1
|
Antoniazzi AM, Unda SR, Norman S, Pomeranz LE, Marongiu R, Stanley SA, Friedman JM, Kaplitt MG. Non-invasive in vivo bidirectional magnetogenetic modulation of pain circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.644041. [PMID: 40166248 PMCID: PMC11957015 DOI: 10.1101/2025.03.18.644041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Primary nociceptors in the dorsal root ganglion (DRG) receive sensory information from discrete parts of the body and are responsible for initiating signaling events that in supraspinal regions will be interpreted as physiological or pathological pain. Genetic, pharmacologic and electric neuromodulation of nociceptor activity in freely moving non-transgenic animals has been shown to be challenging due to many factors including the immunogenicity of non-mammalian proteins, procedure invasiveness and poor temporal precision. Here, we introduce a magnetogenetic strategy that enables remote bidirectional regulation of nociceptor activity. Magnetogenetics utilizes a source of direct magnetic field (DMF) to control neuronal activity in cells that express an anti-ferritin nanobody-TRPV1 receptor fusion protein (Nb-Ft-TRPV1). In our study, AAV2retro-mediated delivery of an excitatory Nb-Ft-TRPV1 construct into the sciatic nerve of wild-type mice resulted in stable long-term transgene expression accompanied by significant reduction of mechanical withdrawal thresholds during DMF exposure, place aversion of the DMF zone and activity changes in the anterior cingulate (ACC) nucleus. Conversely, delivery of an inhibitory variant of the Nb-Ft-TRPV1 construct, engineered to gate chloride ions in response to DMF, led to reversed behavioral manifestations of mechanical allodynia and showed place preference for the DMF zone, suggestive of functional pain relief. Changes in DRG activity were confirmed by post-mortem levels, immediately following DMF exposure, of the activity-induced gene cfos, which increased with the excitatory construct in normal mice and decreased with the inhibitory construct in pain models Our study demonstrates that magnetogenetic channels can achieve long-term expression in the periphery without losing functionality, providing a stable gene therapy system for non-invasive, magnetic field regulation of pain-related neurons for research and potential clinical applications.
Collapse
Affiliation(s)
- Aldana M. Antoniazzi
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Santiago R. Unda
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Sofya Norman
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Lisa E. Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | - Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| |
Collapse
|
2
|
Yang X, Kubican SE, Yi Z, Tong S. Advances in magnetic nanoparticles for molecular medicine. Chem Commun (Camb) 2025; 61:3093-3108. [PMID: 39846549 PMCID: PMC11756346 DOI: 10.1039/d4cc05167j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Magnetic nanoparticles (MNPs) are highly versatile nanomaterials in nanomedicine, owing to their diverse magnetic properties, which can be tailored through variations in size, shape, composition, and exposure to inductive magnetic fields. Over four decades of research have led to the clinical approval or ongoing trials of several MNP formulations, fueling continued innovation. Beyond traditional applications in drug delivery, imaging, and cancer hyperthermia, MNPs have increasingly advanced into molecular medicine. Under external magnetic fields, MNPs can generate mechano- or thermal stimuli to modulate individual molecules or cells deep within tissue, offering precise, remote control of biological processes at cellular and molecular levels. These unique capabilities have opened new avenues in emerging fields such as genome editing, cell therapies, and neuroscience, underpinned by a growing understanding of nanomagnetism and the molecular mechanisms responding to mechanical and thermal cues. Research on MNPs as a versatile synthetic material capable of engineering control at the cellular and molecular levels holds great promise for advancing the frontiers of molecular medicine, including areas such as genome editing and synthetic biology. This review summarizes recent clinical studies showcasing the classical applications of MNPs and explores their integration into molecular medicine, with the goal of inspiring the development of next-generation MNP-based platforms for disease treatment.
Collapse
Affiliation(s)
- Xiaoyue Yang
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sarah E Kubican
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Zhongchao Yi
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sheng Tong
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| |
Collapse
|
3
|
Bandesh K, Motakis E, Nargund S, Kursawe R, Selvam V, Bhuiyan RM, Eryilmaz GN, Krishnan SN, Spracklen CN, Ucar D, Stitzel ML. Single-cell decoding of human islet cell type-specific alterations in type 2 diabetes reveals converging genetic- and state-driven β -cell gene expression defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633590. [PMID: 39896672 PMCID: PMC11785113 DOI: 10.1101/2025.01.17.633590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Pancreatic islets maintain glucose homeostasis through coordinated action of their constituent endocrine and affiliate cell types and are central to type 2 diabetes (T2D) genetics and pathophysiology. Our understanding of robust human islet cell type-specific alterations in T2D remains limited. Here, we report comprehensive single cell transcriptome profiling of 245,878 human islet cells from a 48-donor cohort spanning non-diabetic (ND), pre-diabetic (PD), and T2D states, identifying 14 distinct cell types detected in every donor from each glycemic state. Cohort analysis reveals ~25-30% loss of functional beta cell mass in T2D vs. ND or PD donors resulting from (1) reduced total beta cell numbers/proportions and (2) reciprocal loss of 'high function' and gain of senescent β -cell subpopulations. We identify in T2D β -cells 511 differentially expressed genes (DEGs), including new (66.5%) and validated genes (e.g., FXYD2, SLC2A2, SYT1), and significant neuronal transmission and vitamin A metabolism pathway alterations. Importantly, we demonstrate newly identified DEG roles in human β -cell viability and/or insulin secretion and link 47 DEGs to diabetes-relevant phenotypes in knockout mice, implicating them as potential causal islet dysfunction genes. Additionally, we nominate as candidate T2D causal genes and therapeutic targets 27 DEGs for which T2D genetic risk variants (GWAS SNPs) and pathophysiology (T2D vs. ND) exert concordant expression effects. We provide this freely accessible atlas for data exploration, analysis, and hypothesis testing. Together, this study provides new genomic resources for and insights into T2D pathophysiology and human islet dysfunction.
