1
|
Gomez-Frittelli J, Devienne GF, Travis L, Kyloh MA, Duan X, Hibberd TJ, Spencer NJ, Huguenard JR, Kaltschmidt JA. Synaptic cell adhesion molecule Cdh6 identifies a class of sensory neurons with novel functions in colonic motility. eLife 2025; 13:RP101043. [PMID: 40193178 PMCID: PMC11975370 DOI: 10.7554/elife.101043] [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] [Indexed: 04/09/2025] Open
Abstract
Intrinsic sensory neurons are an essential part of the enteric nervous system (ENS) and play a crucial role in gastrointestinal tract motility and digestion. Neuronal subtypes in the ENS have been distinguished by their electrophysiological properties, morphology, and expression of characteristic markers, notably neurotransmitters and neuropeptides. Here, we investigated synaptic cell adhesion molecules as novel cell-type markers in the ENS. Our work identifies two type II classic cadherins, Cdh6 and Cdh8, specific to sensory neurons in the mouse colon. We show that Cdh6+ neurons demonstrate all other distinguishing classifications of enteric sensory neurons including marker expression of Calcb and Nmu, Dogiel type II morphology and AH-type electrophysiology and IH current. Optogenetic activation of Cdh6+ sensory neurons in distal colon evokes retrograde colonic motor complexes (CMCs), while pharmacologic blockade of rhythmicity-associated current IH disrupts the spontaneous generation of CMCs. These findings provide the first demonstration of selective activation of a single neurochemical and functional class of enteric neurons and demonstrate a functional and critical role for sensory neurons in the generation of CMCs.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford UniversityStanfordUnited States
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
| | - Gabrielle Frederique Devienne
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurology & Neurological Sciences, Stanford UniversityStanfordUnited States
| | - Lee Travis
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Melinda A Kyloh
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San FranciscoSan FranciscoUnited States
| | - Tim J Hibberd
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - John R Huguenard
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurology & Neurological Sciences, Stanford UniversityStanfordUnited States
| | - Julia A Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurosurgery, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
2
|
Hamnett R, Bendrick JL, Saha Z, Robertson K, Lewis CM, Marciano JH, Zhao ET, Kaltschmidt JA. Enteric glutamatergic interneurons regulate intestinal motility. Neuron 2025; 113:1019-1035.e6. [PMID: 39983724 PMCID: PMC11968238 DOI: 10.1016/j.neuron.2025.01.014] [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: 05/07/2024] [Revised: 11/14/2024] [Accepted: 01/23/2025] [Indexed: 02/23/2025]
Abstract
The enteric nervous system (ENS) controls digestion autonomously via a complex neural network within the gut wall. Enteric neurons expressing glutamate have been identified by transcriptomic studies as a distinct subpopulation, and glutamate can affect intestinal motility by modulating enteric neuron activity. However, the nature of glutamatergic neurons, their position within the ENS circuit, and their function in regulating gut motility are unknown. We identify glutamatergic neurons as longitudinally projecting descending interneurons in the small intestine and colon and as a novel class of circumferential neurons only in the colon. Both populations make synaptic contact with diverse neuronal subtypes and signal with multiple neurotransmitters and neuropeptides in addition to glutamate, including acetylcholine and enkephalin. Knocking out the glutamate transporter VGLUT2 from enkephalin neurons disrupts gastrointestinal transit, while ex vivo optogenetic stimulation of glutamatergic neurons initiates colonic propulsive motility. Our results posit glutamatergic neurons as key interneurons that regulate intestinal motility.
Collapse
Affiliation(s)
- Ryan Hamnett
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| | - Jacqueline L Bendrick
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Zinnia Saha
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Cheyanne M Lewis
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Jack H Marciano
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Eric Tianjiao Zhao
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Morales-Soto W, Smith-Edwards KM. Unique properties of proximal and distal colon reflect distinct motor functions. Am J Physiol Gastrointest Liver Physiol 2025; 328:G448-G454. [PMID: 40095602 DOI: 10.1152/ajpgi.00215.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/19/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
The gastrointestinal tract is made up of specialized organs that work in tandem to facilitate digestion. The colon regulates the final steps in this process where complex motor patterns in proximal regions facilitate the formation of fecal pellets that are propelled along the distal colon via self-sustaining neural peristalsis and temporarily stored before defecation. Historically, our understanding of colonic motility has focused primarily on distal regions, and the intrinsic reflex circuits of the enteric nervous system involved in neural peristalsis have been defined, but we do not yet have a clear grasp on the mechanisms orchestrating motor function in proximal regions. New approaches have brought to the forefront the unique structural, neurochemical, and functional characteristics that exist in distinct regions of the mouse and human colon. In this mini-review, we highlight key differences along the proximal-distal colonic axis and discuss how these differences relate to region-specific motor function.
Collapse
Affiliation(s)
- Wilmarie Morales-Soto
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Enteric Neurosciences Program, Mayo Clinic, Rochester, Minnesota, United States
| | - Kristen M Smith-Edwards
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
- Enteric Neurosciences Program, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
4
|
Zipf D, Vogt M, Sathyanarayanan U, Wagdi A, Riebeling J, Patejdl R, Bruegmann T. Direct Stimulation of Gastric Smooth Muscle Cells via G q Proteins With Light. Neurogastroenterol Motil 2025:e70028. [PMID: 40159839 DOI: 10.1111/nmo.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Optogenetics is a cutting-edge approach that can enable direct stimulation of gastric smooth muscle cells (SMC) by combining cell-specific overexpression of light-sensitive proteins with light stimulation. We previously demonstrated that direct optogenetic stimulation of gastric SMC via depolarization can restore contractility and food propulsion and could become a new treatment strategy for gastroparesis. The human receptor Neuropsin (hOPN5) enables activation of Gq signaling with UV light. Herein, we explore this new strategy for direct optogenetic stimulation of gastric SMC. METHODS We used a transgenic mouse model expressing hOPN5 in fusion with eYFP. Antral longitudinal smooth muscle strips were used for isometric force measurements and whole stomachs for intragastric pressure measurements, comparing light stimulation to other stimuli. Adeno-associated virus (AAV) serotypes were screened for efficiency in transducing cultured gastric SMC, and transduced cells were tested by Ca2+ imaging. RESULTS hOPN5 expression was restricted to and found in ~1/3 of SMC in the stomach. UV light induced isometric force and increased intragastric pressure only in transgenic mice similarly to electrical field stimulation and reached approximately 1/3 of the force induced by global depolarization and muscarinic receptor activation. Importantly, optical stimulation remained effective in an ex vivo gastroparesis model. AAV 2.5 was by far the most effective serotype for SMC transduction, and UV light triggered Ca2+ transients in SMC expressing hOPN5. CONCLUSION hOPN5 is a new and effective tool to directly stimulate gastric SMC to control contractility with light. Thus, it is an additional and complementary approach to light-induced membrane depolarization to restore gastric motility.
Collapse
Affiliation(s)
- David Zipf
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Vogt
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Ahmed Wagdi
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Germany, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Johannes Riebeling
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
- Department of General, Visceral and Paediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Else Kröner Fresenius Center for Optogenetic Therapies, University Medical Center Göttingen, Göttingen, Germany
| | - Robert Patejdl
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, University of Rostock, Göttingen, Germany
- Department of Medicine, Health and Medical University Erfurt, Erfurt, Germany
| | - Tobias Bruegmann
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Lower Saxony, Germany, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Else Kröner Fresenius Center for Optogenetic Therapies, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Xie Z, Rose L, Feng J, Zhao Y, Lu Y, Kane H, Hibberd TJ, Hu X, Wang Z, Zang K, Yang X, Richardson Q, Othman R, Venezia O, Zhakyp A, Gao F, Abe N, Vigeland K, Wang H, Branch C, Duizer C, Deng L, Meng X, Zamidar L, Hauptschein M, Bergin R, Dong X, Chiu IM, Kim BS, Spencer NJ, Hu H, Jackson R. Enteric neuronal Piezo1 maintains mechanical and immunological homeostasis by sensing force. Cell 2025:S0092-8674(25)00258-2. [PMID: 40132579 DOI: 10.1016/j.cell.2025.02.031] [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: 05/06/2024] [Revised: 11/19/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
The gastrointestinal (GI) tract experiences a myriad of mechanical forces while orchestrating digestion and barrier immunity. A central conductor of these processes, the enteric nervous system (ENS), detects luminal pressure to regulate peristalsis independently of extrinsic input from the central and peripheral nervous systems. However, how the ∼500 million enteric neurons that reside in the GI tract sense and respond to force remains unknown. Herein, we establish that the mechanosensor Piezo1 is functionally expressed in cholinergic enteric neurons. Optogenetic stimulation of Piezo1+ cholinergic enteric neurons drives colonic motility, while Piezo1 deficiency reduces cholinergic neuronal activity and slows peristalsis. Additionally, Piezo1 deficiency in cholinergic enteric neurons abolishes exercise-induced acceleration of GI motility. Finally, we uncover that enteric neuronal Piezo1 function is required for motility alterations in colitis and acts to prevent aberrant inflammation and tissue damage. This work uncovers how the ENS senses and responds to mechanical force.
