsic factors but also on long-range molecular signals originating in other parts of the body. Here, we provide an overview of inter-organ signals that take part in development and regeneration of the heart. We highlight recent findings and remaining questions. Finally, we discuss the potential of inter-organ modulatory approaches for possible therapeutic use.
Collapse
Affiliation(s)
- Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany
| |
Collapse
|
25
|
Magnuson JT, Leads RR, McGruer V, Qian L, Tanabe P, Roberts AP, Schlenk D. Transcriptomic profiling of miR-203a inhibitor and miR-34b-injected zebrafish (Danio rerio) validates oil-induced neurological, cardiovascular and eye toxicity response pathways. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 254:106356. [PMID: 36423467 DOI: 10.1016/j.aquatox.2022.106356] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 06/16/2023]
Abstract
The global sequencing of microRNA (miRNA; miR) and integration to downstream mRNA expression profiles in early life stages (ELS) of fish following exposure to crude oil determined consistently dysregulated miRNAs regardless of the oil source or fish species. The overlay of differentially expressed miRNAs and mRNAs into in silico software determined that the key roles of these miRNAs were predicted to be involved in cardiovascular, neurological and visually-mediated pathways. Of these, altered expression of miRNAs, miR-203a and miR-34b were predicted to be primary targets of crude oil. To better characterize the effect of these miRNAs to downstream transcript changes, zebrafish embryos were microinjected at 1 h post fertilization (hpf) with either a miR-203a inhibitor or miR-34b. Since both miRs have been shown to be associated with aryl hydrocarbon receptor (AhR) function, benzo(a)pyrene (BaP), a potent AhR agonist, was used as a potential positive control. Transcriptomic profiling was conducted on injected and exposed larvae at 7 and 72 hpf, and eye morphology assessed following exposure at 72 hpf. The top predicted physiological system disease and functions between differentially expressed genes (DEGs) shared with miR-203a inhibitor-injected and miR-34b-injected embryos were involved in brain formation, and the development of the central nervous system and neurons. When DEGs of miR-203a inhibitor-injected embryos were compared with BaP-exposed DEGs, alterations in nervous system development and function, and abnormal morphology of the neurosensory retina, eye and nervous tissue were predicted, consistent with both AhR and non-AhR pathways. When assessed morphologically, the eye area of miR-203a inhibitor and miR-34b-injected and BaP-exposed embryos were significantly reduced. These results suggest that miR-203a inhibition and miR-34b overexpression contribute to neurological, cardiovascular and eye toxicity responses that are caused by oil and PAH exposure in ELS fish, and are likely mediated through both AhR and non-AhR pathways.
Collapse
Affiliation(s)
- Jason T Magnuson
- University of California, Riverside, Department of Environmental Sciences, Riverside, CA, United States of America.
| | - Rachel R Leads
- University of North Texas, Department of Biological Sciences and Advanced Environmental, Research Institute, Denton, TX, United States of America
| | - Victoria McGruer
- University of California, Riverside, Department of Environmental Sciences, Riverside, CA, United States of America
| | - Le Qian
- University of California, Riverside, Department of Environmental Sciences, Riverside, CA, United States of America
| | - Philip Tanabe
- University of California, Riverside, Department of Environmental Sciences, Riverside, CA, United States of America
| | - Aaron P Roberts
- University of North Texas, Department of Biological Sciences and Advanced Environmental, Research Institute, Denton, TX, United States of America
| | - Daniel Schlenk
- University of California, Riverside, Department of Environmental Sciences, Riverside, CA, United States of America; Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang, University, Hangzhou, China
| |
Collapse
|
26
|
Smith JEG, Ashton JL, Argent LP, Cheyne JE, Montgomery JM. Recording plasticity in neuronal activity in the rodent intrinsic cardiac nervous system using calcium imaging techniques. Front Synaptic Neurosci 2023; 15:1104736. [PMID: 37082542 PMCID: PMC10110955 DOI: 10.3389/fnsyn.2023.1104736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/20/2023] [Indexed: 04/22/2023] Open
Abstract
The intrinsic cardiac nervous system (ICNS) is composed of interconnected clusters of neurons called ganglionated plexi (GP) which play a major role in controlling heart rate and rhythm. The function of these neurons is particularly important due to their involvement in cardiac arrhythmias such as atrial fibrillation (AF), and previous work has shown that plasticity in GP neural networks could underpin aberrant activity patterns that drive AF. As research in this field increases, developing new techniques to visualize the complex interactions and plasticity in this GP network is essential. In this study we have developed a calcium imaging method enabling the simultaneous recording of plasticity in neuronal activity from multiple neurons in intact atrial GP networks. Calcium imaging was performed with Cal-520 AM labeling in aged spontaneously hypertensive rats (SHRs), which display both spontaneous and induced AF, and age-matched Wistar Kyoto (WKY) controls to determine the relationship between chronic hypertension, arrhythmia and GP calcium dynamics. Our data show that SHR GPs have significantly larger calcium responses to cholinergic stimulation compared to WKY controls, as determined by both higher amplitude and longer duration calcium responses. Responses were significantly but not fully blocked by hexamethonium, indicating multiple cholinergic receptor subtypes are involved in the calcium response. Given that SHRs are susceptible to cardiac arrhythmias, our data provide evidence for a potential link between arrhythmia and plasticity in calcium dynamics that occur not only in cardiomyocytes but also in the GP neurons of the heart.
