1
|
Wei SC, Cantor AJ, Walleshauser J, Mepani R, Melton K, Bans A, Khekare P, Gupta S, Wang J, Soares C, Kiwan R, Lee J, McCawley S, Jani V, Leong WI, Shahi PK, Chan J, Boivin P, Otoupal P, Pattnaik BR, Gamm DM, Saha K, Gowen BG, Haak-Frendscho M, Janatpour MJ, Silverman AP. Evaluation of subretinally delivered Cas9 ribonucleoproteins in murine and porcine animal models highlights key considerations for therapeutic translation of genetic medicines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630799. [PMID: 39803585 PMCID: PMC11722268 DOI: 10.1101/2024.12.30.630799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Genetic medicines, including CRISPR/Cas technologies, extend tremendous promise for addressing unmet medical need in inherited retinal disorders and other indications; however, there remain challenges for the development of therapeutics. Herein, we evaluate genome editing by engineered Cas9 ribonucleoproteins (eRNP) in vivo via subretinal administration using mouse and pig animal models. Subretinal administration of adenine base editor and double strand break-inducing Cas9 nuclease eRNPs mediate genome editing in both species. Editing occurs in retinal pigmented epithelium (RPE) and photoreceptor cells, with favorable tolerability in both species. Using transgenic reporter strains, we determine that editing primarily occurs close to the site of administration, within the bleb region associated with subretinal injection. Our results show that subretinal administration of eRNPs in mice mediates base editing of up to 12% of the total neural retina, with an average rate of 7% observed at the highest dose tested. In contrast, a substantially lower editing efficiency was observed in minipigs; even with direct quantification of only the treated region, a maximum base editing rate of 1.5%, with an average rate of <1%, was observed. Our data highlight the importance of species consideration in translational studies for genetic medicines targeting the eye and provide an example of a lack of translation between small and larger animal models in the context of subretinal administration of Cas9 eRNPs.
Collapse
Affiliation(s)
| | | | | | | | | | - Ashil Bans
- Spotlight Therapeutics, Hayward, CA, USA
| | | | | | | | | | | | - Jieun Lee
- Spotlight Therapeutics, Hayward, CA, USA
| | | | | | | | - Pawan K. Shahi
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jean Chan
- Spotlight Therapeutics, Hayward, CA, USA
| | | | | | - Bikash R. Pattnaik
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - David M. Gamm
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Krishanu Saha
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute of Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | |
Collapse
|
2
|
Hardy M, Chen Y, Baram TZ, Justice NJ. Targeting corticotropin-releasing hormone receptor type 1 (Crhr1) neurons: validating the specificity of a novel transgenic Crhr1-FlpO mouse. Brain Struct Funct 2024; 230:12. [PMID: 39692887 PMCID: PMC11655595 DOI: 10.1007/s00429-024-02879-0] [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: 08/23/2024] [Accepted: 10/05/2024] [Indexed: 12/19/2024]
Abstract
Corticotropin-releasing hormone (CRH) signaling through its cognate receptors, CRHR1 and CRHR2, contributes to diverse stress-related functions in the mammalian brain. Whereas CRHR2 is predominantly expressed in choroid plexus and blood vessels, CRHR1 is abundantly expressed in neurons in discrete brain regions, including the neocortex, hippocampus and nucleus accumbens. Activation of CRHR1 influences motivated behaviors, emotional states, and learning and memory. However, it is unknown whether alterations in CRHR1 signaling contribute to aberrant motivated behaviors observed, for example, in stressful contexts. These questions require tools to manipulate CRHR1 selectively. Here we describe and validate a novel Crhr1-FlpO mouse. Using bacterial artificial chromosome (BAC) transgenesis, we engineered a transgenic mouse that expresses FlpO recombinase in CRHR1-expressing cells. We used two independent methods to assess the specificity of FlpO to CRHR1-expressing cells. First, we injected Crhr1-FlpO mice with Flp-dependent viruses expressing fluorescent reporter molecules. Additionally, we crossed the Crhr1-FlpO mouse with a transgenic Flp-dependent reporter mouse. CRHR1 and reporter molecules were identified using immunocytochemistry and visualized via confocal microscopy in several brain regions in which CRHR1 expression and function is established. Expression of Flp-dependent viral constructs was highly specific to CRHR1-expressing cells in all regions examined (over 90% co-localization). In accord, robust and specific expression of the Flp-dependent transgenic reporter was observed in a reporter mouse, recapitulating endogenous CRHR1 expression. The Crhr1-FlpO mouse enables selective genetic access to CRHR1-expressing cells within the mouse brain. When combined with Cre-lox or site-specific recombinases, the mouse facilitates intersectional manipulations of CRHR1-expressing neurons.