Collapse
Affiliation(s)
- Zili Xie
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Lillian Rose
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Zhao
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Yisi Lu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Harry Kane
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Xueming Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Zhen Wang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Kaikai Zang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Xingliang Yang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | | | - Rahmeh Othman
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia Venezia
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ademi Zhakyp
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Gao
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Nobuya Abe
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keren Vigeland
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hongshen Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Camren Branch
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Coco Duizer
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Meng
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Lydia Zamidar
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Max Hauptschein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronan Bergin
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Issac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian S Kim
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hongzhen Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Gomez-Frittelli J, Devienne G, Travis L, Kyloh MA, Duan X, Hibberd TJ, Spencer NJ, Huguenard JR, Kaltschmidt JA. Synaptic cell adhesion molecule Cdh6 identifies a class of sensory neurons with novel functions in colonic motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606748. [PMID: 39149241 PMCID: PMC11326146 DOI: 10.1101/2024.08.06.606748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Intrinsic sensory neurons are an essential part of the enteric nervous system (ENS) and play a crucial role in gastrointestinal tract motility and digestion. Neuronal subtypes in the ENS have been distinguished by their electrophysiological properties, morphology, and expression of characteristic markers, notably neurotransmitters and neuropeptides. Here we investigated synaptic cell adhesion molecules as novel cell type markers in the ENS. Our work identifies two Type II classic cadherins, Cdh6 and Cdh8, specific to sensory neurons in the mouse colon. We show that Cdh6+ neurons demonstrate all other distinguishing classifications of enteric sensory neurons including marker expression of Calcb and Nmu, Dogiel type II morphology and AH-type electrophysiology and I H current. Optogenetic activation of Cdh6+ sensory neurons in distal colon evokes retrograde colonic motor complexes (CMCs), while pharmacologic blockade of rhythmicity-associated current I H disrupts the spontaneous generation of CMCs. These findings provide the first demonstration of selective activation of a single neurochemical and functional class of enteric neurons, and demonstrate a functional and critical role for sensory neurons in the generation of CMCs.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford University; Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
| | - Gabrielle Devienne
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Melinda A. Kyloh
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San Francisco; San Francisco, CA, USA
| | - Tim J. Hibberd
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Nick J. Spencer
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - John R. Huguenard
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
7
|
Rykalo N, Riehl L, Kress M. The gut microbiome and the brain. Curr Opin Support Palliat Care 2024; 18:282-291. [PMID: 39250732 DOI: 10.1097/spc.0000000000000717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW The importance of the gut microbiome for human health and well-being is generally accepted, and elucidating the signaling pathways between the gut microbiome and the host offers novel mechanistic insight into the (patho)physiology and multifaceted aspects of healthy aging and human brain functions. RECENT FINDINGS The gut microbiome is tightly linked with the nervous system, and gut microbiota are increasingly emerging as important regulators of emotional and cognitive performance. They send and receive signals for the bidirectional communication between gut and brain via immunological, neuroanatomical, and humoral pathways. The composition of the gut microbiota and the spectrum of metabolites and neurotransmitters that they release changes with increasing age, nutrition, hypoxia, and other pathological conditions. Changes in gut microbiota (dysbiosis) are associated with critical illnesses such as cancer, cardiovascular, and chronic kidney disease but also neurological, mental, and pain disorders, as well as chemotherapies and antibiotics affecting brain development and function. SUMMARY Dysbiosis and a concomitant imbalance of mediators are increasingly emerging both as causes and consequences of diseases affecting the brain. Understanding the microbiota's role in the pathogenesis of these disorders will have major clinical implications and offer new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Nadiia Rykalo
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University Innsbruck, Austria
| | | | | |
Collapse
|
8
|
Wang J, Xiao J. Insights Into Gastrointestinal Motility Through the Use of Optogenetic Sensors. J Neurogastroenterol Motil 2024; 30:512-514. [PMID: 39397627 PMCID: PMC11474550 DOI: 10.5056/jnm24038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Affiliation(s)
- Jing Wang
- Department of Gastroenterology and Hubei Provincial Center and Key Laboratory for the Intestinal and Colorectal Disease, Wuhan University Zhongnan Hospital, Wuhan, China
| | - Jun Xiao
- Department of Gastroenterology and Hubei Provincial Center and Key Laboratory for the Intestinal and Colorectal Disease, Wuhan University Zhongnan Hospital, Wuhan, China
| |
Collapse
|
9
|
Inaba M. Optogenetic techniques for understanding the gut peristalsis during chicken embryonic development. Biochem Soc Trans 2024; 52:1727-1735. [PMID: 39051133 DOI: 10.1042/bst20231337] [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: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Gut peristaltic movements transport ingested materials along the gut axis, which is critical for food digestion and nutrient absorption. While a large amount of studies have been devoted to analyzing the physiological functions of peristalsis in adults, little is known about how the peristaltic system is established during embryogenesis. In recent years, the chicken developing gut has emerged as an excellent model, in which specific sites along the gut axis can be genetically labeled enabling live imaging and optogenetic analyses. This review provides an overview of recent progress in optogenetic studies of gut peristalsis. Analyses with an improved channelrhodopsin-2 variant demonstrated that the peristalsis can artificially be generated in the developing gut. These studies unveiled novel functional coordination between different regions along the gut axis. In addition, imaging with GCaMP6s, a genetically encoded calcium indicator, enabled a fine mapping of developmental changes in the peristaltic patterns as Ca2+ signals. These advanced techniques will broaden our knowledge of how embryonic peristalsis is established at the cellular and molecular level, leading to the understanding of physiological and pathological processes in adult peristalsis.
Collapse
Affiliation(s)
- Masafumi Inaba
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
10
|
Nan K, Wong K, Li D, Ying B, McRae JC, Feig VR, Wang S, Du N, Liang Y, Mao Q, Zhou E, Chen Y, Sang L, Yao K, Zhou J, Li J, Jenkins J, Ishida K, Kuosmanen J, Mohammed Madani WA, Hayward A, Ramadi KB, Yu X, Traverso G. An ingestible, battery-free, tissue-adhering robotic interface for non-invasive and chronic electrostimulation of the gut. Nat Commun 2024; 15:6749. [PMID: 39117667 PMCID: PMC11310346 DOI: 10.1038/s41467-024-51102-5] [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: 10/27/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Ingestible electronics have the capacity to transform our ability to effectively diagnose and potentially treat a broad set of conditions. Current applications could be significantly enhanced by addressing poor electrode-tissue contact, lack of navigation, short dwell time, and limited battery life. Here we report the development of an ingestible, battery-free, and tissue-adhering robotic interface (IngRI) for non-invasive and chronic electrostimulation of the gut, which addresses challenges associated with contact, navigation, retention, and powering (C-N-R-P) faced by existing ingestibles. We show that near-field inductive coupling operating near 13.56 MHz was sufficient to power and modulate the IngRI to deliver therapeutically relevant electrostimulation, which can be further enhanced by a bio-inspired, hydrogel-enabled adhesive interface. In swine models, we demonstrated the electrical interaction of IngRI with the gastric mucosa by recording conductive signaling from the subcutaneous space. We further observed changes in plasma ghrelin levels, the "hunger hormone," while IngRI was activated in vivo, demonstrating its clinical potential in regulating appetite and treating other endocrine conditions. The results of this study suggest that concepts inspired by soft and wireless skin-interfacing electronic devices can be applied to ingestible electronics with potential clinical applications for evaluating and treating gastrointestinal conditions.
Collapse
Affiliation(s)
- Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kiwan Wong
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dengfeng Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong SAR, China
| | - Binbin Ying
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James C McRae
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vivian R Feig
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shubing Wang
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ningjie Du
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuelong Liang
- Department of General Surgery, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qijiang Mao
- Department of General Surgery, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Enjie Zhou
- Department of General Surgery, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yonglin Chen
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lei Sang
- School of Microelectronics, Hefei University of Technology, Hefei, 230601, China
| | - Kuanming Yao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Jingkun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong SAR, China
| | - Jian Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong SAR, China
| | - Joshua Jenkins
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keiko Ishida
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Johannes Kuosmanen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wiam Abdalla Mohammed Madani
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alison Hayward
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Khalil B Ramadi
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
- Tandon School of Engineering, New York University, New York, NY, USA
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China.
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong SAR, China.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, USA.
| |
Collapse
|
11
|
Rahman AA, Ohkura T, Bhave S, Pan W, Ohishi K, Ott L, Han C, Leavitt A, Stavely R, Burns AJ, Goldstein AM, Hotta R. Enteric neural stem cell transplant restores gut motility in mice with Hirschsprung disease. JCI Insight 2024; 9:e179755. [PMID: 39042470 PMCID: PMC11385093 DOI: 10.1172/jci.insight.179755] [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: 01/26/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
The goal of this study was to determine if transplantation of enteric neural stem cells (ENSCs) can rescue the enteric nervous system, restore gut motility, reduce colonic inflammation, and improve survival in the Ednrb-KO mouse model of Hirschsprung disease (HSCR). ENSCs were isolated from mouse intestine, expanded to form neurospheres, and microinjected into the colons of recipient Ednrb-KO mice. Transplanted ENSCs were identified in recipient colons as cell clusters in "neo-ganglia." Immunohistochemical evaluation demonstrated extensive cell migration away from the sites of cell delivery and across the muscle layers. Electrical field stimulation and optogenetics showed significantly enhanced contractile activity of aganglionic colonic smooth muscle following ENSC transplantation and confirmed functional neuromuscular integration of the transplanted ENSC-derived neurons. ENSC injection also partially restored the colonic migrating motor complex. Histological examination revealed a significant reduction in inflammation in ENSC-transplanted aganglionic recipient colon compared with that of sham-operated mice. Interestingly, mice that received cell transplant also had prolonged survival compared with controls. This study demonstrates that ENSC transplantation can improve outcomes in HSCR by restoring gut motility and reducing the severity of Hirschsprung-associated enterocolitis, the leading cause of death in human HSCR.
Collapse
Affiliation(s)
- Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Leah Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abigail Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Sahasrabudhe A, Rupprecht LE, Orguc S, Khudiyev T, Tanaka T, Sands J, Zhu W, Tabet A, Manthey M, Allen H, Loke G, Antonini MJ, Rosenfeld D, Park J, Garwood IC, Yan W, Niroui F, Fink Y, Chandrakasan A, Bohórquez DV, Anikeeva P. Multifunctional microelectronic fibers enable wireless modulation of gut and brain neural circuits. Nat Biotechnol 2024; 42:892-904. [PMID: 37349522 PMCID: PMC11180606 DOI: 10.1038/s41587-023-01833-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Progress in understanding brain-viscera interoceptive signaling is hindered by a dearth of implantable devices suitable for probing both brain and peripheral organ neurophysiology during behavior. Here we describe multifunctional neural interfaces that combine the scalability and mechanical versatility of thermally drawn polymer-based fibers with the sophistication of microelectronic chips for organs as diverse as the brain and the gut. Our approach uses meters-long continuous fibers that can integrate light sources, electrodes, thermal sensors and microfluidic channels in a miniature footprint. Paired with custom-fabricated control modules, the fibers wirelessly deliver light for optogenetics and transfer data for physiological recording. We validate this technology by modulating the mesolimbic reward pathway in the mouse brain. We then apply the fibers in the anatomically challenging intestinal lumen and demonstrate wireless control of sensory epithelial cells that guide feeding behaviors. Finally, we show that optogenetic stimulation of vagal afferents from the intestinal lumen is sufficient to evoke a reward phenotype in untethered mice.