Collapse
Affiliation(s)
- Khushdeep Bandesh
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Efthymios Motakis
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Siddhi Nargund
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Giray Naim Eryilmaz
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Sai Nivedita Krishnan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Cassandra N. Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| | - Michael L. Stitzel
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| |
Collapse
|
4
|
Zhi W, Li Y, Wang L, Hu X. Advancing Neuroscience and Therapy: Insights into Genetic and Non-Genetic Neuromodulation Approaches. Cells 2025; 14:122. [PMID: 39851550 PMCID: PMC11763439 DOI: 10.3390/cells14020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Neuromodulation stands as a cutting-edge approach in the fields of neuroscience and therapeutic intervention typically involving the regulation of neural activity through physical and chemical stimuli. The purpose of this review is to provide an overview and evaluation of different neuromodulation techniques, anticipating a clearer understanding of the future developmental trajectories and the challenges faced within the domain of neuromodulation that can be achieved. This review categorizes neuromodulation techniques into genetic neuromodulation methods (including optogenetics, chemogenetics, sonogenetics, and magnetogenetics) and non-genetic neuromodulation methods (including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, transcranial ultrasound stimulation, photobiomodulation therapy, infrared neuromodulation, electromagnetic stimulation, sensory stimulation therapy, and multi-physical-factor stimulation techniques). By systematically evaluating the principles, mechanisms, advantages, limitations, and efficacy in modulating neuronal activity and the potential applications in interventions of neurological disorders of these neuromodulation techniques, a comprehensive picture is gradually emerging regarding the advantages and challenges of neuromodulation techniques, their developmental trajectory, and their potential clinical applications. This review highlights significant advancements in applying these techniques to treat neurological and psychiatric disorders. Genetic methods, such as sonogenetics and magnetogenetics, have demonstrated high specificity and temporal precision in targeting neuronal populations, while non-genetic methods, such as transcranial magnetic stimulation and photobiomodulation therapy, offer noninvasive and versatile clinical intervention options. The transformative potential of these neuromodulation techniques in neuroscience research and clinical practice is underscored, emphasizing the need for integration and innovation in technologies, the optimization of delivery methods, the improvement of mediums, and the evaluation of toxicity to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Ying Li
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| |
Collapse
|
5
|
Zhai X, Wang Y. Physical modulation and peripheral nerve regeneration: a literature review. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:32. [PMID: 39710804 DOI: 10.1186/s13619-024-00215-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
Peripheral nerve injury (PNI) usually causes severe motor, sensory and autonomic dysfunction. In addition to direct surgical repair, rehabilitation exercises, and traditional physical stimuli, for example, electrical stimulation, have been applied in promoting the clinical recovery of PNI for a long time but showed low efficiency. Recently, significant progress has been made in new physical modulation to promote peripheral nerve regeneration. We hereby review current progress on the mechanism of peripheral nerve regeneration after injury and summarize the new findings and evidence for the application of physical modulation, including electrical stimulation, light, ultrasound, magnetic stimulation, and mechanical stretching in experimental studies and the clinical treatment of patients with PNI.
Collapse
Affiliation(s)
- Xiangwen Zhai
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yuzhong Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong Province, China.
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China.
| |
Collapse
|
6
|
Mirzadeh Z, Faber C. Brain Defense of Glycemia in Health and Diabetes. Diabetes 2024; 73:1952-1966. [PMID: 39401393 PMCID: PMC11579547 DOI: 10.2337/dbi24-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 11/22/2024]
Abstract
The brain coordinates the homeostatic defense of multiple metabolic variables, including blood glucose levels, in the context of ever-changing external and internal environments. The biologically defended level of glycemia (BDLG) is the net result of brain modulation of insulin-dependent mechanisms in cooperation with the islet, and insulin-independent mechanisms through direct innervation and neuroendocrine control of glucose effector tissues. In this article, we highlight evidence from animal and human studies to develop a framework for the brain's core homeostatic functions-sensory/afferent, integration/processing, and motor/efferent-that contribute to the normal BDLG in health and its elevation in diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Chelsea Faber
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| |
Collapse
|
7
|
Herzog H, Zhang L, Fontana L, Neely GG. Impact of non-sugar sweeteners on metabolism beyond sweet taste perception. Trends Endocrinol Metab 2024:S1043-2760(24)00276-5. [PMID: 39551640 DOI: 10.1016/j.tem.2024.10.008] [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: 10/01/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
Non-sugar sweeteners (NSS), low- or no-calorie alternatives to sugar, are marketed for weight loss and improved blood glucose control in people with diabetes. However, their health effects remain controversial. This review provides a brief overview of sweet taste perception and summarizes experimental findings of the impact of NSS on cardiometabolic health in animal models and humans. We also review evidence suggesting that many NSS are not metabolically inert, highlighting the challenges in related human studies. Given the conflicting and unclear data on health outcomes, additional mechanistic studies, particularly in animal models, are necessary to clarify how NSS influence feeding behaviors and energy homoeostasis.