Collapse
Affiliation(s)
- Mason Hardy
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Yuncai Chen
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics and Neurology, University of California-Irvine, Irvine, CA, USA.
| | - Nicholas J Justice
- Brown Foundation Institute of Molecular Medicine of McGovern Medical School, University of Texas Health Sciences Center-Houston, Houston, TX, USA
| |
Collapse
|
3
|
Lusk S, Memos NK, Rauschmayer A, Ray RS. The microbiome is dispensable for normal respiratory function and chemoreflexes in mice. Front Physiol 2024; 15:1481394. [PMID: 39712189 PMCID: PMC11659286 DOI: 10.3389/fphys.2024.1481394] [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: 08/15/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
Increasing evidence indicates an association between microbiome composition and respiratory homeostasis and disease, particularly disordered breathing, such as obstructive sleep apnea. Previous work showing respiratory disruption is limited by the methodology employed to disrupt, eliminate, or remove the microbiome by antibiotic depletion. Our work utilized germ-free mice born without a microbiome and described respiratory alterations. We used whole-body flow through barometric plethysmography to assay conscious and unrestrained C57BL/6J germ-free (GF, n = 24) and specific-pathogen-free (SPF, n = 28) adult mice (with an intact microbiome) in normoxic (21% O2,79% N2) conditions and during challenges in hypercapnic (5% CO2, 21% O2, 74% N2) and hypoxic (10% O2, 90% N2) environments. Following initial plethysmography analysis, we performed fecal transplants to test the ability of gut microbiome establishment to rescue any observed phenotypes. Data were comprehensively analyzed using our newly published respiratory analysis software, Breathe Easy, to identify alterations in respiratory parameters, including ventilatory frequency, tidal volume, ventilation, apnea frequency, and sigh frequency. We also considered possible metabolic changes by analyzing oxygen consumption, carbon dioxide production, and ventilatory equivalents of oxygen. We also assayed GF and SPF neonates in an autoresuscitation assay to understand the effects of the microbiome on cardiorespiratory stressors in early development. We found several differences in baseline and recovery cardiorespiratory parameters in the neonates and differences in body weight at both ages studied. However, there was no difference in the overall survival of the neonates, and in contrast to prior studies utilizing gut microbial depletion, we found no consequential respiratory alterations in GF versus SPF adult mice at baseline or following fecal transplant in any groups. Interestingly, we did see alterations in oxygen consumption in the GF adult mice, which suggests an altered metabolic demand. Results from this study suggest that microbiome alteration in mice may not play as large a role in respiratory outcomes when a less severe methodology to eliminate the microbiome is utilized.