Collapse
Affiliation(s)
- Atharva Sahasrabudhe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura E Rupprecht
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Sirma Orguc
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tural Khudiyev
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tomo Tanaka
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Secure System Platform Research Laboratories, NEC Corporation, Kawasaki, Japan
| | - Joanna Sands
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Weikun Zhu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harrison Allen
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gabriel Loke
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc-Joseph Antonini
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jimin Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Indie C Garwood
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Wei Yan
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Farnaz Niroui
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anantha Chandrakasan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Nagahawatte ND, Avci R, Angeli-Gordon TR, Paskaranandavadivel N, Cheng LK. High-Energy Pacing in the Jejunum Elicits Pulsatile Segmental Contractions. IEEE Trans Biomed Eng 2024; 71:750-757. [PMID: 37729575 DOI: 10.1109/tbme.2023.3317400] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Compromised bowel function is associated with a range of motility disorders such as post-operative ileus and chronic intestinal pseudo-obstruction. Disordered or weak motility compromise the efficient movement of luminal contents necessary for digestion and nutrient absorption. This study investigated the potential of high-energy pacing to enhance contractions in the proximal jejunum of the small intestine. METHODS Pacing pulse parameters (pulse-width: 100 ms, 200 ms, 400 ms, pulse-amplitude: 4 mA, 6 mA, 8 mA) were systematically varied in the in vivo porcine jejunum (n = 7) and the induced contractile responses were evaluated using a video mapping system. Localized segmental contractions were quantified by measuring the intestinal diameter and thereby computing the strain. The impact of pacing parameters on contractile strain was investigated. Finally, histological studies were conducted on paced tissue to assess for potential tissue damage. RESULTS Segmental contractions were successfully induced at all pulse-settings and evaluated across 67 pacing sessions. In response to pacing, the intestine segment at the site of pacing contracted, with diameter reduced by 6-18%. Contractile response significantly increased with increasing pulse-amplitude. However, with increasing pulse-width, the increase in contractile response was significant only between 100 ms and 400 ms. Histology showed no tissue damage occurred when maximal pacing energy (pulse-amplitude = 4-8 mA, pulse-width = 400 ms, 5 minute duration) was applied. CONCLUSION High-energy pacing induced periodic segmental contractions in response to pacing pulses and the contractile strain was proportional to the energy applied on the intestine. The ability to enhance motility through pacing may hold promising therapeutic potential for bowel disorders and awaits clinical translation. SIGNIFICANCE Small intestine pacing elicits localized segmental contractions which increase in magnitude with increasing pulse settings. This study marks the first adaptation of video mapping techniques to track the pacing response in the small intestine.
Collapse
|
14
|
Riehl L, Fürst J, Kress M, Rykalo N. The importance of the gut microbiome and its signals for a healthy nervous system and the multifaceted mechanisms of neuropsychiatric disorders. Front Neurosci 2024; 17:1302957. [PMID: 38249593 PMCID: PMC10797776 DOI: 10.3389/fnins.2023.1302957] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Increasing evidence links the gut microbiome and the nervous system in health and disease. This narrative review discusses current views on the interaction between the gut microbiota, the intestinal epithelium, and the brain, and provides an overview of the communication routes and signals of the bidirectional interactions between gut microbiota and the brain, including circulatory, immunological, neuroanatomical, and neuroendocrine pathways. Similarities and differences in healthy gut microbiota in humans and mice exist that are relevant for the translational gap between non-human model systems and patients. There is an increasing spectrum of metabolites and neurotransmitters that are released and/or modulated by the gut microbiota in both homeostatic and pathological conditions. Dysbiotic disruptions occur as consequences of critical illnesses such as cancer, cardiovascular and chronic kidney disease but also neurological, mental, and pain disorders, as well as ischemic and traumatic brain injury. Changes in the gut microbiota (dysbiosis) and a concomitant imbalance in the release of mediators may be cause or consequence of diseases of the central nervous system and are increasingly emerging as critical links to the disruption of healthy physiological function, alterations in nutrition intake, exposure to hypoxic conditions and others, observed in brain disorders. Despite the generally accepted importance of the gut microbiome, the bidirectional communication routes between brain and gut are not fully understood. Elucidating these routes and signaling pathways in more detail offers novel mechanistic insight into the pathophysiology and multifaceted aspects of brain disorders.
Collapse
Affiliation(s)
| | | | | | - Nadiia Rykalo
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Jiang S, Wu X, Yang F, Rommelfanger NJ, Hong G. Activation of mechanoluminescent nanotransducers by focused ultrasound enables light delivery to deep-seated tissue in vivo. Nat Protoc 2023; 18:3787-3820. [PMID: 37914782 PMCID: PMC11405139 DOI: 10.1038/s41596-023-00895-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/27/2023] [Indexed: 11/03/2023]
Abstract
Light is used extensively in biological and medical research for optogenetic neuromodulation, fluorescence imaging, photoactivatable gene editing and light-based therapies. The major challenge to the in vivo implementation of light-based methods in deep-seated structures of the brain or of internal organs is the limited penetration of photons in biological tissue. The presence of light scattering and absorption has resulted in the development of invasive techniques such as the implantation of optical fibers, the insertion of endoscopes and the surgical removal of overlying tissues to overcome light attenuation and deliver it deep into the body. However, these procedures are highly invasive and make it difficult to reposition and adjust the illuminated area in each animal. Here, we detail a noninvasive approach to deliver light (termed 'deLight') in deep tissue via systemically injected mechanoluminescent nanotransducers that can be gated by using focused ultrasound. This approach achieves localized light emission with sub-millimeter resolution and millisecond response times in any vascularized organ of living mice without requiring invasive implantation of light-emitting devices. For example, deLight enables optogenetic neuromodulation in live mice without a craniotomy or brain implants. deLight provides a generalized method for applications that require a light source in deep tissues in vivo, such as deep-brain fluorescence imaging and photoactivatable genome editing. The implementation of the entire protocol for an in vivo application takes ~1-2 weeks.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Xiang Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Fan Yang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Nicholas J Rommelfanger
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Cassidy RM, Flores EM, Trinh Nguyen AK, Cheruvu SS, Uribe RA, Krachler AM, Odem MA. Systematic analysis of proximal midgut- and anorectal-originating contractions in larval zebrafish using event feature detection and supervised machine learning algorithms. Neurogastroenterol Motil 2023; 35:e14675. [PMID: 37743702 PMCID: PMC10841157 DOI: 10.1111/nmo.14675] [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] [Received: 08/12/2021] [Revised: 07/16/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Zebrafish larvae are translucent, allowing in vivo analysis of gut development and physiology, including gut motility. While recent progress has been made in measuring gut motility in larvae, challenges remain which can influence results, such as how data are interpreted, opportunities for technical user error, and inconsistencies in methods. METHODS To overcome these challenges, we noninvasively introduced Nile Red fluorescent dye to fill the intraluminal gut space in zebrafish larvae and collected serial confocal microscopic images of gut fluorescence. We automated the detection of fluorescent-contrasted contraction events against the median-subtracted signal and compared it to manually annotated gut contraction events across anatomically defined gut regions. Supervised machine learning (multiple logistic regression) was then used to discriminate between true contraction events and noise. To demonstrate, we analyzed motility in larvae under control and reserpine-treated conditions. We also used automated event detection analysis to compare unfed and fed larvae. KEY RESULTS Automated analysis retained event features for proximal midgut-originating retrograde and anterograde contractions and anorectal-originating retrograde contractions. While manual annotation showed reserpine disrupted gut motility, machine learning only achieved equivalent contraction discrimination in controls and failed to accurately identify contractions after reserpine due to insufficient intraluminal fluorescence. Automated analysis also showed feeding had no effect on the frequency of anorectal-originating contractions. CONCLUSIONS & INFERENCES Automated event detection analysis rapidly and accurately annotated contraction events, including the previously neglected phenomenon of anorectal contractions. However, challenges remain to discriminate contraction events based on intraluminal fluorescence under treatment conditions that disrupt functional motility.
Collapse
Affiliation(s)
- Ryan M. Cassidy
- Brown Foundation Institute of Molecular Medicine, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Erika M. Flores
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Anh K. Trinh Nguyen
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Sai S. Cheruvu
- Department of Integrative Biology and Pharmacology,
McGovern Medical School at UTHealth, Houston, TX 77030, USA
| | - Rosa A. Uribe
- Department of Biosciences, Rice University, Houston, TX
77005, USA
| | - Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Max A. Odem
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| |
Collapse
|
17
|
Hotta R, Rahman A, Bhave S, Stavely R, Pan W, Srinivasan S, de Couto G, Rodriguez-Borlado L, Myers R, Burns AJ, Goldstein AM. Transplanted ENSCs form functional connections with intestinal smooth muscle and restore colonic motility in nNOS-deficient mice. Stem Cell Res Ther 2023; 14:232. [PMID: 37667277 PMCID: PMC10478362 DOI: 10.1186/s13287-023-03469-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Enteric neuropathies, which result from abnormalities of the enteric nervous system, are associated with significant morbidity and high health-care costs, but current treatments are unsatisfactory. Cell-based therapy offers an innovative approach to replace the absent or abnormal enteric neurons and thereby restore gut function. METHODS Enteric neuronal stem cells (ENSCs) were isolated from the gastrointestinal tract of Wnt1-Cre;R26tdTomato mice and generated neurospheres (NS). NS transplants were performed via injection into the mid-colon mesenchyme of nNOS-/- mouse, a model of colonic dysmotility, using either 1 (n = 12) or 3 (n = 12) injections (30 NS per injection) targeted longitudinally 1-2 mm apart. Functional outcomes were assessed up to 6 weeks later using electromyography (EMG), electrical field stimulation (EFS), optogenetics, and by measuring colorectal motility. RESULTS Transplanted ENSCs formed nitrergic neurons in the nNOS-/- recipient colon. Multiple injections of ENSCs resulted in a significantly larger area of coverage compared to single injection alone and were associated with a marked improvement in colonic function, demonstrated by (1) increased colonic muscle activity by EMG recording, (2) faster rectal bead expulsion, and (3) increased fecal pellet output in vivo. Organ bath studies revealed direct neuromuscular communication by optogenetic stimulation of channelrhodopsin-expressing ENSCs and restoration of smooth muscle relaxation in response to EFS. CONCLUSIONS These results demonstrate that transplanted ENSCs can form effective neuromuscular connections and improve colonic motor function in a model of colonic dysmotility, and additionally reveal that multiple sites of cell delivery led to an improved response, paving the way for optimized clinical trial design.
Collapse
Affiliation(s)
- Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Ahmed Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Shriya Srinivasan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Geoffrey de Couto
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Luis Rodriguez-Borlado
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Richard Myers
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Alan J Burns
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Zhao S, Tong W. An Optogenetics-based Approach to Regulate Colonic Contractions by Modulating the Activity of the Interstitial Cells of Cajal in Mice. J Neurogastroenterol Motil 2023; 29:388-399. [PMID: 37417266 PMCID: PMC10334192 DOI: 10.5056/jnm22181] [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: 10/25/2022] [Revised: 02/16/2023] [Accepted: 03/26/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND/AIMS The interstitial cells of Cajal (ICC) are pacemaker cells in the gastrointestinal (GI) tract. We examined whether the activity of ICC could be stimulated to control colonic contractions. An optogenetics-based mouse model in which the light-sensitive protein channelrhodopsin-2 (ChR2) was expressed was used to accomplish cell specific, direct stimulation of ICC. METHODS An inducible site-specific Cre-loxP recombination system was used to generate KitCreERT2/+;ROSAChR2(H134R)/tdTomato/+ mice in which ChR2(H134R), a variant of ChR2, was genetically expressed in ICC after tamoxifen administration. Genotyping and immunofluorescence analysis were performed to confirm gene fusion and expression. Isometric force recordings were performed to measure changes in contractions in the colonic muscle strips. RESULTS ChR2 was specifically expressed in Kit-labeled ICC. The isometric force recordings showed that the contractions of the colonic muscle strips changed under 470 nm blue light. Light stimulation evoked premature low-frequency and high amplitude (LFHA) contractions and enhanced the frequency of the LFHA contractions. The light-evoked contractions were blocked by T16Ainh-A01, an antagonist of anoctamin 1 channels that are expressed selectively in ICC in colonic muscles. CONCLUSIONS Our study demonstrates a potentially feasible approach to stimulate the activity of ICC by optogenetics. The colonic motor patterns of muscle strips, especially LFHA contractions, can be regulated by 470 nm light via ChR2, which is expressed in ICC.