Collapse
Affiliation(s)
- Joscelin E. G. Smith
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Pūtahi Manawa, Centre for Heart Research, Auckland, New Zealand
| | - Jesse L. Ashton
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Pūtahi Manawa, Centre for Heart Research, Auckland, New Zealand
| | - Liam P. Argent
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Pūtahi Manawa, Centre for Heart Research, Auckland, New Zealand
| | | | - Johanna M. Montgomery
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Pūtahi Manawa, Centre for Heart Research, Auckland, New Zealand
- *Correspondence: Johanna M. Montgomery,
| |
Collapse
|
27
|
Bilkic I, Sotelo D, Anujarerat S, Ortiz NR, Alonzo M, El Khoury R, Loyola CC, Joddar B. Development of an extrusion-based 3D-printing strategy for clustering of human neural progenitor cells. Heliyon 2022; 8:e12250. [PMID: 36636220 PMCID: PMC9830177 DOI: 10.1016/j.heliyon.2022.e12250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/28/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
3D bioprinting offers a simplified solution for the engineering of complex tissue parts for in-vitro drug discovery or, in-vivo implantation. However, significant amount of challenges exist in 3D bioprinting of neural tissues, as these are sensitive cell types to handle via extrusion bioprinting techniques. We assessed the feasibility of bioprinting human neural progenitor cells (NPCs) in 3D hydrogel lattices using a fibrinogen-alginate-chitosan bioink, previously optimized for neural-cell growth, and subsequently modified for structural support during extrusion printing, in this study. The original bioink used in this study was made by adding optimized amounts of high- and medium-viscosity alginate to the fibrinogen-chitosan-based bioink and making it extrudable under shear pressure. The mechanically robust 3D constructs promoted NPC cluster formation and maintained their morphology and viability during the entire culture period. This strategy may be useful for co-culturing of NPCs along with other cell types such as cardiac, vascular, and other cells during 3D bioprinting.
Collapse
Affiliation(s)
- Ines Bilkic
- Department of Chemical Engineering and Materials Research Laboratory, University of California, Santa Barbara, CA 93106, USA
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Diana Sotelo
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Stephanie Anujarerat
- Department of Chemical Engineering and Materials Research Laboratory, University of California, Santa Barbara, CA 93106, USA
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Nickolas R. Ortiz
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Matthew Alonzo
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX, 79968, USA
| | - Raven El Khoury
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX, 79968, USA
| | - Carla C. Loyola
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX, 79968, USA
| | - Binata Joddar
- Inspired Materials and Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso, El Paso, TX, 79968, USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX, 79968, USA
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, 500 W. University Avenue, El Paso, TX, 79968, USA
| |
Collapse
|
28
|
Narkar A, Feaster TK, Casciola M, Blinova K. Human in vitro neurocardiac coculture (ivNCC) assay development for evaluating cardiac contractility modulation. Physiol Rep 2022; 10:e15498. [PMID: 36325586 PMCID: PMC9630755 DOI: 10.14814/phy2.15498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