Collapse
Affiliation(s)
- Savannah Lusk
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Nicoletta K. Memos
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Andrea Rauschmayer
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Russell S. Ray
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Baylor College of Medicine, McNair Medical Institute, Houston, TX, United States
| |
Collapse
|
4
|
Regan SB, Medhi D, White TB, Jiang YZ, Jia S, Deng Q, Jasin M. Megabase-scale loss of heterozygosity provoked by CRISPR-Cas9 DNA double-strand breaks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615517. [PMID: 39386534 PMCID: PMC11463394 DOI: 10.1101/2024.09.27.615517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Harnessing DNA double-strand breaks (DSBs) is a powerful approach for gene editing, but it may provoke loss of heterozygosity (LOH), which predisposes to tumorigenesis. To interrogate this risk, we developed a two- color flow cytometry-based system (Flo-LOH), detecting LOH in ∼5% of cells following a DSB. After this initial increase, cells with LOH decrease due to a competitive disadvantage with parental cells, but if isolated, they stably propagate. Segmental loss from terminal deletions with de novo telomere addition and nonreciprocal translocations is observed as well as whole chromosome loss, especially following a centromeric DSB. LOH spans megabases distal from the DSB, but also frequently tens of megabases centromere-proximal. Inhibition of microhomology-mediated end joining massively increases LOH, which is synergistically increased with concomitant inhibition of canonical nonhomologous end joining. The capacity for large-scale LOH must therefore be considered when using DSB-based gene editing, especially in conjunction with end joining inhibition.
Collapse
|
5
|
Mittal S, Arenkiel BR, Lyons-Warren AM. Arcuate dopaminergic/GABAergic neurons project within the hypothalamus and to the median eminence. J Neurophysiol 2024; 132:943-952. [PMID: 39108212 PMCID: PMC11427037 DOI: 10.1152/jn.00086.2024] [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: 02/29/2024] [Revised: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024] Open
Abstract
Cotransmission, meaning the release of multiple neurotransmitters from one synapse, allows for increased diversity of signaling in the brain. Dopamine (DA) and γ-aminobutyric acid (GABA) are known to coexpress in many regions such as the olfactory bulb and the ventral tegmental area. Tuberoinfundibular dopaminergic neurons (TIDA) in the arcuate nucleus of the hypothalamus (Arc) project to the median eminence (ME) and regulate prolactin release from the pituitary, and prior work suggests dopaminergic Arc neurons also cotransmit GABA. However, the extent of cotransmission, and the projection patterns of these neurons have not been fully revealed. Here, we used a genetic intersectional reporter expression approach to selectively label cells that express both tyrosine hydroxylase (TH) and vesicular GABA transporter (VGAT). Through this approach, we identified cells capable of both DA and GABA cotransmission in the Arc, periventricular (Pe), paraventricular (Pa), ventromedial, and the dorsolateral hypothalamic nuclei, in addition to a novel population in the caudate putamen. The highest density of labeled cells was in the Arc, 6.68% of DAPI-labeled cells at Bregma -2.06 mm, and in the Pe, 2.83% of DAPI-labeled cells at Bregma -1.94 mm. Next, we evaluated the projections of these DA/GABA cells by injecting an mCherry virus that fluoresces in DA/GABA cells. We observed a cotransmitting DA/GABA population, with projections within the Arc, and to the Pa and ME. These data suggest DA/GABA Arc neurons are involved in prolactin release as a subset of TIDA neurons. Further investigation will elucidate the interactions of dopamine and GABA in the hypothalamus.NEW & NOTEWORTHY Cotransmitting dopaminergic (DA) and γ-aminobutyric acid (GABA)ergic (DA/GABA) neurons contribute to the complexity of neural circuits. Using a new genetic technique, we characterized the locations, density, and projections of hypothalamic DA/GABA neurons. DA/GABA cells are mostly in the arcuate nucleus (Arc), from which they project locally within the arcuate, to the median eminence (ME), and to the paraventricular (Pa) nucleus. There is also a small and previously unreported group of DA/GABA cells in the caudate putamen.