Collapse
Affiliation(s)
- Song Zhao
- Division of Gastric and Colorectal Surgery, Department of General Surgery, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, China
| | - Weidong Tong
- Division of Gastric and Colorectal Surgery, Department of General Surgery, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, China
| |
Collapse
|
19
|
Shikaya Y, Inaba M, Tadokoro R, Utsunomiya S, Takahashi Y. Optogenetic control of gut movements reveals peristaltic wave-mediated induction of cloacal contractions and reactivation of impaired gut motility. Front Physiol 2023; 14:1175951. [PMID: 37293264 PMCID: PMC10245550 DOI: 10.3389/fphys.2023.1175951] [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: 02/28/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Gut peristalsis, recognized as a wave-like progression along the anterior-posterior gut axis, plays a pivotal role in the transportation, digestion, and absorption of ingested materials. The embryonic gut, which has not experienced ingested materials, undergoes peristalsis offering a powerful model for studying the intrinsic mechanisms underlying the gut motility. It has previously been shown in chicken embryos that acute contractions of the cloaca (an anus-like structure) located at the posterior end of the hindgut are tightly coupled with the arrival of hindgut-derived waves. To further scrutinize the interactions between hindgut and cloaca, we here developed an optogenetic method that produced artificial waves in the hindgut. A variant form of channelrhodopsin-2 (ChR2(D156C)), permitting extremely large photocurrents, was expressed in the muscle component of the hindgut of chicken embryos using Tol2-mediated gene transfer and in ovo electroporation techniques. The D156C-expressing hindgut responded efficiently to local pulses of blue light: local contractions emerge at an ectopic site in the hindgut, which were followed by peristaltic waves that reached to the endpoint of the hindgut. Markedly, the arrival of the optogenetically induced waves caused concomitant contractions of the cloaca, revealing that the hindgut-cloaca coordination is mediated by signals triggered by peristaltic waves. Moreover, a cloaca undergoing pharmacologically provoked aberrant contractions could respond to pulsed blue light irradiation. Together, the optogenetic technology developed in this study for inducing gut peristalsis paves the way to study the gut movement and also to explore therapeutic methodology for peristaltic disorders.
Collapse
|
20
|
Zhao S, Zhang T, Tong W. Application of optogenetics in the study of gastrointestinal motility: A mini review. JOURNAL OF INNOVATIVE OPTICAL HEALTH SCIENCES 2023; 16. [DOI: 10.1142/s1793545822300130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Disorders of gastrointestinal (GI) motility are associated with various symptoms such as nausea, vomiting, and constipation. However, the underlying causes of impaired GI motility remain unclear, which has led to variation in the efficacy of therapies to treat GI dysfunction. Optogenetics is a novel approach through which target cells can be precisely controlled by light and has shown great potential in GI motility research. Here, we summarized recent studies of GI motility patterns utilizing optogenetic devices and focused on the ability of opsins, which are genetically expressed in different types of cells in the gut, to regulate the excitability of target cells. We hope that our review of recent findings regarding optogenetic control of GI cells broadens the scope of application for optogenetics in GI motility studies.
Collapse
Affiliation(s)
- Song Zhao
- Department of General Surgery, Gastric and Colorectal Surgery Division, Army Medical Center (Daping Hospital), Army Medical University Chongqing, P. R. China
| | - Ting Zhang
- Department of General Surgery, The 983th Hospital of Joint Logistic Support Force of People’s Liberation Army, Tianjin, P. R. China
| | - Weidong Tong
- Department of General Surgery, Gastric and Colorectal Surgery Division, Army Medical Center (Daping Hospital), Army Medical University Chongqing, P. R. China
| |
Collapse
|
21
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
22
|
Zhang C, Pan C, Chan KF, Gao J, Yang Z, Leung KKC, Jin D, Wang Y, Xia N, Ning Z, Wang X, Jiang S, Zhang Z, Wang Q, Hao B, Chiu PWY, Zhang L. Wirelessly powered deformable electronic stent for noninvasive electrical stimulation of lower esophageal sphincter. SCIENCE ADVANCES 2023; 9:eade8622. [PMID: 36888700 PMCID: PMC9995080 DOI: 10.1126/sciadv.ade8622] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Electrical stimulation is a promising method to modulate gastrointestinal disorders. However, conventional stimulators need invasive implantation and removal surgeries associated with risks of infection and secondary injuries. Here, we report a battery-free and deformable electronic esophageal stent for wireless stimulation of the lower esophageal sphincter in a noninvasive fashion. The stent consists of an elastic receiver antenna infilled with liquid metal (eutectic gallium-indium), a superelastic nitinol stent skeleton, and a stretchable pulse generator that jointly enables 150% axial elongation and 50% radial compression for transoral delivery through the narrow esophagus. The compliant stent adaptive to the dynamic environment of the esophagus can wirelessly harvest energy through deep tissue. Continuous electrical stimulations delivered by the stent in vivo using pig models significantly increase the pressure of the lower esophageal sphincter. The electronic stent provides a noninvasive platform for bioelectronic therapies in the gastrointestinal tract without the need for open surgery.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Chengfeng Pan
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- The State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Kai Fung Chan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Multi-Scale Medical Robotics Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Jinyang Gao
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Zhengxin Yang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Kevin Kai Chung Leung
- Multi-Scale Medical Robotics Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Dongdong Jin
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Yuqiong Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Neng Xia
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Zhipeng Ning
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xin Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Shuai Jiang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Zifeng Zhang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Qinglong Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Bo Hao
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Philip Wai Yan Chiu
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Multi-Scale Medical Robotics Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Li Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Multi-Scale Medical Robotics Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
23
|
Zhou L, Zhang Y, Cao G, Zhang C, Zheng C, Meng G, Lai Y, Zhou Z, Liu Z, Liu Z, Guo F, Dong X, Liang Z, Wang Y, Guo S, Zhou X, Jiang H, Yu L. Wireless Self-Powered Optogenetic System for Long-Term Cardiac Neuromodulation to Improve Post-MI Cardiac Remodeling and Malignant Arrhythmia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205551. [PMID: 36698262 PMCID: PMC10037959 DOI: 10.1002/advs.202205551] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Autonomic imbalance is an important characteristic of patients after myocardial infarction (MI) and adversely contributes to post-MI cardiac remodeling and ventricular arrhythmias (VAs). A previous study proved that optogenetic modulation could precisely inhibit cardiac sympathetic hyperactivity and prevent acute ischemia-induced VAs. Here, a wireless self-powered optogenetic modulation system is introduced, which achieves long-term precise cardiac neuromodulation in ambulatory canines. The wireless self-powered optical system based on a triboelectric nanogenerator is powered by energy harvested from body motion and realized the effective optical illumination that is required for optogenetic neuromodulation (ON). It is further demonstrated that long-term ON significantly mitigates MI-induced sympathetic remodeling and hyperactivity, and improves a variety of clinically relevant outcomes such as improves ventricular dysfunction, reduces infarct size, increases electrophysiological stability, and reduces susceptibility to VAs. These novel insights suggest that wireless ON holds translational potential for the clinical treatment of arrhythmia and other cardiovascular diseases related to sympathetic hyperactivity. Moreover, this innovative self-powered optical system may provide an opportunity to develop implantable/wearable and self-controllable devices for long-term optogenetic therapy.
Collapse
Affiliation(s)
- Liping Zhou
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Yuanzheng Zhang
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
- Hubei Yangtze Memory LaboratoriesKey Laboratory of Artificial Micro, and Nano‐structures of Ministry of EducationSchool of Physics and TechnologyWuhan UniversityWuhan430072P. R. China
| | - Gang Cao
- Biomedical CenterCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430072P. R. China
| | - Chi Zhang
- Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430072P. R. China
| | - Chen Zheng
- Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430072P. R. China
| | - Guannan Meng
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Yanqiu Lai
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Zhen Zhou
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Zhihao Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Zihan Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Fuding Guo
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Xin Dong
- Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430072P. R. China
| | - Zhizhuo Liang
- Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430072P. R. China
| | - Yueyi Wang
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Shishang Guo
- Hubei Yangtze Memory LaboratoriesKey Laboratory of Artificial Micro, and Nano‐structures of Ministry of EducationSchool of Physics and TechnologyWuhan UniversityWuhan430072P. R. China
| | - Xiaoya Zhou
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Hong Jiang
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| | - Lilei Yu
- Department of CardiologyRenmin Hospital of Wuhan UniversityHubei Key Laboratory of Autonomic Nervous System ModulationCardiac Autonomic Nervous System Research Center of Wuhan UniversityTaikang Center for Life and Medical SciencesWuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhan430060P. R. China
| |
Collapse
|
24
|
Chen W, Liao L, Huang Z, Lu Y, Lin Y, Pei Y, Yi S, Huang C, Cao H, Tan B. Patchouli alcohol improved diarrhea-predominant irritable bowel syndrome by regulating excitatory neurotransmission in the myenteric plexus of rats. Front Pharmacol 2022; 13:943119. [PMID: 36452228 PMCID: PMC9703083 DOI: 10.3389/fphar.2022.943119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/31/2022] [Indexed: 09/07/2023] Open
Abstract
Background and Purpose: Irritable bowel syndrome (IBS) is usually associated with chronic gastrointestinal disorders. Its most common subtype is accompanied with diarrhea (IBS-D). The enteric nervous system (ENS) modulates major gastrointestinal motility and functions whose aberration may induce IBS-D. The enteric neurons are susceptible to long-term neurotransmitter level alterations. The patchouli alcohol (PA), extracted from Pogostemonis Herba, has been reported to regulate neurotransmitter release in the ENS, while its effectiveness against IBS-D and the underlying mechanism remain unknown. Experimental Approach: In this study, we established an IBS-D model in rats through chronic restraint stress. We administered the rats with 5, 10, and 20 mg/kg of PA for intestinal and visceral examinations. The longitudinal muscle myenteric plexus (LMMP) neurons were further immunohistochemically stained for quantitative, morphological, and neurotransmitters analyses. Key Results: We found that PA decreased visceral sensitivity, diarrhea symptoms and intestinal transit in the IBS-D rats. Meanwhile, 10 and 20 mg/kg of PA significantly reduced the proportion of excitatory LMMP neurons in the distal colon, decreased the number of acetylcholine (Ach)- and substance P (SP)-positive neurons in the distal colon and restored the levels of Ach and SP in the IBS-D rats. Conclusion and Implications: These findings indicated that PA modulated LMMP excitatory neuron activities, improved intestinal motility and alleviated IBS-induced diarrheal symptoms, suggesting the potential therapeutic efficacy of PA against IBS-D.
