skip to main content


Title: Optical vagus nerve modulation of heart and respiration via heart-injected retrograde AAV
Abstract Vagus nerve stimulation has shown many benefits for disease therapies but current approaches involve imprecise electrical stimulation that gives rise to off-target effects, while the functionally relevant pathways remain poorly understood. One method to overcome these limitations is the use of optogenetic techniques, which facilitate targeted neural communication with light-sensitive actuators (opsins) and can be targeted to organs of interest based on the location of viral delivery. Here, we tested whether retrograde adeno-associated virus (rAAV2-retro) injected in the heart can be used to selectively express opsins in vagus nerve fibers controlling cardiac function. Furthermore, we investigated whether perturbations in cardiac function could be achieved with photostimulation at the cervical vagus nerve. Viral injection in the heart resulted in robust, primarily afferent, opsin reporter expression in the vagus nerve, nodose ganglion, and brainstem. Photostimulation using both one-photon stimulation and two-photon holography with a GRIN-lens incorporated nerve cuff, was tested on the pilot-cohort of injected mice. Changes in heart rate, surface electrocardiogram, and respiratory responses were observed in response to both one- and two-photon photostimulation. The results demonstrate feasibility of retrograde labeling for organ targeted optical neuromodulation.  more » « less
Award ID(s):
1631912
NSF-PAR ID:
10304183
Author(s) / Creator(s):
; ; ; ; ; ; ; ; ; ;
Date Published:
Journal Name:
Scientific Reports
Volume:
11
Issue:
1
ISSN:
2045-2322
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. New Findings

    What is the topic of this review?

    The vagus nerve is a crucial regulator of cardiovascular homeostasis, and its activity is linked to heart health. Vagal activity originates from two brainstem nuclei: the nucleus ambiguus (fast lane) and the dorsal motor nucleus of the vagus (slow lane), nicknamed for the time scales that they require to transmit signals.

    What advances does it highlight?

    Computational models are powerful tools for organizing multi‐scale, multimodal data on the fast and slow lanes in a physiologically meaningful way. A strategy is laid out for how these models can guide experiments aimed at harnessing the cardiovascular health benefits of differential activation of the fast and slow lanes.

    Abstract

    The vagus nerve is a key mediator of brain–heart signaling, and its activity is necessary for cardiovascular health. Vagal outflow stems from the nucleus ambiguus, responsible primarily for fast, beat‐to‐beat regulation of heart rate and rhythm, and the dorsal motor nucleus of the vagus, responsible primarily for slow regulation of ventricular contractility. Due to the high‐dimensional and multimodal nature of the anatomical, molecular and physiological data on neural regulation of cardiac function, data‐derived mechanistic insights have proven elusive. Elucidating insights has been complicated further by the broad distribution of the data across heart, brain and peripheral nervous system circuits. Here we lay out an integrative framework based on computational modelling for combining these disparate and multi‐scale data on the two vagal control lanes of the cardiovascular system. Newly available molecular‐scale data, particularly single‐cell transcriptomic analyses, have augmented our understanding of the heterogeneous neuronal states underlying vagally mediated fast and slow regulation of cardiac physiology. Cellular‐scale computational models built from these data sets represent building blocks that can be combined using anatomical and neural circuit connectivity, neuronal electrophysiology, and organ/organismal‐scale physiology data to create multi‐system, multi‐scale models that enablein silicoexploration of the fast versus slow lane vagal stimulation. The insights from the computational modelling and analyses will guide new experimental questions on the mechanisms regulating the fast and slow lanes of the cardiac vagus toward exploiting targeted vagal neuromodulatory activity to promote cardiovascular health.