Collapse
Affiliation(s)
- Somya Mittal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States
| | - Ariel M Lyons-Warren
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, United States
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
6
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
7
|
Lanza DG, Mao J, Lorenzo I, Liao L, Seavitt JR, Ljungberg MC, Simpson EM, DeMayo FJ, Heaney JD. An oocyte-specific Cas9-expressing mouse for germline CRISPR/Cas9-mediated genome editing. Genesis 2024; 62:e23589. [PMID: 38523431 PMCID: PMC10987075 DOI: 10.1002/dvg.23589] [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: 12/22/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/26/2024]
Abstract
Cas9 transgenes can be employed for genome editing in mouse zygotes. However, using transgenic instead of exogenous Cas9 to produce gene-edited animals creates unique issues including ill-defined transgene integration sites, the potential for prolonged Cas9 expression in transgenic embryos, and increased genotyping burden. To overcome these issues, we generated mice harboring an oocyte-specific, Gdf9 promoter driven, Cas9 transgene (Gdf9-Cas9) targeted as a single copy into the Hprt1 locus. The X-linked Hprt1 locus was selected because it is a defined integration site that does not influence transgene expression, and breeding of transgenic males generates obligate transgenic females to serve as embryo donors. Using microinjections and electroporation to introduce sgRNAs into zygotes derived from transgenic dams, we demonstrate that Gdf9-Cas9 mediates genome editing as efficiently as exogenous Cas9 at several loci. We show that genome editing efficiency is independent of transgene inheritance, verifying that maternally derived Cas9 facilitates genome editing. We also show that paternal inheritance of Gdf9-Cas9 does not mediate genome editing, confirming that Gdf9-Cas9 is not expressed in embryos. Finally, we demonstrate that off-target mutagenesis is equally rare when using transgenic or exogenous Cas9. Together, these results show that the Gdf9-Cas9 transgene is a viable alternative to exogenous Cas9.
Collapse
Affiliation(s)
- Denise G. Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA 77030
| | - Jianqiang Mao
- Department of Molecular & Cellular Biology, Baylor College of Medicine Houston, TX, USA 77030
| | - Isabel Lorenzo
- Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA 77030
| | - Lan Liao
- Department of Molecular & Cellular Biology, Baylor College of Medicine Houston, TX, USA 77030
| | - John R. Seavitt
- Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA 77030
- Present address: The Jackson Laboratory 600 Main St., Bar Harbor, Maine, ME, USA 04609
| | - M. Cecilia Ljungberg
- Department of Pediatrics – Neurology, Baylor College of Medicine Houston, TX, USA 77030
- Duncan Neurological Research Institute, Texas Children’s Hospital Houston, TX, USA 77030
| | - Elizabeth M. Simpson
- Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital Department of Medical Genetics, The University of British Columbia Vancouver, British Columbia V5Z 4H4, Canada
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory National Institute of Environmental Health Sciences Research Triangle Park, NC, USA 27709
| | - Jason D. Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA 77030
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, USA 77030
| |
Collapse
|
8
|
Hutchison V, Lynch A, Gutierrez-Gamez AM, Chen J. Inducible tricolor reporter mouse for parallel imaging of lysosomes, mitochondria, and microtubules. J Cell Biol 2024; 223:e202305086. [PMID: 37917008 PMCID: PMC10621751 DOI: 10.1083/jcb.202305086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/06/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Cell type-specific use of the same DNA blueprint generates diverse cell types. Such diversity must also be executed via differential deployment of the same subcellular machinery. However, our understanding of the size, distribution, and dynamics of subcellular machinery in native tissues and their connection to cellular diversity remains limited. We generate and characterize an inducible tricolor reporter mouse, dubbed "Kaleidoscope," for simultaneous imaging of lysosomes, mitochondria, and microtubules in any cell type and at a single-cell resolution. The expected subcellular compartments are labeled in culture and in tissues with no impact on cellular and organismal viability. Quantitative and live imaging of the tricolor reporter captures cell type-specific organelle features and kinetics in the lung, as well as their changes after Sendai virus infection. Yap/Taz mutant lung epithelial cells undergo accelerated lamellar body maturation, a subcellular manifestation of their molecular defects. A comprehensive toolbox of reporters for all subcellular structures is expected to transform our understanding of cell biology in tissues.