Collapse
Affiliation(s)
- Wanyu Chen
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Liao
- Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Guangzhou, China
| | - Zitong Huang
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yulin Lu
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukang Lin
- College of Integrated Chinese and Western Medicines, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Pei
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shulin Yi
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Huang
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Tan
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
25
|
Robinson CM, Short NE, Riglar DT. Achieving spatially precise diagnosis and therapy in the mammalian gut using synthetic microbial gene circuits. Front Bioeng Biotechnol 2022; 10:959441. [PMID: 36118573 PMCID: PMC9478464 DOI: 10.3389/fbioe.2022.959441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian gut and its microbiome form a temporally dynamic and spatially heterogeneous environment. The inaccessibility of the gut and the spatially restricted nature of many gut diseases translate into difficulties in diagnosis and therapy for which novel tools are needed. Engineered bacterial whole-cell biosensors and therapeutics have shown early promise at addressing these challenges. Natural and engineered sensing systems can be repurposed in synthetic genetic circuits to detect spatially specific biomarkers during health and disease. Heat, light, and magnetic signals can also activate gene circuit function with externally directed spatial precision. The resulting engineered bacteria can report on conditions in situ within the complex gut environment or produce biotherapeutics that specifically target host or microbiome activity. Here, we review the current approaches to engineering spatial precision for in vivo bacterial diagnostics and therapeutics using synthetic circuits, and the challenges and opportunities this technology presents.
Collapse
Affiliation(s)
| | | | - David T. Riglar
- Section of Structural and Synthetic Biology, Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Role of Ion Channels in the Chemotransduction and Mechanotransduction in Digestive Function and Feeding Behavior. Int J Mol Sci 2022; 23:ijms23169358. [PMID: 36012643 PMCID: PMC9409042 DOI: 10.3390/ijms23169358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The gastrointestinal tract constantly communicates with the environment, receiving and processing a wide range of information. The contents of the gastrointestinal tract and the gastrointestinal tract generate mechanical and chemical signals, which are essential for regulating digestive function and feeding behavior. There are many receptors here that sense intestinal contents, including nutrients, microbes, hormones, and small molecule compounds. In signal transduction, ion channels are indispensable as an essential component that can generate intracellular ionic changes or electrical signals. Ion channels generate electrical activity in numerous neurons and, more importantly, alter the action of non-neurons simply and effectively, and also affect satiety, molecular secretion, intestinal secretion, and motility through mechanisms of peripheral sensation, signaling, and altered cellular function. In this review, we focus on the identity of ion channels in chemosensing and mechanosensing in the gastrointestinal tract.
Collapse
|
27
|
Ricci MF, Béla SR, Barbosa JL, Moraes MM, Mazzeti AL, Bahia MT, Horta LS, Santiago HDC, Cruz JS, Capettini LDSA, Arantes RME. A Potential Role of Cholinergic Dysfunction on Impaired Colon Motility in Experimental Intestinal Chagas Disease. J Neurogastroenterol Motil 2022; 28:483-500. [PMID: 35799242 PMCID: PMC9274474 DOI: 10.5056/jnm21074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/21/2021] [Accepted: 10/11/2021] [Indexed: 12/01/2022] Open
Abstract
Background/Aims Chagasic megacolon is caused by Trypanosoma cruzi, which promotes in several cases, irreversible segmental colonic dilation. This alteration is the major anatomic-clinical disorder, characterized by the enteric nervous system and muscle wall structural damage. Herein, we investigate how T. cruzi-induced progressive colonic structural changes modulate the colonic contractile pattern activity. Methods We developed a murine model of T. cruzi-infection that reproduced long-term modifications of the enlarged colon. We evaluated colonic and total intestinal transit time in animals. The patterns of motor response at several time intervals between the acute and chronic phases were evaluated using the organ bath assays. Enteric motor neurons were stimulated by electric field stimulation. The responses were analyzed in the presence of the nicotinic and muscarinic acetylcholine receptor antagonists. Western blot was performed to evaluate the expression of nicotinic and muscarinic receptors. The neurotransmitter expression was analyzed by real-time polymerase chain reaction. Results In the chronic phase of infection, there was decreased intestinal motility associated with decreased amplitude and rhythmicity of intestinal contractility. Pharmacological tests suggested a defective response mediated by acetylcholine receptors. The contractile response induced by acetylcholine was decreased by atropine in the acute phase while the lack of its action in the chronic phase was associated with tissue damage, and decreased expression of choline acetyltransferase, nicotinic subunits of acetylcholine receptors, and neurotransmitters. Conclusions T. cruzi-induced damage of smooth muscles was accompanied by motility disorders such as decreased intestinal peristalsis and cholinergic system response impairment. This study allows integration of the natural history of Chagasic megacolon motility disorders and opens new perspectives for the design of effective therapeutic.
Collapse
Affiliation(s)
- Mayra F Ricci
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Samantha R Béla
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Joana L Barbosa
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Michele M Moraes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Ana L Mazzeti
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brasil
| | - Maria T Bahia
- Escola de Medicina & Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Laila S Horta
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Helton da C Santiago
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Jader S Cruz
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Luciano dos S A Capettini
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Rosa M E Arantes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| |
Collapse
|
28
|
Hee Lee J, Lee S, Kim D, Jae Lee K. Implantable Micro-Light-Emitting Diode (µLED)-based optogenetic interfaces toward human applications. Adv Drug Deliv Rev 2022; 187:114399. [PMID: 35716898 DOI: 10.1016/j.addr.2022.114399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/29/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022]
Abstract
Optogenetics has received wide attention in biomedical fields because of itsadvantages in temporal precision and spatial resolution. Beyond contributions to important advances in fundamental research, optogenetics is inspiring a shift towards new methods of improving human well-being and treating diseases. Soft, flexible and biocompatible systems using µLEDs as a light source have been introduced to realize brain-compatible optogenetic implants, but there are still many technical challenges to overcome before their human applications. In this review, we address progress in the development of implantable µLED probes and recent achievements in (i) device engineering design, (ii) driving power, (iii) multifunctionality and (iv) closed-loop systems. (v) Expanded optogenetic applications based on remarkable advances in µLED implants will also be discussed.
Collapse
Affiliation(s)
- Jae Hee Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sinjeong Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Keon Jae Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
29
|
Gonzales J, Gulbransen BD. Purines help determine the gut's sweet tooth. Purinergic Signal 2022; 18:245-247. [PMID: 35639305 DOI: 10.1007/s11302-022-09871-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jacques Gonzales
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| | - Brian D Gulbransen
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
30
|
Drumm BT, Cobine CA, Baker SA. Insights on gastrointestinal motility through the use of optogenetic sensors and actuators. J Physiol 2022; 600:3031-3052. [PMID: 35596741 DOI: 10.1113/jp281930] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 11/08/2022] Open
Abstract
The muscularis of the gastrointestinal (GI) tract consists of smooth muscle cells (SMCs) and various populations of interstitial cells of Cajal (ICC), platelet-derived growth factor receptor α+ (PDGFRα+ ) cells, as well as excitatory and inhibitory enteric motor nerves. SMCs, ICC and PDGFRα+ cells form an electrically coupled syncytium, which together with inputs from the enteric nervous system (ENS) regulate GI motility. Early studies evaluating Ca2+ signalling behaviours in the GI tract relied upon indiscriminate loading of tissues with Ca2+ dyes. These methods lacked the means to study activity in specific cells of interest without encountering contamination from other cells within the preparation. Development of mice expressing optogenetic sensors (GCaMP, RCaMP) has allowed visualization of Ca2+ signalling behaviours in a cell specific manner. Additionally, availability of mice expressing optogenetic modulators (channelrhodopsins or halorhodospins) has allowed manipulation of specific signalling pathways using light. GCaMP expressing animals have been used to characterize Ca2+ signalling behaviours of distinct classes of ICC and SMCs throughout the GI musculature. These findings illustrate how Ca2+ signalling in ICC is fundamental in GI muscles, contributing to tone in sphincters, pacemaker activity in rhythmic muscles and relaying enteric signals to SMCs. Animals that express channelrhodopsin in specific neuronal populations have been used to map neural circuitry and to examine post junctional neural effects on GI motility. Thus, optogenetic approaches provide a novel means to examine the contribution of specific cell types to the regulation of motility patterns within complex multi-cellular systems. Abstract Figure Legends Optogenetic activators and sensors can be used to investigate the complex multi-cellular nature of the gastrointestinal (GI tract). Optogenetic activators that are activated by light such as channelrhodopsins (ChR2), OptoXR and halorhodopsinss (HR) proteins can be genetically encoded into specific cell types. This can be used to directly activate or silence specific GI cells such as various classes of enteric neurons, smooth muscle cells (SMC) or interstitial cells, such as interstitial cells of Cajal (ICC). Optogenetic sensors that are activated by different wavelengths of light such as green calmodulin fusion protein (GCaMP) and red CaMP (RCaMP) make high resolution of sub-cellular Ca2+ signalling possible within intact tissues of specific cell types. These tools can provide unparalleled insight into mechanisms underlying GI motility and innervation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland.,Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Caroline A Cobine
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Salah A Baker
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
31
|
Benevides ES, Sunshine MD, Rana S, Fuller DD. Optogenetic activation of the diaphragm. Sci Rep 2022; 12:6503. [PMID: 35444167 PMCID: PMC9021282 DOI: 10.1038/s41598-022-10240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Impaired diaphragm activation is common in many neuromuscular diseases. We hypothesized that expressing photoreceptors in diaphragm myofibers would enable light stimulation to evoke functional diaphragm activity, similar to endogenous bursts. In a mouse model, adeno-associated virus (AAV) encoding channelrhodopsin-2 (AAV9-CAG-ChR2-mVenus, 6.12 × 1011 vg dose) was delivered to the diaphragm using a minimally invasive method of microinjection to the intrapleural space. At 8-18 weeks following AAV injection, mice were anesthetized and studied during spontaneous breathing. We first showed that diaphragm electromyographic (EMG) potentials could be evoked with brief presentations of light, using a 473 nm high intensity LED. Evoked potential amplitude increased with intensity or duration of the light pulse. We next showed that in a paralyzed diaphragm, trains of light pulses evoked diaphragm EMG activity which resembled endogenous bursting, and this was sufficient to generate respiratory airflow. Light-evoked diaphragm EMG bursts showed no diminution after up to one hour of stimulation. Histological evaluation confirmed transgene expression in diaphragm myofibers. We conclude that intrapleural delivery of AAV9 can drive expression of ChR2 in the diaphragm and subsequent photostimulation can evoke graded compound diaphragm EMG activity similar to endogenous inspiratory bursting.