     
    more » « less
  2. null (Ed.)
    We have discovered a cardiac-inducing RNA (CIR) in the axolotl, Ambystoma mexicanum, (a salamander) and two cardiac inducing RNAs (CIR-6 and CIR-30) in human heart that have the ability to induce the differentiation of non-muscle cells, including induced pluripotent stem cells from human skin, mouse embryonic stem cells, and mouse fibroblasts into cardiomyocytes in vitro. Although the primary sequences of salamander and human RNAs are not homologous, their secondary structures are very similar and we believe account for their shared unique abilities to promote differentiation of non-muscle cells into definitive cardiomyocytes. We are beginning to explore the potential for repair/regeneration of cardiac muscle in vivo using mouse and rat models with induced acute myocardial infarctions (AMI) to determine if pluripotent stem cells or fibroblasts transfected with the human CIRs or CIRs alone injected into the damaged areas of the hearts can effect repair of the damaged cardiac muscle tissue, and return the infarcted hearts and the AMI animal models to pre-heart-attack function again. If cardiac cells damaged in heart attacks can be replaced with living, functioning cardiomyocytes, patients with heart disease would be able to have normal heart function restored and could return to normal pre-heart-attack activity levels. Understanding how CIR transforms non-muscle cells into vigorously contracting, functional cardiac muscle and effectively replacing damaged heart cells with newly-formed cardiac muscle tissue would represent a major breakthrough in modern biology and medicine with the potential to have a significant impact on the survival rate and quality of life of millions of individuals worldwide who suffer heart attacks each year. 
    more » « less
  3. Rationale: NAA15 (N-alpha-acetyltransferase 15) is a component of the NatA (N-terminal acetyltransferase complex). The mechanism by which NAA15 haploinsufficiency causes congenital heart disease remains unknown. To better understand molecular processes by which NAA15 haploinsufficiency perturbs cardiac development, we introduced NAA15 variants into human induced pluripotent stem cells (iPSCs) and assessed the consequences of these mutations on RNA and protein expression. Objective: We aim to understand the role of NAA15 haploinsufficiency in cardiac development by investigating proteomic effects on NatA complex activity and identifying proteins dependent upon a full amount of NAA15. Methods and Results: We introduced heterozygous loss of function, compound heterozygous, and missense residues (R276W) in iPSCs using CRISPR/Cas9. Haploinsufficient NAA15 iPSCs differentiate into cardiomyocytes, unlike NAA15 -null iPSCs, presumably due to altered composition of NatA. Mass spectrometry analyses reveal ≈80% of identified iPSC NatA targeted proteins displayed partial or complete N-terminal acetylation. Between null and haploinsufficient NAA15 cells, N-terminal acetylation levels of 32 and 9 NatA-specific targeted proteins were reduced, respectively. Similar acetylation loss in few proteins occurred in NAA15 R276W induced pluripotent stem cells. In addition, steady-state protein levels of 562 proteins were altered in both null and haploinsufficient NAA15 cells; 18 were ribosomal-associated proteins. At least 4 proteins were encoded by genes known to cause autosomal dominant congenital heart disease. Conclusions: These studies define a set of human proteins that requires a full NAA15 complement for normal synthesis and development. A 50% reduction in the amount of NAA15 alters levels of at least 562 proteins and N-terminal acetylation of only 9 proteins. One or more modulated proteins are likely responsible for NAA15-haploinsufficiency mediated congenital heart disease. Additionally, genetically engineered induced pluripotent stem cells provide a platform for evaluating the consequences of amino acid sequence variants of unknown significance on NAA15 function. 
    more » « less
  4. Abstract

    To treat diseases associated with vagal nerve control of peripheral organs, it is necessary to selectively activate efferent and afferent fibers in the vagus. As a result of the nerve’s complex anatomy, fiber-specific activation proves challenging. Spatially selective neuromodulation using micromagnetic stimulation(μMS) is showing incredible promise. This neuromodulation technique uses microcoils(μcoils) to generate magnetic fields by powering them with a time-varying current. Following the principles of Faraday’s law of induction, a highly directional electric field is induced in the nerve from the magnetic field. In this study on rodent cervical vagus, a solenoidalμcoil was oriented at an angle to left and right branches of the nerve. The aim of this study was to measure changes in the mean arterial pressure (MAP) and heart rate (HR) followingμMS of the vagus. Theμcoils were powered by a single-cycle sinusoidal current varying in pulse widths(PW = 100, 500, and 1000μsec) at a frequency of 20 Hz. Under the influence of isoflurane,μMS of the left vagus at 1000μsec PW led to an average drop in MAP of 16.75 mmHg(n = 7). In contrast,μMS of the right vagus under isoflurane resulted in an average drop of 11.93 mmHg in the MAP(n = 7). Surprisingly, there were no changes in HR to either right or left vagalμMS suggesting the drop in MAP associated with vagusμMS was the result of stimulation of afferent, but not efferent fibers. In urethane anesthetized rats, no changes in either MAP or HR were observed uponμMS of the right or left vagus(n = 3). These findings suggest the choice of anesthesia plays a key role in determining the efficacy ofμMS on the vagal nerve. Absence of HR modulation uponμMS could offer alternative treatment options using VNS with fewer heart-related side-effects.

     
    more » « less
  5. Abstract

    Ischemic heart injury causes permanent cardiomyocyte loss and fibrosis impairing cardiac function. Tissue derived biomaterials have shown promise as an injectable treatment for the post‐ischemic heart. Specifically, decellularized extracellular matrix (dECM) is a protein rich suspension that forms a therapeutic hydrogel once injected and improves the heart post‐injury response in rodents and pig models. Current dECM‐derived biomaterials are delivered to the heart as a liquid dECM hydrogel precursor or colloidal suspension, which gels over several minutes. To increase the functionality of the dECM therapy, an injectable solid dECM microparticle formulation derived from heart tissue to control sizing and extend stability in aqueous conditions is developed. When delivered into the infarcted mouse heart, these dECM microparticles protect cardiac function promote vessel density and reduce left ventricular remodeling by sustained delivery of biomolecules. Longer retention, higher stiffness, and slower protein release of dECM microparticles are noted compared to liquid dECM hydrogel precursor. In addition, the dECM microparticle can be developed as a platform for macromolecule delivery. Together, the results suggest that dECM microparticles can be developed as a modular therapy for heart injury.

     
    more » « less