Collapse
Affiliation(s)
- Vera Hutchison
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Anne Lynch
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Lusk S, Ward CS, Chang A, Twitchell-Heyne A, Fattig S, Allen G, Jankowsky J, Ray R. An automated respiratory data pipeline for waveform characteristic analysis. J Physiol 2023; 601:4767-4806. [PMID: 37786382 PMCID: PMC10841337 DOI: 10.1113/jp284363] [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: 01/12/2023] [Accepted: 08/11/2023] [Indexed: 10/04/2023] Open
Abstract
Comprehensive and accurate analysis of respiratory and metabolic data is crucial to modelling congenital, pathogenic and degenerative diseases converging on autonomic control failure. A lack of tools for high-throughput analysis of respiratory datasets remains a major challenge. We present Breathe Easy, a novel open-source pipeline for processing raw recordings and associated metadata into operative outcomes, publication-worthy graphs and robust statistical analyses including QQ and residual plots for assumption queries and data transformations. This pipeline uses a facile graphical user interface for uploading data files, setting waveform feature thresholds and defining experimental variables. Breathe Easy was validated against manual selection by experts, which represents the current standard in the field. We demonstrate Breathe Easy's utility by examining a 2-year longitudinal study of an Alzheimer's disease mouse model to assess contributions of forebrain pathology in disordered breathing. Whole body plethysmography has become an important experimental outcome measure for a variety of diseases with primary and secondary respiratory indications. Respiratory dysfunction, while not an initial symptom in many of these disorders, often drives disability or death in patient outcomes. Breathe Easy provides an open-source respiratory analysis tool for all respiratory datasets and represents a necessary improvement upon current analytical methods in the field. KEY POINTS: Respiratory dysfunction is a common endpoint for disability and mortality in many disorders throughout life. Whole body plethysmography in rodents represents a high face-value method for measuring respiratory outcomes in rodent models of these diseases and disorders. Analysis of key respiratory variables remains hindered by manual annotation and analysis that leads to low throughput results that often exclude a majority of the recorded data. Here we present a software suite, Breathe Easy, that automates the process of data selection from raw recordings derived from plethysmography experiments and the analysis of these data into operative outcomes and publication-worthy graphs with statistics. We validate Breathe Easy with a terabyte-scale Alzheimer's dataset that examines the effects of forebrain pathology on respiratory function over 2 years of degeneration.
Collapse
Affiliation(s)
- Savannah Lusk
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher S. Ward
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andersen Chang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Shaun Fattig
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Genevera Allen
- Departments of Electrical and Computer Engineering, Statistics, and Computer Science, Rice University, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Joanna Jankowsky
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Russell Ray
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- McNair Medical Institute, Houston, TX 77030, USA
| |
Collapse
|
10
|
McKinney A, Hu M, Hoskins A, Mohammadyar A, Naeem N, Jing J, Patel SS, Sheth BR, Jiang X. Cellular composition and circuit organization of the locus coeruleus of adult mice. eLife 2023; 12:e80100. [PMID: 36734517 PMCID: PMC9934863 DOI: 10.7554/elife.80100] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 02/01/2023] [Indexed: 02/04/2023] Open
Abstract
The locus coeruleus (LC) houses the vast majority of noradrenergic neurons in the brain and regulates many fundamental functions, including fight and flight response, attention control, and sleep/wake cycles. While efferent projections of the LC have been extensively investigated, little is known about its local circuit organization. Here, we performed large-scale multipatch recordings of noradrenergic neurons in adult mouse LC to profile their morpho-electric properties while simultaneously examining their interactions. LC noradrenergic neurons are diverse and could be classified into two major morpho-electric types. While fast excitatory synaptic transmission among LC noradrenergic neurons was not observed in our preparation, these mature LC neurons connected via gap junction at a rate similar to their early developmental stage and comparable to other brain regions. Most electrical connections form between dendrites and are restricted to narrowly spaced pairs or small clusters of neurons of the same type. In addition, more than two electrically coupled cell pairs were often identified across a cohort of neurons from individual multicell recording sets that followed a chain-like organizational pattern. The assembly of LC noradrenergic neurons thus follows a spatial and cell-type-specific wiring principle that may be imposed by a unique chain-like rule.