Collapse
Affiliation(s)
- Ethan S Benevides
- Rehabilitation Science PhD Program, University of Florida, Gainesville, Florida, USA.,Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Michael D Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA. .,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA. .,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
32
|
Yang Y, Wu M, Wegener AJ, Vázquez-Guardado A, Efimov AI, Lie F, Wang T, Ma Y, Banks A, Li Z, Xie Z, Huang Y, Good CH, Kozorovitskiy Y, Rogers JA. Preparation and use of wireless reprogrammable multilateral optogenetic devices for behavioral neuroscience. Nat Protoc 2022; 17:1073-1096. [PMID: 35173306 PMCID: PMC9311268 DOI: 10.1038/s41596-021-00672-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/01/2021] [Indexed: 11/08/2022]
Abstract
Wireless battery-free optogenetic devices enable behavioral neuroscience studies in groups of animals with minimal interference to natural behavior. Real-time independent control of optogenetic stimulation through near-field communication dramatically expands the realm of applications of these devices in broad contexts of neuroscience research. Dissemination of these tools with advanced functionalities to the neuroscience community requires protocols for device manufacturing and experimental implementation. This protocol describes detailed procedures for fabrication, encapsulation and implantation of recently developed advanced wireless devices in head- and back-mounted forms. In addition, procedures for standard implementation of experimental systems in mice are provided. This protocol aims to facilitate the application of wireless optogenetic devices in advanced optogenetic experiments involving groups of freely moving rodents and complex environmental designs. The entire protocol lasts ~3-5 weeks.
Collapse
Affiliation(s)
- Yiyuan Yang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Amy J Wegener
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Abraham Vázquez-Guardado
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Andrew I Efimov
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | | | - Taoyi Wang
- Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Yuhang Ma
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Anthony Banks
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Neurolux Inc., Evanston, IL, USA
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Zhengwei Li
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Zhaoqian Xie
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
| | - Yonggang Huang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Cameron H Good
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA.
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA.
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA.
| | - John A Rogers
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Neurolux Inc., Evanston, IL, USA.
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Northwestern University, Evanston, IL, USA.
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA.
- Department of Computer Science, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
33
|
Hong S, Kim WS, Han Y, Cherukuri R, Jung H, Campos C, Wu Q, Park SI. Optogenetic Targeting of Mouse Vagal Afferents Using an Organ-specific, Scalable, Wireless Optoelectronic Device. Bio Protoc 2022; 12:e4341. [PMID: 35592610 PMCID: PMC8918220 DOI: 10.21769/bioprotoc.4341] [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: 01/06/2022] [Revised: 08/19/2021] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
Optogenetics has the potential to transform the study of the peripheral nervous system (PNS), but the complex anatomy of the PNS poses unique challenges for the focused delivery of light to specific tissues. This protocol describes the fabrication of a wireless telemetry system for studying peripheral sensory pathways. Unlike existing wireless approaches, the low-power wireless telemetry offers organ specificity via a sandwiched pre-curved tether, and enables high-throughput analysis of behavioral experiments with a channel isolation strategy. We describe the technical procedures for the construction of these devices, the wireless power transmission (TX) system with antenna coils, and their implementation for in vivo experimental applications. In total, the timeline of the procedure, including device fabrication, implantation, and preparation to begin in vivo experimentation can be completed in ~2-4 weeks. Implementation of these devices allows for chronic (>1 month) wireless optogenetic manipulation of peripheral neural pathways in freely behaving animals navigating homecage environments (up to 8).
Collapse
Affiliation(s)
- Sungcheol Hong
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Woo Seok Kim
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Yong Han
- Department of Pediatrics, Baylor College of Medicine, Houston, US
| | - Rahul Cherukuri
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Haemin Jung
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Carlos Campos
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, US
| | - Qi Wu
- Department of Pediatrics, Baylor College of Medicine, Houston, US
| | - Sung Il Park
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
,Institute for Neuroscience, Texas A&M University, College Station, US
,Center of Remote Health Sciences and Technologies, Texas A&M University, College Station, US
,
*For correspondence:
| |
Collapse
|
34
|
Feng J, Hibberd TJ, Luo J, Yang P, Xie Z, Travis L, Spencer NJ, Hu H. Modification of Neurogenic Colonic Motor Behaviours by Chemogenetic Ablation of Calretinin Neurons. Front Cell Neurosci 2022; 16:799717. [PMID: 35317196 PMCID: PMC8934436 DOI: 10.3389/fncel.2022.799717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/31/2022] [Indexed: 12/31/2022] Open
Abstract
How the enteric nervous system determines the pacing and propagation direction of neurogenic contractions along the colon remains largely unknown. We used a chemogenetic strategy to ablate enteric neurons expressing calretinin (CAL). Mice expressing human diphtheria toxin receptor (DTR) in CAL neurons were generated by crossing CAL-ires-Cre mice with Cre-dependent ROSA26-DTR mice. Immunohistochemical analysis revealed treatment with diphtheria toxin incurred a 42% reduction in counts of Hu-expressing colonic myenteric neurons (P = 0.036), and 57% loss of CAL neurons (comprising ∼25% of all Hu neurons; P = 0.004) compared to control. As proportions of Hu-expressing neurons, CAL neurons that contained nitric oxide synthase (NOS) were relatively spared (control: 15 ± 2%, CAL-DTR: 13 ± 1%; P = 0.145), while calretinin neurons lacking NOS were significantly reduced (control: 26 ± 2%, CAL-DTR: 18 ± 5%; P = 0.010). Colonic length and pellet sizes were significantly reduced without overt inflammation or changes in ganglionic density. Interestingly, colonic motor complexes (CMCs) persisted with increased frequency (mid-colon interval 111 ± 19 vs. 189 ± 24 s, CAL-DTR vs. control, respectively, P < 0.001), decreased contraction size (mid-colon AUC 26 ± 24 vs. 59 ± 13 gram/seconds, CAL-DTR vs. control, respectively, P < 0.001), and lacked preferential anterograde migration (P < 0.001). The functional effects of modest calretinin neuron ablation, particularly increased neurogenic motor activity frequencies, differ from models that incur general enteric neuron loss, and suggest calretinin neurons may contribute to pacing, force, and polarity of CMCs in the large bowel.
Collapse
Affiliation(s)
- Jing Feng
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tim J. Hibberd
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Jialie Luo
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Pu Yang
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Zili Xie
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Lee Travis
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Nick J. Spencer
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
- *Correspondence: Nick J. Spencer,
| | - Hongzhen Hu
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
- Hongzhen Hu,
| |
Collapse
|
35
|
Schonkeren SL, Küthe TT, Idris M, Bon-Frauches AC, Boesmans W, Melotte V. The gut brain in a dish: Murine primary enteric nervous system cell cultures. Neurogastroenterol Motil 2022; 34:e14215. [PMID: 34236124 PMCID: PMC9285479 DOI: 10.1111/nmo.14215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/22/2021] [Accepted: 06/01/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND The enteric nervous system (ENS) is an extensive neural network embedded in the wall of the gastrointestinal tract that regulates digestive function and gastrointestinal homeostasis. The ENS consists of two main cell types; enteric neurons and enteric glial cells. In vitro techniques allow simplified investigation of ENS function, and different culture methods have been developed over the years helping to understand the role of ENS cells in health and disease. PURPOSE This review focuses on summarizing and comparing available culture protocols for the generation of primary ENS cells from adult mice, including dissection of intestinal segments, enzymatic digestions, surface coatings, and culture media. In addition, the potential of human ENS cultures is also discussed.
Collapse
Affiliation(s)
- Simone L Schonkeren
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Tara T Küthe
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Musa Idris
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ana C Bon-Frauches
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Werend Boesmans
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Veerle Melotte
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
36
|
Buchanan KL, Rupprecht LE, Kaelberer MM, Sahasrabudhe A, Klein ME, Villalobos JA, Liu WW, Yang A, Gelman J, Park S, Anikeeva P, Bohórquez DV. The preference for sugar over sweetener depends on a gut sensor cell. Nat Neurosci 2022; 25:191-200. [PMID: 35027761 PMCID: PMC8825280 DOI: 10.1038/s41593-021-00982-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022]
Abstract
Guided by gut sensory cues, humans and animals prefer nutritive sugars over non-caloric sweeteners, but how the gut steers such preferences remains unknown. In the intestine, neuropod cells synapse with vagal neurons to convey sugar stimuli to the brain within seconds. Here, we found that cholecystokinin (CCK)-labeled duodenal neuropod cells differentiate and transduce luminal stimuli from sweeteners and sugars to the vagus nerve using sweet taste receptors and sodium glucose transporters. The two stimulus types elicited distinct neural pathways: while sweetener stimulated purinergic neurotransmission, sugar stimulated glutamatergic neurotransmission. To probe the contribution of these cells to behavior, we developed optogenetics for the gut lumen by engineering a flexible fiberoptic. We showed that preference for sugar over sweetener in mice depends on neuropod cell glutamatergic signaling. By swiftly discerning the precise identity of nutrient stimuli, gut neuropod cells serve as the entry point to guide nutritive choices. Buchanan, Rupprecht, Kaelberer and colleagues show that the preference for sugar over sweetener in mice depends on gut neuropod cells. Akin to other sensor cells, neuropod cells swiftly communicate the precise identity of stimuli to drive food choices.
Collapse
Affiliation(s)
- Kelly L Buchanan
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Duke University School of Medicine, Durham, NC, USA
| | - Laura E Rupprecht
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Department of Medicine, Duke University, Durham, NC, USA
| | - M Maya Kaelberer
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Department of Medicine, Duke University, Durham, NC, USA
| | - Atharva Sahasrabudhe
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marguerita E Klein
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Department of Medicine, Duke University, Durham, NC, USA
| | - Jorge A Villalobos
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Department of Medicine, Duke University, Durham, NC, USA
| | - Winston W Liu
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Duke University School of Medicine, Durham, NC, USA.,Department of Neurobiology, Duke University, Durham, NC, USA
| | - Annabelle Yang
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Trinity College of Arts & Sciences, Duke University, Durham, NC, USA
| | - Justin Gelman
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.,Trinity College of Arts & Sciences, Duke University, Durham, NC, USA
| | - Seongjun Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Polina Anikeeva
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.,Departments of Materials Science & Engineering and Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA. .,Department of Medicine, Duke University, Durham, NC, USA. .,Department of Neurobiology, Duke University, Durham, NC, USA. .,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA. .,MSRB-I, room 221A, 203 Research Drive, Durham, NC, USA.
| |
Collapse
|
37
|
Meerschaert KA, Davis BM, Smith-Edwards KM. New Insights on Extrinsic Innervation of the Enteric Nervous System and Non-neuronal Cell Types That Influence Colon Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:133-139. [PMID: 36587153 DOI: 10.1007/978-3-031-05843-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The enteric nervous system not only innervates the colon to execute various functions in a semi-autonomous manner but also receives neural input from three extrinsic sources, (1) vagal, (2) thoracolumbar (splanchnic), and (3) lumbosacral (pelvic) pathways, that permit bidirectional communication between the colon and central nervous system. Extrinsic pathways signal sensory input via afferent fibers, as well as motor autonomic output via parasympathetic or sympathetic efferent fibers, but the shared and unique roles for each pathway in executing sensory-motor control of colon function have not been well understood. Here, we describe the recently developed approaches that have provided new insights into the diverse mechanisms utilized by extrinsic pathways to influence colon functions related to visceral sensation, motility, and inflammation. Based on the cumulative results from anatomical, molecular, and functional studies, we propose pathway-specific functions for vagal, thoracolumbar, and lumbosacral innervation of the colon.