Collapse
Affiliation(s)
- Herbert Herzog
- St Vincent's Centre for Applied Medical Research, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia.
| | - Lei Zhang
- St Vincent's Centre for Applied Medical Research, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia
| | - Luigi Fontana
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - G Gregory Neely
- Dr John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
8
|
Unda SR, Pomeranz LE, Marongiu R, Yu X, Kelly L, Hassanzadeh G, Molina H, Vaisey G, Wang P, Dyke JP, Fung EK, Grosenick L, Zirkel R, Antoniazzi AM, Norman S, Liston CM, Schaffer C, Nishimura N, Stanley SA, Friedman JM, Kaplitt MG. Bidirectional regulation of motor circuits using magnetogenetic gene therapy. SCIENCE ADVANCES 2024; 10:eadp9150. [PMID: 39383230 PMCID: PMC11463271 DOI: 10.1126/sciadv.adp9150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Here, we report a magnetogenetic system, based on a single anti-ferritin nanobody-TRPV1 receptor fusion protein, which regulated neuronal activity when exposed to magnetic fields. Adeno-associated virus (AAV)-mediated delivery of a floxed nanobody-TRPV1 into the striatum of adenosine-2a receptor-Cre drivers resulted in motor freezing when placed in a magnetic resonance imaging machine or adjacent to a transcranial magnetic stimulation device. Functional imaging and fiber photometry confirmed activation in response to magnetic fields. Expression of the same construct in the striatum of wild-type mice along with a second injection of an AAVretro expressing Cre into the globus pallidus led to similar circuit specificity and motor responses. Last, a mutation was generated to gate chloride and inhibit neuronal activity. Expression of this variant in the subthalamic nucleus in PitX2-Cre parkinsonian mice resulted in reduced c-fos expression and motor rotational behavior. These data demonstrate that magnetogenetic constructs can bidirectionally regulate activity of specific neuronal circuits noninvasively in vivo using clinically available devices.
Collapse
Affiliation(s)
- Santiago R. Unda
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Lisa E. Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Jonathan P. Dyke
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Edward K. Fung
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Logan Grosenick
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Rick Zirkel
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Aldana M. Antoniazzi
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Sofya Norman
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Conor M. Liston
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Chris Schaffer
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Nozomi Nishimura
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Jeffrey M. Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
9
|
Thorens B. Neuronal glucose sensing mechanisms and circuits in the control of insulin and glucagon secretion. Physiol Rev 2024; 104:1461-1486. [PMID: 38661565 DOI: 10.1152/physrev.00038.2023] [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: 09/29/2023] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024] Open
Abstract
Glucose homeostasis is mainly under the control of the pancreatic islet hormones insulin and glucagon, which, respectively, stimulate glucose uptake and utilization by liver, fat, and muscle and glucose production by the liver. The balance between the secretions of these hormones is under the control of blood glucose concentrations. Indeed, pancreatic islet β-cells and α-cells can sense variations in glycemia and respond by an appropriate secretory response. However, the secretory activity of these cells is also under multiple additional metabolic, hormonal, and neuronal signals that combine to ensure the perfect control of glycemia over a lifetime. The central nervous system (CNS), which has an almost absolute requirement for glucose as a source of metabolic energy and thus a vital interest in ensuring that glycemic levels never fall below ∼5 mM, is equipped with populations of neurons responsive to changes in glucose concentrations. These neurons control pancreatic islet cell secretion activity in multiple ways: through both branches of the autonomic nervous system, through the hypothalamic-pituitary-adrenal axis, and by secreting vasopressin (AVP) in the blood at the level of the posterior pituitary. Here, we present the autonomic innervation of the pancreatic islets; the mechanisms of neuron activation by a rise or a fall in glucose concentration; how current viral tracing, chemogenetic, and optogenetic techniques allow integration of specific glucose sensing neurons in defined neuronal circuits that control endocrine pancreas function; and, finally, how genetic screens in mice can untangle the diversity of the hypothalamic mechanisms controlling the response to hypoglycemia.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Choi SH, Shin J, Park C, Lee JU, Lee J, Ambo Y, Shin W, Yu R, Kim JY, Lah JD, Shin D, Kim G, Noh K, Koh W, Lee CJ, Lee JH, Kwak M, Cheon J. In vivo magnetogenetics for cell-type-specific targeting and modulation of brain circuits. NATURE NANOTECHNOLOGY 2024; 19:1333-1343. [PMID: 38956320 DOI: 10.1038/s41565-024-01694-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 05/05/2024] [Indexed: 07/04/2024]
Abstract
Neuromodulation technologies are crucial for investigating neuronal connectivity and brain function. Magnetic neuromodulation offers wireless and remote deep brain stimulations that are lacking in optogenetic- and wired-electrode-based tools. However, due to the limited understanding of working principles and poorly designed magnetic operating systems, earlier magnetic approaches have yet to be utilized. Furthermore, despite its importance in neuroscience research, cell-type-specific magnetic neuromodulation has remained elusive. Here we present a nanomaterials-based magnetogenetic toolbox, in conjunction with Cre-loxP technology, to selectively activate genetically encoded Piezo1 ion channels in targeted neuronal populations via torque generated by the nanomagnetic actuators in vitro and in vivo. We demonstrate this cell-type-targeting magnetic approach for remote and spatiotemporal precise control of deep brain neural activity in multiple behavioural models, such as bidirectional feeding control, long-term neuromodulation for weight control in obese mice and wireless modulation of social behaviours in multiple mice in the same physical space. Our study demonstrates the potential of cell-type-specific magnetogenetics as an effective and reliable research tool for life sciences, especially in wireless, long-term and freely behaving animals.