Two of the most prominent organ systems, the nervous and the cardiovascular systems, are intricately connected to maintain homeostasis in mammals. Recent years have shown tremendous efforts toward therapeutic modulation of cardiac contractility and electrophysiology by electrical stimulation. Neuronal innervation and cardiac ganglia regulation are often overlooked when developing in vitro models for cardiac devices, but it is likely that peripheral nervous system plays a role in the clinical effects. We developed an in vitro neurocardiac coculture (ivNCC) model system to study cardiac and neuronal interplay using human induced pluripotent stem cell (hiPSC) technology. We demonstrated significant expression and colocalization of cardiac markers including troponin, α-actinin, and neuronal marker peripherin in neurocardiac coculture. To assess functional coupling between the cardiomyocytes and neurons, we evaluated nicotine-induced β-adrenergic norepinephrine effect and found beat rate was significantly increased in ivNCC as compared to monoculture alone. The developed platform was used as a nonclinical model for the assessment of cardiac medical devices that deliver nonexcitatory electrical pulses to the heart during the absolute refractory period of the cardiac cycle, that is, cardiac contractility modulation (CCM) therapy. Robust coculture response was observed at 14 V/cm (5 V, 64 mA), monophasic, 2 ms pulse duration for pacing and 20 V/cm (7 V, 90 mA) phase amplitude, biphasic, 5.14 ms pulse duration for CCM. We observed that the CCM effect and kinetics were more pronounced in coculture as compared to cardiac monoculture, supporting a hypothesis that some part of CCM mechanism of action can be attributed to peripheral nervous system stimulation. This study provides novel characterization of CCM effects on hiPSC-derived neurocardiac cocultures. This innervated human heart model can be further extended to investigate arrhythmic mechanisms, neurocardiac safety, and toxicity post-chronic exposure to materials, drugs, and medical devices. We present data on acute CCM electrical stimulation effects on a functional and optimized coculture using commercially available hiPSC-derived cardiomyocytes and neurons. Moreover, this study provides an in vitro human heart model to evaluate neuronal innervation and cardiac ganglia regulation of contractility by applying CCM pulse parameters that closely resemble clinical setting. This ivNCC platform provides a potential tool for investigating aspects of cardiac and neurological device safety and performance.
Collapse
Affiliation(s)
- Akshay Narkar
- Center for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Tromondae K. Feaster
- Center for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Maura Casciola
- Center for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Ksenia Blinova
- Center for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
29
|
Bufo MR, Guidotti M, De Faria C, Mofid Y, Bonnet-Brilhault F, Wardak C, Aguillon-Hernandez N. Autonomic tone in children and adults: Pupillary, electrodermal and cardiac activity at rest. Int J Psychophysiol 2022; 180:68-78. [PMID: 35914548 DOI: 10.1016/j.ijpsycho.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/24/2022]
Abstract
Considering the suspected involvement of the autonomic nervous system (ANS) in several neurodevelopmental disorders, a description of its tonus in typical populations and of its maturation between childhood and adulthood is necessary. We aimed to arrive at a better understanding of the maturation of the sympathetic (SNS) and parasympathetic (PNS) tonus by comparing children and adults at rest, via recordings of multiple ANS indices. We recorded simultaneously pupil diameter, electrodermal activity (EDA) and cardiac activity (RR interval and HRV: heart rate variability) in 29 children (6-12 years old) and 30 adults (20-42 years old) during a 5-min rest period. Children exhibited lower RR intervals, higher LF peak frequencies, and lower LF/HF (low frequency/high frequency) ratios compared to adults. Children also produced more spontaneous EDA peaks, reflected in a larger EDA AUC (area under the curve), in comparison with adults. Finally, children displayed a larger median pupil diameter and a higher pupillary hippus frequency than adults. Our results converged towards higher SNS and PNS tones in children compared to adults. Childhood would thus be characterized by a high autonomic tone, possibly reflecting a physiological state compatible with developmental acquisitions.