Collapse
Affiliation(s)
- Andrew McKinney
- Neuroscience Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Ming Hu
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | | | | | | | - Junzhan Jing
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Saumil S Patel
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Bhavin R Sheth
- Department of Electrical and Computer Engineering, University of HoustonHoustonUnited States
- Center for NeuroEngineering and Cognitive Science, University of HoustonHoustonUnited States
| | - Xiaolong Jiang
- Neuroscience Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Ophthalmology, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
11
|
Ncube D, Tallafuss A, Serafin J, Bruckner J, Farnsworth DR, Miller AC, Eisen JS, Washbourne P. A conserved transcriptional fingerprint of multi-neurotransmitter neurons necessary for social behavior. BMC Genomics 2022; 23:675. [PMID: 36175871 PMCID: PMC9523972 DOI: 10.1186/s12864-022-08879-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Background An essential determinant of a neuron’s functionality is its neurotransmitter phenotype. We previously identified a defined subpopulation of cholinergic neurons required for social orienting behavior in zebrafish. Results We transcriptionally profiled these neurons and discovered that they are capable of synthesizing both acetylcholine and GABA. We also established a constellation of transcription factors and neurotransmitter markers that can be used as a “transcriptomic fingerprint” to recognize a homologous neuronal population in another vertebrate. Conclusion Our results suggest that this transcriptomic fingerprint and the cholinergic-GABAergic neuronal subtype that it defines are evolutionarily conserved. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08879-w.
Collapse
Affiliation(s)
- Denver Ncube
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Alexandra Tallafuss
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Jen Serafin
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Joseph Bruckner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Dylan R Farnsworth
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Judith S Eisen
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Philip Washbourne
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
12
|
Hunt PJ, Kochukov M, Pekarek BT, Belfort BD, Romero JM, Swanson JL, Arenkiel BR. Co-transmitting neurons in the lateral septal nucleus exhibit features of neurotransmitter switching. IBRO Neurosci Rep 2022; 12:390-398. [PMID: 35601692 PMCID: PMC9121281 DOI: 10.1016/j.ibneur.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/09/2022] [Indexed: 01/07/2023] Open
Abstract
The lateral septal nucleus (LSN) is a highly interconnected region of the central brain whose activity regulates widespread circuitry. As such, the mechanisms that govern neuronal activity within the LSN have far-reaching implications on numerous brain-wide nuclei, circuits, and behaviors. We found that GABAergic neurons within the LSN express markers that mediate the release of acetylcholine (ACh). Moreover, we show that these vGATLSN neurons release both GABA and ACh onto local glutamatergic LSN neurons. Using both short-term and long-term neuronal labeling techniques we observed expression of the cholinergic neuron marker Choline Acetyltransferase (ChAT) in vGATLSN neurons. These findings provide evidence of cholinergic neurotransmission from vGATLSN neurons, and provide an impetus to examine dynamic co-neurotransmission changes as a potential mechanism that contributes to neuronal and circuit-wide plasticity within the LSN.
Collapse
Affiliation(s)
- Patrick J. Hunt
- Genetics and Genomics Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Mikhail Kochukov
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Brandon T. Pekarek
- Genetics and Genomics Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Benjamin D.W. Belfort
- Genetics and Genomics Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Juan M. Romero
- Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Neuroscience Graduate Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Jessica L. Swanson
- Genetics and Genomics Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund Street, Houston, TX 77030, USA
| |
Collapse
|