Collapse
Affiliation(s)
- Seo-Hyun Choi
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Jihye Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Chanhyun Park
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Jung-Uk Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Jaegyeong Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Yuko Ambo
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Ri Yu
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Ju-Young Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Jungsu David Lah
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Donghun Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Gooreum Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Wuhyun Koh
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - C Justin Lee
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| | - Minsuk Kwak
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Tian W, Jia Q, Lin J, Luo J, He D, Yang J, Guo T, Guo H, Guo Y, Zhang W, Chen F, Ye Y, Liu J, Xu M, Deng C, Cui B, Su D, Wang H, Lu Y, Xiao J, Liu H, Yang J, Hou Z, Wang S. Remote neurostimulation through an endogenous ion channel using a near-infrared light-activatable nanoagonist. SCIENCE ADVANCES 2024; 10:eadn0367. [PMID: 39121219 PMCID: PMC11313869 DOI: 10.1126/sciadv.adn0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/02/2024] [Indexed: 08/11/2024]
Abstract
The development of noninvasive approaches to precisely control neural activity in mammals is highly desirable. Here, we used the ion channel transient receptor potential ankyrin-repeat 1 (TRPA1) as a proof of principle, demonstrating remote near-infrared (NIR) activation of endogenous neuronal channels in mice through an engineered nanoagonist. This achievement enables specific neurostimulation in nongenetically modified mice. Initially, target-based screening identified flavins as photopharmacological agonists, allowing for the photoactivation of TRPA1 in sensory neurons upon ultraviolet A/blue light illumination. Subsequently, upconversion nanoparticles (UCNPs) were customized with an emission spectrum aligned to flavin absorption and conjugated with flavin adenine dinucleotide, creating a nanoagonist capable of NIR activation of TRPA1. Following the intrathecal injection of the nanoagonist, noninvasive NIR stimulation allows precise bidirectional control of nociception in mice through remote activation of spinal TRPA1. This study demonstrates a noninvasive NIR neurostimulation method with the potential for adaptation to various endogenous ion channels and neural processes by combining photochemical toolboxes with customized UCNPs.
Collapse
Affiliation(s)
- Weifeng Tian
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Institute of Organoid Technology, Kunming Medical University, Kunming, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Qi Jia
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiewen Lin
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Luo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dongmei He
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Yang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tao Guo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huiling Guo
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yusheng Guo
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, The Affiliated TCM Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, GMU-GIBH Joint School of Life Sciences, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Wenjie Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Feiyu Chen
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ying Ye
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingjing Liu
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Mindong Xu
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chengjie Deng
- Cell Biology and Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Boxiang Cui
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Deyuan Su
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hao Wang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Lu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Heng Liu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, The Affiliated TCM Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, GMU-GIBH Joint School of Life Sciences, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Zhiyao Hou
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shu Wang
- The Affiliated TCM Hospital of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Hernández-Morales M, Morales-Weil K, Han SM, Han V, Tran T, Benner EJ, Pegram K, Meanor J, Miller EW, Kramer RH, Liu C. Electrophysiological Mechanisms and Validation of Ferritin-Based Magnetogenetics for Remote Control of Neurons. J Neurosci 2024; 44:e1717232024. [PMID: 38777598 PMCID: PMC11270515 DOI: 10.1523/jneurosci.1717-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Magnetogenetics was developed to remotely control genetically targeted neurons. A variant of magnetogenetics uses magnetic fields to activate transient receptor potential vanilloid (TRPV) channels when coupled with ferritin. Stimulation with static or RF magnetic fields of neurons expressing these channels induces Ca2+ transients and modulates behavior. However, the validity of ferritin-based magnetogenetics has been questioned due to controversies surrounding the underlying mechanisms and deficits in reproducibility. Here, we validated the magnetogenetic approach Ferritin-iron Redistribution to Ion Channels (FeRIC) using electrophysiological (Ephys) and imaging techniques. Previously, interference from RF stimulation rendered patch-clamp recordings inaccessible for magnetogenetics. We solved this limitation for FeRIC, and we studied the bioelectrical properties of neurons expressing TRPV4 (nonselective cation channel) and transmembrane member 16A (TMEM16A; chloride-permeable channel) coupled to ferritin (FeRIC channels) under RF stimulation. We used cultured neurons obtained from the rat hippocampus of either sex. We show that RF decreases the membrane resistance (Rm) and depolarizes the membrane potential in neurons expressing TRPV4FeRIC RF does not directly trigger action potential firing but increases the neuronal basal spiking frequency. In neurons expressing TMEM16AFeRIC, RF decreases the Rm, hyperpolarizes the membrane potential, and decreases the spiking frequency. Additionally, we corroborated the previously described biochemical mechanism responsible for RF-induced activation of ferritin-coupled ion channels. We solved an enduring problem for ferritin-based magnetogenetics, obtaining direct Ephys evidence of RF-induced activation of ferritin-coupled ion channels. We found that RF does not yield instantaneous changes in neuronal membrane potentials. Instead, RF produces responses that are long-lasting and moderate, but effective in controlling the bioelectrical properties of neurons.
Collapse
Affiliation(s)
- Miriam Hernández-Morales
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Koyam Morales-Weil
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Sang Min Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Victor Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Tiffany Tran
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Jenna Meanor
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Evan W Miller
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Department of Chemistry, University of California, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| |
Collapse
|
13
|
Alfihed S, Majrashi M, Ansary M, Alshamrani N, Albrahim SH, Alsolami A, Alamari HA, Zaman A, Almutairi D, Kurdi A, Alzaydi MM, Tabbakh T, Al-Otaibi F. Non-Invasive Brain Sensing Technologies for Modulation of Neurological Disorders. BIOSENSORS 2024; 14:335. [PMID: 39056611 PMCID: PMC11274405 DOI: 10.3390/bios14070335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
The non-invasive brain sensing modulation technology field is experiencing rapid development, with new techniques constantly emerging. This study delves into the field of non-invasive brain neuromodulation, a safer and potentially effective approach for treating a spectrum of neurological and psychiatric disorders. Unlike traditional deep brain stimulation (DBS) surgery, non-invasive techniques employ ultrasound, electrical currents, and electromagnetic field stimulation to stimulate the brain from outside the skull, thereby eliminating surgery risks and enhancing patient comfort. This study explores the mechanisms of various modalities, including transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS), highlighting their potential to address chronic pain, anxiety, Parkinson's disease, and depression. We also probe into the concept of closed-loop neuromodulation, which personalizes stimulation based on real-time brain activity. While we acknowledge the limitations of current technologies, our study concludes by proposing future research avenues to advance this rapidly evolving field with its immense potential to revolutionize neurological and psychiatric care and lay the foundation for the continuing advancement of innovative non-invasive brain sensing technologies.