Collapse
Affiliation(s)
- Maria Rosa Bufo
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Marco Guidotti
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France; Centre universitaire de pédopsychiatrie, CHRU de Tours, Tours, France; Centre Hospitalier du Chinonais, Saint-Benoît-la-Forêt, France
| | - Cindie De Faria
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Yassine Mofid
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France; Centre universitaire de pédopsychiatrie, CHRU de Tours, Tours, France
| | - Claire Wardak
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | |
Collapse
|
30
|
Yang D, Dai X, Xing Y, Tang X, Yang G, Harrison AG, Cahoon J, Li H, Lv X, Yu X, Wang P, Wang H. Intrinsic cardiac adrenergic cells contribute to LPS-induced myocardial dysfunction. Commun Biol 2022; 5:96. [PMID: 35079095 PMCID: PMC8789803 DOI: 10.1038/s42003-022-03007-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023] Open
Abstract
Intrinsic cardiac adrenergic (ICA) cells regulate both developing and adult cardiac physiological and pathological processes. However, the role of ICA cells in septic cardiomyopathy is unknown. Here we show that norepinephrine (NE) secretion from ICA cells is increased through activation of Toll-like receptor 4 (TLR4) to aggravate myocardial TNF-α production and dysfunction by lipopolysaccharide (LPS). In ICA cells, LPS activated TLR4-MyD88/TRIF-AP-1 signaling that promoted NE biosynthesis through expression of tyrosine hydroxylase, but did not trigger TNF-α production due to impairment of p65 translocation. In a co-culture consisting of LPS-treated ICA cells and cardiomyocytes, the upregulation and secretion of NE from ICA cells activated cardiomyocyte β1-adrenergic receptor driving Ca2+/calmodulin-dependent protein kinase II (CaMKII) to crosstalk with NF-κB and mitogen-activated protein kinase pathways. Importantly, blockade of ICA cell-derived NE prevented LPS-induced myocardial dysfunction. Our findings suggest that ICA cells may be a potential therapeutic target for septic cardiomyopathy.
Collapse
Affiliation(s)
- Duomeng Yang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiaomeng Dai
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yun Xing
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiangxu Tang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Guang Yang
- Department of Pathogen biology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Andrew G Harrison
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Jason Cahoon
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Hongmei Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiuxiu Lv
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiaohui Yu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Penghua Wang
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
31
|
Gruscheski L, Brand T. The Role of POPDC Proteins in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis 2021; 8:160. [PMID: 34940515 PMCID: PMC8706714 DOI: 10.3390/jcdd8120160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
The Popeye domain-containing (POPDC) gene family, consisting of Popdc1 (also known as Bves), Popdc2, and Popdc3, encodes transmembrane proteins abundantly expressed in striated muscle. POPDC proteins have recently been identified as cAMP effector proteins and have been proposed to be part of the protein network involved in cAMP signaling. However, their exact biochemical activity is presently poorly understood. Loss-of-function mutations in animal models causes abnormalities in skeletal muscle regeneration, conduction, and heart rate adaptation after stress. Likewise, patients carrying missense or nonsense mutations in POPDC genes have been associated with cardiac arrhythmias and limb-girdle muscular dystrophy. In this review, we introduce the POPDC protein family, and describe their structure function, and role in cAMP signaling. Furthermore, the pathological phenotypes observed in zebrafish and mouse models and the clinical and molecular pathologies in patients carrying POPDC mutations are described.
Collapse
Affiliation(s)
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
32
|
Stoyek MR, Hortells L, Quinn TA. From Mice to Mainframes: Experimental Models for Investigation of the Intracardiac Nervous System. J Cardiovasc Dev Dis 2021; 8:149. [PMID: 34821702 PMCID: PMC8620975 DOI: 10.3390/jcdd8110149] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/17/2023] Open
Abstract
The intracardiac nervous system (IcNS), sometimes referred to as the "little brain" of the heart, is involved in modulating many aspects of cardiac physiology. In recent years our fundamental understanding of autonomic control of the heart has drastically improved, and the IcNS is increasingly being viewed as a therapeutic target in cardiovascular disease. However, investigations of the physiology and specific roles of intracardiac neurons within the neural circuitry mediating cardiac control has been hampered by an incomplete knowledge of the anatomical organisation of the IcNS. A more thorough understanding of the IcNS is hoped to promote the development of new, highly targeted therapies to modulate IcNS activity in cardiovascular disease. In this paper, we first provide an overview of IcNS anatomy and function derived from experiments in mammals. We then provide descriptions of alternate experimental models for investigation of the IcNS, focusing on a non-mammalian model (zebrafish), neuron-cardiomyocyte co-cultures, and computational models to demonstrate how the similarity of the relevant processes in each model can help to further our understanding of the IcNS in health and disease.