Collapse
Affiliation(s)
| | - Brian M Davis
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
38
|
Barth BB, Spencer NJ, Grill WM. Activation of ENS Circuits in Mouse Colon: Coordination in the Mouse Colonic Motor Complex as a Robust, Distributed Control System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:113-123. [PMID: 36587151 DOI: 10.1007/978-3-031-05843-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The characteristic motor patterns of the colon are coordinated by the enteric nervous system (ENS) and involve enterochromaffin (EC) cells, enteric glia, smooth muscle fibers, and interstitial cells. While the fundamental control mechanisms of colonic motor patterns are understood, greater complexity in the circuitry underlying motor patterns has been revealed by recent advances in the field. We review these recent advances and new findings from our laboratories that provide insights into how the ENS coordinates motor patterns in the isolated mouse colon. We contextualize these observations by describing the neuromuscular system underling the colonic motor complex (CMC) as a robust, distributed control system. Framing the colonic motor complex as a control system reveals a new perspective on the coordinated motor patterns in the colon. We test the control system by applying electrical stimulation in the isolated mouse colon to disrupt the coordination and propagation of the colonic motor complex.
Collapse
Affiliation(s)
- Bradley B Barth
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
39
|
Prospero AG, Pinto LA, Matos RVR, Soares GA, Oliveira RB, Mascarenhas S, Miranda JRDA. New device for active gastric mechanical stimulation. Neurogastroenterol Motil 2021; 33:e14169. [PMID: 33969918 DOI: 10.1111/nmo.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 04/16/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastroparesis is a chronic stomach disorder and effective treatment is the aim of different strategies. Alternative therapies consist of an electrical stimulation of the stomach to evoke a response in the gastric activity. We present the development and in vivo application of an electromagnet system to induce a mechanical stimulus in the stomach aiming for gastric contractile responses. METHODS The electromagnet system consisted of an implantable magnet and an external drive coil. We implanted the magnet at the greater curvature of the gastric body in rats. We applied an alternating current to the drive coils, inducing mechanical stimulation of the gastric wall. We measured the gastric contraction activity and gastric electrical activity in response to the stimulus using AC biosusceptometry and electrogastrography. Moreover, we used the phenol red to evaluate the stimulus effects on gastrointestinal transit. KEY RESULTS The stimulus increased the spectral intensity and signal-to-noise ratio significantly of gastric contraction activity and gastric electrical activity. Furthermore, we found a lower phenol red retention in the stomach in rats without stimulus. No significant differences were found in frequency and root mean square amplitude. CONCLUSIONS & INFERENCES We developed a new simple electromagnet system that evoked a contraction and gastric electrical response using a mechanical stimulus and decreased gastric emptying time. The system is an accessible tool and may contribute to gastroparesis studies in animals.
Collapse
Affiliation(s)
- Andre Gonçalves Prospero
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Leonardo Antonio Pinto
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Ronaldo Vitor Reis Matos
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Guilherme Augusto Soares
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| | - Ricardo Brandt Oliveira
- Faculdade de Medicina de Ribeirão Preto, USP/University of São Paulo, Ribeirão Preto, Brazil
| | | | - José Ricardo de Arruda Miranda
- Departamento de Biofísica e Farmacologia, Laboratório de Biomagnetismo, UNESP/São Paulo State University, Botucatu, Brazil
| |
Collapse
|
40
|
Nestor-Kalinoski A, Smith-Edwards KM, Meerschaert K, Margiotta JF, Rajwa B, Davis BM, Howard MJ. Unique Neural Circuit Connectivity of Mouse Proximal, Middle, and Distal Colon Defines Regional Colonic Motor Patterns. Cell Mol Gastroenterol Hepatol 2021; 13:309-337.e3. [PMID: 34509687 PMCID: PMC8703201 DOI: 10.1016/j.jcmgh.2021.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Colonic motor patterns have been described by a number of different groups, but the neural connectivity and ganglion architecture supporting patterned motor activity have not been elucidated. Our goals were to describe quantitatively, by region, the structural architecture of the mouse enteric nervous system and use functional calcium imaging, pharmacology, and electrical stimulation to show regional underpinnings of different motor patterns. METHODS Excised colon segments from mice expressing the calcium indicator GCaMP6f or GCaMP6s were used to examine spontaneous and evoked (pharmacologic or electrical) changes in GCaMP-mediated fluorescence and coupled with assessment of colonic motor activity, immunohistochemistry, and confocal imaging. Three-dimensional image reconstruction and statistical methods were used to describe quantitatively mouse colon myenteric ganglion structure, neural and vascular network patterning, and neural connectivity. RESULTS In intact colon, regionally specific myenteric ganglion size, architecture, and neural circuit connectivity patterns along with neurotransmitter-receptor expression underlie colonic motor patterns that define functional differences along the colon. Region-specific effects on spontaneous, evoked, and chemically induced neural activity contribute to regional motor patterns, as does intraganglionic functional connectivity. We provide direct evidence of neural circuit structural and functional regional differences that have only been inferred in previous investigations. We include regional comparisons between quantitative measures in mouse and human colon that represent an important advance in showing the usefulness and relevance of the mouse system for translation to the human colon. CONCLUSIONS There are several neural mechanisms dependent on myenteric ganglion architecture and functional connectivity that underlie neurogenic control of patterned motor function in the mouse colon.
Collapse
Affiliation(s)
- Andrea Nestor-Kalinoski
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Kristen M Smith-Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kimberly Meerschaert
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joseph F Margiotta
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana
| | - Brian M Davis
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marthe J Howard
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.
| |
Collapse
|
41
|
Keshmiri Neghab H, Soheilifar MH, Grusch M, Ortega MM, Esmaeeli Djavid G, Saboury AA, Goliaei B. The state of the art of biomedical applications of optogenetics. Lasers Surg Med 2021; 54:202-216. [PMID: 34363230 DOI: 10.1002/lsm.23463] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Optogenetics has opened new insights into biomedical research with the ability to manipulate and control cellular activity using light in combination with genetically engineered photosensitive proteins. By stimulating with light, this method provides high spatiotemporal and high specificity resolution, which is in contrast to conventional pharmacological or electrical stimulation. Optogenetics was initially introduced to control neural activities but was gradually extended to other biomedical fields. STUDY DESIGN In this paper, firstly, we summarize the current optogenetic tools stimulated by different light sources, including lasers, light-emitting diodes, and laser diodes. Second, we outline the variety of biomedical applications of optogenetics not only for neuronal circuits but also for various kinds of cells and tissues from cardiomyocytes to ganglion cells. Furthermore, we highlight the potential of this technique for treating neurological disorders, cardiac arrhythmia, visual impairment, hearing loss, and urinary bladder diseases as well as clarify the mechanisms underlying cancer progression and control of stem cell differentiation. CONCLUSION We sought to summarize the various types of promising applications of optogenetics to treat a broad spectrum of disorders. It is conceivable to expect that optogenetics profits a growing number of patients suffering from a range of different diseases in the near future.
Collapse
Affiliation(s)
- Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | | | - Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Ali Akbar Saboury
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Bahram Goliaei
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
42
|
Wood JD. Motor behavior of mouse large intestine: A Minireview. Neurogastroenterol Motil 2021; 33:e14206. [PMID: 34145934 DOI: 10.1111/nmo.14206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023]
Abstract
Mice with a recessive gene which reduces the number of ganglion cells of the distal colon and rectum and produces megacolon, imitating Hirschsprung disease, are discussed as a model for integrative control of the large intestinal smooth musculature by the enteric division of the autonomic nervous system (ie, the brain-in-the-gut). Investigative approaches, such as propulsion of artificial pellets in preparations of whole colon in organ baths in vitro and innovative approaches capitalizing on neurogenetic technologies (eg, optogenetics), are considered in view of potential application in the development of novel therapeutic mechanisms to selectively evoke and control gastrointestinal motility patterns, such as the small intestinal digestive motility pattern, interdigestive pattern, and reversed direction of powerful propulsive motility during emesis. This minireview relates to the paper titled: "Motor patterns in the proximal and distal mouse colon which underlie formation and propulsion of feces," appearing in this issue of Neurogastroenterology and Motility.
Collapse
Affiliation(s)
- Jackie Dale Wood
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
43
|
Patrono E, Svoboda J, Stuchlík A. Schizophrenia, the gut microbiota, and new opportunities from optogenetic manipulations of the gut-brain axis. Behav Brain Funct 2021; 17:7. [PMID: 34158061 PMCID: PMC8218443 DOI: 10.1186/s12993-021-00180-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia research arose in the twentieth century and is currently rapidly developing, focusing on many parallel research pathways and evaluating various concepts of disease etiology. Today, we have relatively good knowledge about the generation of positive and negative symptoms in patients with schizophrenia. However, the neural basis and pathophysiology of schizophrenia, especially cognitive symptoms, are still poorly understood. Finding new methods to uncover the physiological basis of the mental inabilities related to schizophrenia is an urgent task for modern neuroscience because of the lack of specific therapies for cognitive deficits in the disease. Researchers have begun investigating functional crosstalk between NMDARs and GABAergic neurons associated with schizophrenia at different resolutions. In another direction, the gut microbiota is getting increasing interest from neuroscientists. Recent findings have highlighted the role of a gut-brain axis, with the gut microbiota playing a crucial role in several psychopathologies, including schizophrenia and autism. There have also been investigations into potential therapies aimed at normalizing altered microbiota signaling to the enteric nervous system (ENS) and the central nervous system (CNS). Probiotics diets and fecal microbiota transplantation (FMT) are currently the most common therapies. Interestingly, in rodent models of binge feeding, optogenetic applications have been shown to affect gut colony sensitivity, thus increasing colonic transit. Here, we review recent findings on the gut microbiota–schizophrenia relationship using in vivo optogenetics. Moreover, we evaluate if manipulating actors in either the brain or the gut might improve potential treatment research. Such research and techniques will increase our knowledge of how the gut microbiota can manipulate GABA production, and therefore accompany changes in CNS GABAergic activity.