Collapse
Affiliation(s)
- Salman Alfihed
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Majed Majrashi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Muhammad Ansary
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Naif Alshamrani
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Shahad H. Albrahim
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Abdulrahman Alsolami
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Hala A. Alamari
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Adnan Zaman
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Dhaifallah Almutairi
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Abdulaziz Kurdi
- Advanced Materials Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Mai M. Alzaydi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Thamer Tabbakh
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Faisal Al-Otaibi
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
14
|
García-González J, Garcia-Gonzalez S, Liou L, O'Reilly PF. The Gene Expression Landscape of Disease Genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.20.24309121. [PMID: 38947033 PMCID: PMC11213058 DOI: 10.1101/2024.06.20.24309121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Fine-mapping and gene-prioritisation techniques applied to the latest Genome-Wide Association Study (GWAS) results have prioritised hundreds of genes as causally associated with disease. Here we leverage these recently compiled lists of high-confidence causal genes to interrogate where in the body disease genes operate. Specifically, we combine GWAS summary statistics, gene prioritisation results and gene expression RNA-seq data from 46 tissues and 204 cell types in relation to 16 major diseases (including 8 cancers). In tissues and cell types with well-established relevance to the disease, the prioritised genes typically have higher absolute and relative (i.e. tissue/cell specific) expression compared to non-prioritised 'control' genes. Examples include brain tissues in psychiatric disorders (P-value < 1×10-7), microglia cells in Alzheimer's Disease (P-value = 9.8×10-3) and colon mucosa in colorectal cancer (P-value < 1×10-3). We also observe significantly higher expression for disease genes in multiple tissues and cell types with no established links to the corresponding disease. While some of these results may be explained by cell types that span multiple tissues, such as macrophages in brain, blood, lung and spleen in relation to Alzheimer's disease (P-values < 1×10-3), the cause for others is unclear and motivates further investigation that may provide novel insights into disease etiology. For example, mammary tissue in Type 2 Diabetes (P-value < 1×10-7); reproductive tissues such as breast, uterus, vagina, and prostate in Coronary Artery Disease (P-value < 1×10-4); and motor neurons in psychiatric disorders (P-value < 3×10-4). In the GTEx dataset, tissue type is the major predictor of gene expression but the contribution of each predictor (tissue, sample, subject, batch) varies widely among disease-associated genes. Finally, we highlight genes with the highest levels of gene expression in relevant tissues to guide functional follow-up studies. Our results could offer novel insights into the tissues and cells involved in disease initiation, inform drug target and delivery strategies, highlighting potential off-target effects, and exemplify the relative performance of different statistical tests for linking disease genes with tissue and cell type gene expression.
Collapse
Affiliation(s)
- Judit García-González
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Saul Garcia-Gonzalez
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Lathan Liou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Paul F O'Reilly
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| |
Collapse
|
15
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-z] [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: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
16
|
Chen C, Chen H, Wang P, Wang X, Wang X, Chen C, Pan W. Reactive Oxygen Species Activate a Ferritin-Linked TRPV4 Channel under a Static Magnetic Field. ACS Chem Biol 2024; 19:1151-1160. [PMID: 38648729 DOI: 10.1021/acschembio.4c00090] [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: 04/25/2024]
Abstract
Magnetogenetics has shown great potential for cell function and neuromodulation using heat or force effects under different magnetic fields; however, there is still a contradiction between experimental effects and underlying mechanisms by theoretical computation. In this study, we aimed to investigate the role of reactive oxygen species (ROS) in mechanical force-dependent regulation from a physicochemical perspective. The transient receptor potential vanilloid 4 (TRPV4) cation channels fused to ferritin (T4F) were overexpressed in HEK293T cells and exposed to static magnetic fields (sMF, 1.4-5.0 mT; gradient: 1.62 mT/cm). An elevation of ROS levels was found under sMF in T4F-overexpressing cells, which could lead to lipid oxidation. Compared with the overexpression of TRPV4, ferritin in T4F promoted the generation of ROS under the stimulation of sMF, probably related to the release of iron ions from ferritin. Then, the resulting ROS regulated the opening of the TRPV4 channel, which was attenuated by the direct addition of ROS inhibitors or an iron ion chelator, highlighting a close relationship among iron release, ROS production, and TRPV4 channel activation. Taken together, these findings indicate that the produced ROS under sMF act on the TRPV4 channel, regulating the influx of calcium ions. The study would provide a scientific basis for the application of magnetic regulation in cellular or neural regulation and disease treatment and contribute to the development of the more sensitive regulatory technology.
Collapse
Affiliation(s)
- Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chuanfang Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Weidong Pan
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| |
Collapse
|
17
|
Chen C, Chen H, Wang P, Wang X, Wang X, Chen C. Ca 2+ Overload Decreased Cellular Viability in Magnetic Hyperthermia without a Macroscopic Temperature Rise. ACS Biomater Sci Eng 2024; 10:2995-3005. [PMID: 38654432 DOI: 10.1021/acsbiomaterials.3c01875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Magnetic hyperthermia is a crucial medical engineering technique for treating diseases, which usually uses alternating magnetic fields (AMF) to interplay with magnetic substances to generate heat. Recently, it has been found that in some cases, there is no detectable temperature increment after applying an AMF, which caused corresponding effects surprisingly. The mechanisms involved in this phenomenon are not yet fully understood. In this study, we aimed to explore the role of Ca2+ overload in the magnetic hyperthermia effect without a perceptible temperature rise. A cellular system expressing the fusion proteins TRPV1 and ferritin was prepared. The application of an AMF (518 kHz, 16 kA/m) could induce the fusion protein to release a large amount of iron ions, which then participates in the production of massive reactive oxygen radicals (ROS). Both ROS and its induced lipid oxidation enticed the opening of ion channels, causing intracellular Ca2+ overload, which further led to decreased cellular viability. Taken together, Ca2+ overload triggered by elevated ROS and the induced oxidation of lipids contributes to the magnetic hyperthermia effect without a perceptible temperature rise. These findings would be beneficial for expanding the application of temperature-free magnetic hyperthermia, such as in cellular and neural regulation, design of new cancer treatment methods.