Collapse
Affiliation(s)
- Matthew R. Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
| | - Luis Hortells
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg–Bad Krozingen, 79110 Freiburg, Germany;
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
- School of Biomedical Engineering, Dalhousie University, Halifax, NS 15000, Canada
| |
Collapse
|
33
|
Cerritelli F, Frasch MG, Antonelli MC, Viglione C, Vecchi S, Chiera M, Manzotti A. A Review on the Vagus Nerve and Autonomic Nervous System During Fetal Development: Searching for Critical Windows. Front Neurosci 2021; 15:721605. [PMID: 34616274 PMCID: PMC8488382 DOI: 10.3389/fnins.2021.721605] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
The autonomic nervous system (ANS) is one of the main biological systems that regulates the body's physiology. Autonomic nervous system regulatory capacity begins before birth as the sympathetic and parasympathetic activity contributes significantly to the fetus' development. In particular, several studies have shown how vagus nerve is involved in many vital processes during fetal, perinatal, and postnatal life: from the regulation of inflammation through the anti-inflammatory cholinergic pathway, which may affect the functioning of each organ, to the production of hormones involved in bioenergetic metabolism. In addition, the vagus nerve has been recognized as the primary afferent pathway capable of transmitting information to the brain from every organ of the body. Therefore, this hypothesis paper aims to review the development of ANS during fetal and perinatal life, focusing particularly on the vagus nerve, to identify possible "critical windows" that could impact its maturation. These "critical windows" could help clinicians know when to monitor fetuses to effectively assess the developmental status of both ANS and specifically the vagus nerve. In addition, this paper will focus on which factors-i.e., fetal characteristics and behaviors, maternal lifestyle and pathologies, placental health and dysfunction, labor, incubator conditions, and drug exposure-may have an impact on the development of the vagus during the above-mentioned "critical window" and how. This analysis could help clinicians and stakeholders define precise guidelines for improving the management of fetuses and newborns, particularly to reduce the potential adverse environmental impacts on ANS development that may lead to persistent long-term consequences. Since the development of ANS and the vagus influence have been shown to be reflected in cardiac variability, this paper will rely in particular on studies using fetal heart rate variability (fHRV) to monitor the continued growth and health of both animal and human fetuses. In fact, fHRV is a non-invasive marker whose changes have been associated with ANS development, vagal modulation, systemic and neurological inflammatory reactions, and even fetal distress during labor.
Collapse
Affiliation(s)
- Francesco Cerritelli
- Research and Assistance for Infants to Support Experience Lab, Foundation Center for Osteopathic Medicine Collaboration, Pescara, Italy
| | - Martin G. Frasch
- Department of Obstetrics and Gynecology and Center on Human Development and Disability, University of Washington, Seattle, WA, United States
| | - Marta C. Antonelli
- Facultad de Medicina, Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”, Universidad de Buenos Aires, Buenos Aires, Argentina
- Department of Obstetrics and Gynecology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Chiara Viglione
- Research and Assistance for Infants to Support Experience Lab, Foundation Center for Osteopathic Medicine Collaboration, Pescara, Italy
| | - Stefano Vecchi
- Research and Assistance for Infants to Support Experience Lab, Foundation Center for Osteopathic Medicine Collaboration, Pescara, Italy
| | - Marco Chiera
- Research and Assistance for Infants to Support Experience Lab, Foundation Center for Osteopathic Medicine Collaboration, Pescara, Italy
| | - Andrea Manzotti
- Research and Assistance for Infants to Support Experience Lab, Foundation Center for Osteopathic Medicine Collaboration, Pescara, Italy
- Department of Pediatrics, Division of Neonatology, “V. Buzzi” Children's Hospital, Azienda Socio-Sanitaria Territoriale Fatebenefratelli Sacco, Milan, Italy
- Research Department, Istituto Osteopatia Milano, Milan, Italy
| |
Collapse
|
34
|
Harper AA, Adams DJ. Electrical properties and synaptic transmission in mouse intracardiac ganglion neurons in situ. Physiol Rep 2021; 9:e15056. [PMID: 34582125 PMCID: PMC8477906 DOI: 10.14814/phy2.15056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/25/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
The intrinsic cardiac nervous system represents the final site of signal integration for neurotransmission to the myocardium to enable local control of cardiac performance. The electrophysiological characteristics and ganglionic transmission of adult mouse intrinsic cardiac ganglion (ICG) neurons were investigated using a whole-mount ganglion preparation of the excised right atrial ganglion plexus and intracellular microelectrode recording techniques. The passive and active electrical properties of ICG neurons and synaptic transmission including synaptic response strength and efficacy as a function of stimulation frequency were examined. The resting membrane potential and input resistance of ICG neurons were -47.9 ± 4.0 mV and 197.2 ± 81.5 MΩ, respectively. All neurons had somatic action potentials with overshoots of >+15 mV and after-hyperpolarizations having an average of 10 mV amplitude and ~45 ms half duration. Phasic discharge activities were recorded from the majority of neurons studied and several types of excitatory synaptic responses were recorded following inputs from the vagus or interganglionic nerve trunk(s). Most postganglionic neurons (>75%) received a strong, suprathreshold synaptic input and reliably followed high-frequency repetitive nerve stimulation up to at least 50 Hz. Nerve-evoked synaptic transmission was blocked by extracellular Cd2+ , ω-conotoxin CVIE, or α-conotoxin RegIIA, a selective α3-containing nicotinic acetylcholine receptor antagonist. Synaptic transmission and the electrical properties of murine ICG neurons contribute to the pattern of discharge which regulates chronotropic, dromotropic, and inotropic elements of cardiac function.