Collapse
Affiliation(s)
- Enrico Patrono
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| | - Jan Svoboda
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic
| | - Aleš Stuchlík
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| |
Collapse
|
44
|
Pandelakis M, Delgado E, Ebrahimkhani MR. CRISPR-Based Synthetic Transcription Factors In Vivo: The Future of Therapeutic Cellular Programming. Cell Syst 2021; 10:1-14. [PMID: 31972154 DOI: 10.1016/j.cels.2019.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/14/2019] [Accepted: 10/09/2019] [Indexed: 01/04/2023]
Abstract
Pinpoint control over endogenous gene expression in vivo has long been a fevered dream for clinicians and researchers alike. With the recent repurposing of programmable, RNA-guided DNA endonucleases from the CRISPR bacterial immune system, this dream is becoming a powerful reality. Engineered CRISPR/Cas9-based transcriptional regulators and epigenome editors have enabled researchers to perturb endogenous gene expression in vivo, allowing for the therapeutic reprogramming of cell and tissue behavior. For this technology to be of maximal use, a variety of technological hurdles still need to be addressed. Better understanding of the design principle controlling gene expression together with technologies that enable spatiotemporal control of transcriptional engineering are fundamental for rational design, improved efficacy, and ultimately safe translation to humans. In this review, we will discuss recent advances and integrative strategies that can help pave the path toward a new class of transcriptional therapeutics.
Collapse
Affiliation(s)
- Matthew Pandelakis
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, USA
| | - Elizabeth Delgado
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, USA
| | - Mo R Ebrahimkhani
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, USA; Department of Pathology, Division of Experimental Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
45
|
Abstract
The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.
Collapse
|
46
|
Barth BB, Travis L, Spencer NJ, Grill WM. Control of colonic motility using electrical stimulation to modulate enteric neural activity. Am J Physiol Gastrointest Liver Physiol 2021; 320:G675-G687. [PMID: 33624530 PMCID: PMC8238160 DOI: 10.1152/ajpgi.00463.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 01/31/2023]
Abstract
Electrical stimulation of the enteric nervous system (ENS) is an attractive approach to modify gastrointestinal transit. Colonic motor complexes (CMCs) occur with a periodic rhythm, but the ability to elicit a premature CMC depends, at least in part, upon the intrinsic refractory properties of the ENS, which are presently unknown. The objectives of this study were to record myoelectric complexes (MCs, the electrical correlates of CMCs) in the smooth muscle and 1) determine the refractory periods of MCs, 2) inform and evaluate closed-loop stimulation to repetitively evoke MCs, and 3) identify stimulation methods to suppress MC propagation. We dissected the colon from male and female C57BL/6 mice, preserving the integrity of intrinsic circuitry while removing the extrinsic nerves, and measured properties of spontaneous and evoked MCs in vitro. Hexamethonium abolished spontaneous and evoked MCs, confirming the necessary involvement of the ENS for electrically evoked MCs. Electrical stimulation reduced the mean interval between evoked and spontaneous CMCs (24.6 ± 3.5 vs. 70.6 ± 15.7 s, P = 0.0002, n = 7). The absolute refractory period was 4.3 s (95% confidence interval (CI) = 2.8-5.7 s, R2 = 0.7315, n = 8). Electrical stimulation applied during fluid distention-evoked MCs led to an arrest of MC propagation, and following stimulation, MC propagation resumed at an increased velocity (n = 9). The timing parameters of electrical stimulation increased the rate of evoked MCs and the duration of entrainment of MCs, and the refractory period provides insight into timing considerations for designing neuromodulation strategies to treat colonic dysmotility.NEW & NOTEWORTHY Maintained physiological distension of the isolated mouse colon induces rhythmic cyclic myoelectric complexes (MCs). MCs evoked repeatedly by closed-loop electrical stimulation entrain MCs more frequently than spontaneously occurring MCs. Electrical stimulation delivered at the onset of a contraction temporarily suppresses the propagation of MC contractions. Controlled electrical stimulation can either evoke MCs or temporarily delay MCs in the isolated mouse colon, depending on timing relative to ongoing activity.
Collapse
Affiliation(s)
- Bradley B Barth
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Lee Travis
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina
- Department of Neurobiology, Duke University, Durham, North Carolina
- Department of Neurosurgery, Duke University, Durham, North Carolina
| |
Collapse
|
47
|
Fujii K, Nakajo K, Egashira Y, Yamamoto Y, Kitada K, Taniguchi K, Kawai M, Tomiyama H, Kawakami K, Uchiyama K, Ono F. Gastrointestinal Neurons Expressing HCN4 Regulate Retrograde Peristalsis. Cell Rep 2021; 30:2879-2888.e3. [PMID: 32130893 DOI: 10.1016/j.celrep.2020.02.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/31/2022] Open
Abstract
Peristalsis is indispensable for physiological function of the gut. The enteric nervous system (ENS) plays an important role in regulating peristalsis. While the neural network regulating anterograde peristalsis, which migrates from the oral end to the anal end, is characterized to some extent, retrograde peristalsis remains unresolved with regards to its neural regulation. Using forward genetics in zebrafish, we reveal that a population of neurons expressing a hyperpolarization-activated nucleotide-gated channel HCN4 specifically regulates retrograde peristalsis. When HCN4 channels are blocked by an HCN channel inhibitor or morpholinos blocking the protein expression, retrograde peristalsis is specifically attenuated. Conversely, when HCN4(+) neurons expressing channelrhodopsin are activated by illumination, retrograde peristalsis is enhanced while anterograde peristalsis remains unchanged. We propose that HCN4(+) neurons in the ENS forward activating signals toward the oral end and simultaneously stimulate local circuits regulating the circular muscle.
Collapse
Affiliation(s)
- Kensuke Fujii
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Nakajo
- Department of Physiology, Osaka Medical College, Takatsuki, Japan; Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | | | | | - Kazuya Kitada
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Masaru Kawai
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Hideki Tomiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan
| | - Kazuhisa Uchiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Fumihito Ono
- Department of Physiology, Osaka Medical College, Takatsuki, Japan.
| |
Collapse
|
48
|
Vogt M, Schulz B, Wagdi A, Lebert J, van Belle GJ, Christoph J, Bruegmann T, Patejdl R. Direct optogenetic stimulation of smooth muscle cells to control gastric contractility. Am J Cancer Res 2021; 11:5569-5584. [PMID: 33859764 PMCID: PMC8039938 DOI: 10.7150/thno.53883] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
Rationale: Antral peristalsis is responsible for gastric emptying. Its failure is called gastroparesis and often caused by dysfunction of enteric neurons and interstitial cells of Cajal (ICC). Current treatment options, including gastric electrical stimulation, are non-satisfying and may improve symptoms but commonly fail to restore gastric emptying. Herein, we explore direct optogenetic stimulation of smooth muscle cells (SMC) via the light-gated non-selective cation channel Channelrhodopsin2 (ChR2) to control gastric motor function. Methods: We used a transgenic mouse model expressing ChR2 in fusion with eYFP under the control of the chicken-β-actin promoter. We performed patch clamp experiments to quantify light-induced currents in isolated SMC, Ca2+ imaging and isometric force measurements of antral smooth muscle strips as well as pressure recordings of intact stomachs to evaluate contractile responses. Light-induced propulsion of gastric contents from the isolated stomach preparation was quantified in video recordings. We furthermore tested optogenetic stimulation in a gastroparesis model induced by neuronal- and ICC-specific damage through methylene blue photo-toxicity. Results: In the stomachs, eYFP signals were restricted to SMC in which blue light (460 nm) induced inward currents typical for ChR2. These depolarizing currents led to contractions in antral smooth muscle strips that were stronger than those triggered by supramaximal electrical field stimulation and comparable to those evoked by global depolarization with high K+ concentration. In the intact stomach, panoramic illumination efficiently increased intragastric pressure achieving 239±46% (n=6) of the pressure induced by electrical field stimulation and triggered gastric transport. Within the gastroparesis model, electric field stimulation completely failed but light still efficiently generated pressure waves. Conclusions: We demonstrate direct optogenetic stimulation of SMC to control gastric contractility. This completely new approach could allow for the restoration of motility in gastroparesis in the future.
Collapse
|
49
|
Organ-specific, multimodal, wireless optoelectronics for high-throughput phenotyping of peripheral neural pathways. Nat Commun 2021; 12:157. [PMID: 33420038 PMCID: PMC7794361 DOI: 10.1038/s41467-020-20421-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/12/2020] [Indexed: 11/17/2022] Open
Abstract
The vagus nerve supports diverse autonomic functions and behaviors important for health and survival. To understand how specific components of the vagus contribute to behaviors and long-term physiological effects, it is critical to modulate their activity with anatomical specificity in awake, freely behaving conditions using reliable methods. Here, we introduce an organ-specific scalable, multimodal, wireless optoelectronic device for precise and chronic optogenetic manipulations in vivo. When combined with an advanced, coil-antenna system and a multiplexing strategy for powering 8 individual homecages using a single RF transmitter, the proposed wireless telemetry enables low cost, high-throughput, and precise functional mapping of peripheral neural circuits, including long-term behavioral and physiological measurements. Deployment of these technologies reveals an unexpected role for stomach, non-stretch vagal sensory fibers in suppressing appetite and demonstrates the durability of the miniature wireless device inside harsh gastric conditions. Advances in wireless technologies have enabled internalisation of light sources, but organ specific illumination is challenging. Here, the authors present a durable, multimodal, wireless system enabling optogenetic stimulation of peripheral neurons within organs.
Collapse
|
50
|
Shahriari D, Rosenfeld D, Anikeeva P. Emerging Frontier of Peripheral Nerve and Organ Interfaces. Neuron 2020; 108:270-285. [PMID: 33120023 DOI: 10.1016/j.neuron.2020.09.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023]
Abstract
The development of new tools to interface with the nervous system, empowered by advances in electronics and materials science, has transformed neuroscience and is informing therapies for neurological and mental conditions. Although the vast majority of neural engineering research has focused on advancing tools to study the brain, understanding the peripheral nervous system and other organs can similarly benefit from these technologies. To realize this vision, the neural interface technologies need to address the biophysical, mechanical, and chemical challenges posed by the peripheral nerves and organs. In this Perspective, we discuss design considerations and recent technological advances to modulate electrical signaling outside the central nervous system. The innovations in bioelectronics borne out of interdisciplinary collaborations between biologists and physical scientists may not only advance fundamental study of peripheral (neuro)physiology but also empower clinical interventions for conditions including neurological, gastrointestinal, and immune dysfunction.
Collapse
Affiliation(s)
- Dena Shahriari
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|