Collapse
Affiliation(s)
- Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chuanfang Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| |
Collapse
|
18
|
Guan W, Gao H, Liu Y, Sun S, Li G. Application of magnetism in tissue regeneration: recent progress and future prospects. Regen Biomater 2024; 11:rbae048. [PMID: 38939044 PMCID: PMC11208728 DOI: 10.1093/rb/rbae048] [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: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 06/29/2024] Open
Abstract
Tissue regeneration is a hot topic in the field of biomedical research in this century. Material composition, surface topology, light, ultrasonic, electric field and magnetic fields (MFs) all have important effects on the regeneration process. Among them, MFs can provide nearly non-invasive signal transmission within biological tissues, and magnetic materials can convert MFs into a series of signals related to biological processes, such as mechanical force, magnetic heat, drug release, etc. By adjusting the MFs and magnetic materials, desired cellular or molecular-level responses can be achieved to promote better tissue regeneration. This review summarizes the definition, classification and latest progress of MFs and magnetic materials in tissue engineering. It also explores the differences and potential applications of MFs in different tissue cells, aiming to connect the applications of magnetism in various subfields of tissue engineering and provide new insights for the use of magnetism in tissue regeneration.
Collapse
Affiliation(s)
- Wenchao Guan
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Hongxia Gao
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yaqiong Liu
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Shaolan Sun
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
19
|
Chen H, Xie Y, Zhang M, Huang J, Jiang W, Zhang R, Li C, Du X, Chen H, Nie Q, Liang S, Tan Q, Yang J, Jin M, Huang S, Kuang L, Su N, Qi H, Luo X, Xu X, Deng C, Chen L, Luo F. An Hsp70 promoter-based mouse for heat shock-induced gene modulation. J Mol Med (Berl) 2024; 102:693-707. [PMID: 38492027 DOI: 10.1007/s00109-024-02433-9] [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: 01/04/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.
Collapse
Affiliation(s)
- Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Mei Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Department of Chinese Medicine Rehabilitation, Chongqing Emergency Medical Center, Chongqing University Central Hospital), Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Hua Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiang Nie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shuo Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
20
|
Unda SR, Pomeranz LE, Marongiu R, Yu X, Kelly L, Hassanzadeh G, Molina H, Vaisey G, Wang P, Dyke JP, Fung EK, Grosenick L, Zirkel R, Antoniazzi AM, Norman S, Liston CM, Schaffer C, Nishimura N, Stanley SA, Friedman JM, Kaplitt MG. Bidirectional Regulation of Motor Circuits Using Magnetogenetic Gene Therapy Short: Magnetogenetic Regulation of Motor Circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.13.548699. [PMID: 37503198 PMCID: PMC10369996 DOI: 10.1101/2023.07.13.548699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Here we report a novel suite of magnetogenetic tools, based on a single anti-ferritin nanobody-TRPV1 receptor fusion protein, which regulated neuronal activity when exposed to magnetic fields. AAV-mediated delivery of a floxed nanobody-TRPV1 into the striatum of adenosine 2a receptor-cre driver mice resulted in motor freezing when placed in an MRI or adjacent to a transcranial magnetic stimulation (TMS) device. Functional imaging and fiber photometry both confirmed activation of the target region in response to the magnetic fields. Expression of the same construct in the striatum of wild-type mice along with a second injection of an AAVretro expressing cre into the globus pallidus led to similar circuit specificity and motor responses. Finally, a mutation was generated to gate chloride and inhibit neuronal activity. Expression of this variant in subthalamic nucleus in PitX2-cre parkinsonian mice resulted in reduced local c-fos expression and motor rotational behavior. These data demonstrate that magnetogenetic constructs can bidirectionally regulate activity of specific neuronal circuits non-invasively in-vivo using clinically available devices.
Collapse
Affiliation(s)
- Santiago R. Unda
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Lisa E. Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | - Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai, 200433
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | - Jonathan P. Dyke
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Edward K. Fung
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Logan Grosenick
- Department of Psychiatry, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Rick Zirkel
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aldana M. Antoniazzi
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Sofya Norman
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Conor M. Liston
- Department of Psychiatry, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Chris Schaffer
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nozomi Nishimura
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| |
Collapse
|
21
|
Pomeranz L, Li R, Yu X, Kelly L, Hassanzadeh G, Molina H, Gross D, Brier M, Vaisey G, Wang P, Jimenez-Gonzalez M, Garcia-Ocana A, Dordick J, Friedman J, Stanley S. Magnetogenetic cell activation using endogenous ferritin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.20.545120. [PMID: 37786709 PMCID: PMC10541561 DOI: 10.1101/2023.06.20.545120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The ability to precisely control the activity of defined cell populations enables studies of their physiological roles and may provide therapeutic applications. While prior studies have shown that magnetic activation of ferritin-tagged ion channels allows cell-specific modulation of cellular activity, the large size of the constructs made the use of adeno-associated virus, AAV, the vector of choice for gene therapy, impractical. In addition, simple means for generating magnetic fields of sufficient strength have been lacking. Toward these ends, we first generated a novel anti-ferritin nanobody that when fused to transient receptor potential cation channel subfamily V member 1, TRPV1, enables direct binding of the channel to endogenous ferritin in mouse and human cells. This smaller construct can be delivered in a single AAV and we validated that it robustly enables magnetically induced cell activation in vitro. In parallel, we developed a simple benchtop electromagnet capable of gating the nanobody-tagged channel in vivo. Finally, we showed that delivering these new constructs by AAV to pancreatic beta cells in combination with the benchtop magnetic field delivery stimulates glucose-stimulated insulin release to improve glucose tolerance in mice in vivo. Together, the novel anti-ferritin nanobody, nanobody-TRPV1 construct and new hardware advance the utility of magnetogenetics in animals and potentially humans.