Collapse
Affiliation(s)
- Alexander A. Harper
- Illawarra Health and Medical Research Institute (IHMRI)University of WollongongWollongongNew South WalesAustralia
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI)University of WollongongWollongongNew South WalesAustralia
| |
Collapse
|
35
|
Scalco A, Moro N, Mongillo M, Zaglia T. Neurohumoral Cardiac Regulation: Optogenetics Gets Into the Groove. Front Physiol 2021; 12:726895. [PMID: 34531763 PMCID: PMC8438220 DOI: 10.3389/fphys.2021.726895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022] Open
Abstract
The cardiac autonomic nervous system (ANS) is the main modulator of heart function, adapting contraction force, and rate to the continuous variations of intrinsic and extrinsic environmental conditions. While the parasympathetic branch dominates during rest-and-digest sympathetic neuron (SN) activation ensures the rapid, efficient, and repeatable increase of heart performance, e.g., during the "fight-or-flight response." Although the key role of the nervous system in cardiac homeostasis was evident to the eyes of physiologists and cardiologists, the degree of cardiac innervation, and the complexity of its circuits has remained underestimated for too long. In addition, the mechanisms allowing elevated efficiency and precision of neurogenic control of heart function have somehow lingered in the dark. This can be ascribed to the absence of methods adequate to study complex cardiac electric circuits in the unceasingly moving heart. An increasing number of studies adds to the scenario the evidence of an intracardiac neuron system, which, together with the autonomic components, define a little brain inside the heart, in fervent dialogue with the central nervous system (CNS). The advent of optogenetics, allowing control the activity of excitable cells with cell specificity, spatial selectivity, and temporal resolution, has allowed to shed light on basic neuro-cardiology. This review describes how optogenetics, which has extensively been used to interrogate the circuits of the CNS, has been applied to untangle the knots of heart innervation, unveiling the cellular mechanisms of neurogenic control of heart function, in physiology and pathology, as well as those participating to brain-heart communication, back and forth. We discuss existing literature, providing a comprehensive view of the advancement in the understanding of the mechanisms of neurogenic heart control. In addition, we weigh the limits and potential of optogenetics in basic and applied research in neuro-cardiology.
Collapse
Affiliation(s)
- Arianna Scalco
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Nicola Moro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Tania Zaglia
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
36
|
Choi S, Baudot M, Vivas O, Moreno CM. Slowing down as we age: aging of the cardiac pacemaker's neural control. GeroScience 2021; 44:1-17. [PMID: 34292477 PMCID: PMC8811107 DOI: 10.1007/s11357-021-00420-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022] Open
Abstract
The cardiac pacemaker ignites and coordinates the contraction of the whole heart, uninterruptedly, throughout our entire life. Pacemaker rate is constantly tuned by the autonomous nervous system to maintain body homeostasis. Sympathetic and parasympathetic terminals act over the pacemaker cells as the accelerator and the brake pedals, increasing or reducing the firing rate of pacemaker cells to match physiological demands. Despite the remarkable reliability of this tissue, the pacemaker is not exempt from the detrimental effects of aging. Mammals experience a natural and continuous decrease in the pacemaker rate throughout the entire lifespan. Why the pacemaker rhythm slows with age is poorly understood. Neural control of the pacemaker is remodeled from birth to adulthood, with strong evidence of age-related dysfunction that leads to a downshift of the pacemaker. Such evidence includes remodeling of pacemaker tissue architecture, alterations in the innervation, changes in the sympathetic acceleration and the parasympathetic deceleration, and alterations in the responsiveness of pacemaker cells to adrenergic and cholinergic modulation. In this review, we revisit the main evidence on the neural control of the pacemaker at the tissue and cellular level and the effects of aging on shaping this neural control.