Collapse
Affiliation(s)
- Lisa Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Rosemary Li
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai, 200433
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel Gross
- Current address, Dept. of Radiology, Weill Cornell Medicine, 1300 York Avenue New York, NY 10065
| | - Matthew Brier
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocana
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010
| | - Jonathan Dordick
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Jeffrey Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Sarah Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
Mundell JW, Brier MI, Orloff E, Stanley SA, Dordick JS. Alternating magnetic fields drive stimulation of gene expression via generation of reactive oxygen species. iScience 2024; 27:109186. [PMID: 38420587 PMCID: PMC10901079 DOI: 10.1016/j.isci.2024.109186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/23/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Magnetogenetics represents a method for remote control of cellular function. Previous work suggests that generation of reactive oxygen species (ROS) initiates downstream signaling. Herein, a chemical biology approach was used to elucidate further the mechanism of radio frequency-alternating magnetic field (RF-AMF) stimulation of a TRPV1-ferritin magnetogenetics platform that leads to Ca2+ flux. RF-AMF stimulation of HEK293T cells expressing TRPV1-ferritin resulted in ∼30% and ∼140% increase in intra- and extracellular ROS levels, respectively. Mutations to specific cysteine residues in TRPV1 responsible for ROS sensitivity eliminated RF-AMF driven Ca2+-dependent transcription of secreted embryonic alkaline phosphatase (SEAP). Using a non-tethered (to TRPV1) ferritin also eliminated RF-AMF driven SEAP production, and using specific inhibitors, ROS-activated TRPV1 signaling involves protein kinase C, NADPH oxidase, and the endoplasmic reticulum. These results suggest ferritin-dependent ROS activation of TRPV1 plays a key role in the initiation of magnetogenetics, and provides relevance for potential applications in medicine and biotechnology.
Collapse
Affiliation(s)
- Jordan W. Mundell
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Matthew I. Brier
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Everest Orloff
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan S. Dordick
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Departments of Biomedical Engineering and Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
23
|
Yi Z, Yang X, Liang Y, Chapelin F, Tong S. Enhancing ROS-Inducing Nanozyme through Intraparticle Electron Transport. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305974. [PMID: 37771197 PMCID: PMC10922328 DOI: 10.1002/smll.202305974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/01/2023] [Indexed: 09/30/2023]
Abstract
Iron oxide nanoparticles (IONPs) have garnered significant attention as a promising platform for reactive oxygen species (ROS)-dependent disease treatment, owing to their remarkable biocompatibility and Fenton catalytic activity. However, the low catalytic activity of IONPs is a major hurdle in their clinical translation. To overcome this challenge, IONPs of different compositions are examined for their Fenton reaction under pharmacologically relevant conditions. The results show that wüstite (FeO) nanoparticles exhibit higher catalytic activity than magnetite (Fe3 O4 ) or maghemite (γ-Fe2 O3 ) of matched size and coating, despite having a similar surface oxidation state. Further analyses suggest that the high catalytic activity of wüstite nanoparticles can be attributed to the presence of internal low-valence iron (Fe0 and Fe2+ ), which accelerates the recycling of surface Fe3+ to Fe2+ through intraparticle electron transport. Additionally, ultrasmall wüstite nanoparticles are generated by tuning the thermodecomposition-based nanocrystal synthesis, resulting in a Fenton reaction rate 5.3 times higher than that of ferumoxytol, an FDA-approved IONP. Compared with ferumoxytol, wüstite nanoparticles substantially increase the level of intracellular ROS in mouse mammary carcinoma cells. This study presents a novel mechanism and pivotal improvement for the development of highly efficient ROS-inducing nanozymes, thereby expanding the horizons for their therapeutic applications.
Collapse
Affiliation(s)
- Zhongchao Yi
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, 40536, USA
| | - Xiaoyue Yang
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, 40536, USA
| | - Ying Liang
- New York Blood Center, New York, NY, 10065, USA
| | - Fanny Chapelin
- Shu Chien - Gene Lay Department of Bioengineering & Department of Radiology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sheng Tong
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
24
|
Juras JA, Pitra S, Smith BN. Systemic Glucose Regulation by a Hindbrain Inhibitory Circuit in a Mouse Model of Type 1 Diabetes. Neuroendocrinology 2024; 114:302-312. [PMID: 38194945 DOI: 10.1159/000536142] [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: 07/14/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Previous work showed that increasing the electrical activity of inhibitory neurons in the dorsal vagal complex (DVC) is sufficient to increase whole-body glucose concentration in normoglycemic mice. Here we tested the hypothesis that deactivating GABAergic neurons in the dorsal hindbrain of hyperglycemic mice decreases synaptic inhibition of parasympathetic motor neurons in the dorsal motor nucleus of the vagus (DMV) and reduces systemic glucose levels. METHODS Chemogenetic activation or inactivation of GABAergic neurons in the nucleus tractus solitarius (NTS) was used to assess effects of modulating parasympathetic output on blood glucose concentration in normoglycemic and hyperglycemic mice. Patch-clamp electrophysiology in vitro was used to assess cellular effects of chemogenetic manipulation of NTS GABA neurons. RESULTS Chemogenetic activation of GABAergic NTS neurons in normoglycemic mice increased their action potential firing, resulting in increased inhibitory synaptic input to DMV motor neurons and elevated blood glucose concentration. Deactivation of GABAergic DVC neurons in normoglycemic mice altered their electrical activity but did not alter systemic glucose levels. Conversely, stimulation of GABAergic DVC neurons in mice that were hyperglycemic subsequent to treatment with streptozotocin changed their electrical activity but did not alter whole-body glucose concentration, while deactivation of this inhibitory circuit significantly decreased circulating glucose concentration. Peripheral administration of a brain impermeant muscarinic acetylcholine receptor antagonist abolished these effects. CONCLUSION Disinhibiting vagal motor neurons decreases hyperglycemia in a mouse model of type 1 diabetes. This inhibitory brainstem circuit emerges as a key parasympathetic regulator of whole-body glucose homeostasis that undergoes functional plasticity in hyperglycemic conditions.