Collapse
Affiliation(s)
- Sabrina Choi
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Matthias Baudot
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Oscar Vivas
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Claudia M Moreno
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
37
|
Odnoshivkina YG, Petrov AM. The Role of Neuro-Cardiac Junctions
in Sympathetic Regulation of the Heart. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Wallace MJ, El Refaey M, Mesirca P, Hund TJ, Mangoni ME, Mohler PJ. Genetic Complexity of Sinoatrial Node Dysfunction. Front Genet 2021; 12:654925. [PMID: 33868385 PMCID: PMC8047474 DOI: 10.3389/fgene.2021.654925] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The pacemaker cells of the cardiac sinoatrial node (SAN) are essential for normal cardiac automaticity. Dysfunction in cardiac pacemaking results in human sinoatrial node dysfunction (SND). SND more generally occurs in the elderly population and is associated with impaired pacemaker function causing abnormal heart rhythm. Individuals with SND have a variety of symptoms including sinus bradycardia, sinus arrest, SAN block, bradycardia/tachycardia syndrome, and syncope. Importantly, individuals with SND report chronotropic incompetence in response to stress and/or exercise. SND may be genetic or secondary to systemic or cardiovascular conditions. Current management of patients with SND is limited to the relief of arrhythmia symptoms and pacemaker implantation if indicated. Lack of effective therapeutic measures that target the underlying causes of SND renders management of these patients challenging due to its progressive nature and has highlighted a critical need to improve our understanding of its underlying mechanistic basis of SND. This review focuses on current information on the genetics underlying SND, followed by future implications of this knowledge in the management of individuals with SND.
Collapse
Affiliation(s)
- Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Pietro Mesirca
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matteo E. Mangoni
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
39
|
Bordoni B, Escher AR. Osteopathic Palpation of the Heart. Cureus 2021; 13:e14187. [PMID: 33816036 PMCID: PMC8008978 DOI: 10.7759/cureus.14187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/17/2022] Open
Abstract
In the panorama of scientific literature, there is a paucity of literature on how to palpate the heart area in the osteopathic setting and relevant indications on which palpatory sensations the clinician should perceive during the evaluation. The article reviews the fascial anatomy of the heart area and the heart movements derived from magnetic resonance imaging (MRI) studies and describes the landmarks used by the cardiac surgeon to visualize the mediastinal area. The text sets out possible suggestions for a more adequate osteopathic palpatory evaluation and describes any tactile sensations arising from the patient's chest. To the knowledge of the authors, this is the first article that seeks to lay solid foundations for the improvement of osteopathic manual medicine in the cardiology field.
Collapse
Affiliation(s)
- Bruno Bordoni
- Physical Medicine and Rehabilitation, Foundation Don Carlo Gnocchi, Milan, ITA
| | - Allan R Escher
- Anesthesiology/Pain Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| |
Collapse
|
40
|
Tissue Chips and Microphysiological Systems for Disease Modeling and Drug Testing. MICROMACHINES 2021; 12:mi12020139. [PMID: 33525451 PMCID: PMC7911320 DOI: 10.3390/mi12020139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Tissue chips (TCs) and microphysiological systems (MPSs) that incorporate human cells are novel platforms to model disease and screen drugs and provide an alternative to traditional animal studies. This review highlights the basic definitions of TCs and MPSs, examines four major organs/tissues, identifies critical parameters for organization and function (tissue organization, blood flow, and physical stresses), reviews current microfluidic approaches to recreate tissues, and discusses current shortcomings and future directions for the development and application of these technologies. The organs emphasized are those involved in the metabolism or excretion of drugs (hepatic and renal systems) and organs sensitive to drug toxicity (cardiovascular system). This article examines the microfluidic/microfabrication approaches for each organ individually and identifies specific examples of TCs. This review will provide an excellent starting point for understanding, designing, and constructing novel TCs for possible integration within MPS.
Collapse
|