Collapse
Affiliation(s)
- J Anna Juras
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Soledad Pitra
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Bret N Smith
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
25
|
Zhuang S, He M, Feng J, Peng S, Jiang H, Li Y, Hua N, Zheng Y, Ye Q, Hu M, Nie Y, Yu P, Yue X, Qian J, Yang W. Near-Infrared Photothermal Manipulates Cellular Excitability and Animal Behavior in Caenorhabditis elegans. SMALL METHODS 2023; 7:e2300848. [PMID: 37681531 DOI: 10.1002/smtd.202300848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/12/2023] [Indexed: 09/09/2023]
Abstract
Near-infrared (NIR) photothermal manipulation has emerged as a promising and noninvasive technology for neuroscience research and disease therapy for its deep tissue penetration. NIR stimulated techniques have been used to modulate neural activity. However, due to the lack of suitable in vivo control systems, most studies are limited to the cellular level. Here, a NIR photothermal technique is developed to modulate cellular excitability and animal behaviors in Caenorhabditis elegans in vivo via the thermosensitive transient receptor potential vanilloid 1 (TRPV1) channel with an FDA-approved photothermal agent indocyanine green (ICG). Upon NIR stimuli, exogenous expression of TRPV1 in AFD sensory neurons causes Ca2+ influx, leading to increased neural excitability and reversal behaviors, in the presence of ICG. The GABAergic D-class motor neurons can also be activated by NIR irradiation, resulting in slower thrashing behaviors. Moreover, the photothermal manipulation is successfully applied in different types of muscle cells (striated muscles and nonstriated muscles), enhancing muscular excitability, causing muscle contractions and behavior changes in vivo. Altogether, this study demonstrates a noninvasive method to precisely regulate the excitability of different types of cells and related behaviors in vivo by NIR photothermal manipulation, which may be applied in mammals and clinical therapy.
Collapse
Affiliation(s)
- Siyi Zhuang
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mubin He
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China
| | - Jiaqi Feng
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Shiyi Peng
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China
| | - Haochen Jiang
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yunhao Li
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ning Hua
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yujie Zheng
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qizhen Ye
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Miaojin Hu
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ying Nie
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Peilin Yu
- Department of Toxicology, Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiaomin Yue
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun Qian
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China
| | - Wei Yang
- Department of Biophysics, Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
26
|
Wang L, Chang G, Yang M, Xu Z, Wang J, Xu H, He M, Dai L, Zhao Y, Ji Z, Zhang L. The Noninvasive Sonothermogenetics Used for Neuromodulation in M1 Region of Mice Brain by Overexpression of TRPV1. Neuroscience 2023; 527:22-36. [PMID: 37482284 DOI: 10.1016/j.neuroscience.2023.07.019] [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: 02/26/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023]
Abstract
Sonogenetics is preferred for neuroregulation and the treatment of brain diseases due to its noninvasive properties. Ultrasonic stimulation produces thermal and mechanical effects, among others. Since transient receptor potential vanilloid 1 (TRPV1) could be activated at 42 °C, it is overexpressed in the M1 region of the mouse motor cortex to sense the change of temperature upon being stimulated by focused ultrasound. Whether the heat generated by ultrasonic stimulation could activate TRPV1 in the M1 region and induce changes in electromyography (EMG) signals collected from the mice's triceps was carefully verified. The position of the focused ultrasound and the temperature of the tissue at the location of the focused position were simulated using COMSOL software and verified via experiments. For Neuro-2a cells with TRPV1 overexpression, 42 °C could activate the TRPV1 and induce calcium influx. For mice with TRPV1 overexpression in the M1 region, tissue temperature of >42 °C in the M1 region induces an increased number of cfos, suggesting that neurons with overexpressed TRPV1 in the M1 region can be activated using focused ultrasound. Furthermore, when the temperature is >42 °C, the peak-to-peak value of the EMG signal for mice with TRPV1 overexpression in the M1 region was higher than that for mice without TRPV1 overexpression. The immunohistochemical results showed that ultrasound was not harmful to the stimulation site. The noninvasive ultrasound stimulation combined with thermosensitive protein TRPV1 overexpressed in neurocytes as sonothermogenetics technology has great potential to be used for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Lulu Wang
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Guanglei Chang
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Miaomiao Yang
- School of Basic Medical Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Zhaobin Xu
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Jianye Wang
- Institute of Physics, Henan Academy of Sciences, Zhengzhou 450046, China
| | - Hongliang Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan Province, China
| | - Meixia He
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Liping Dai
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Yang Zhao
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Zhenyu Ji
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China.
| | - Liguo Zhang
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China.
| |
Collapse
|
27
|
Meng A, Ameroso D, Rios M. mGluR5 in Astrocytes in the Ventromedial Hypothalamus Regulates Pituitary Adenylate Cyclase-Activating Polypeptide Neurons and Glucose Homeostasis. J Neurosci 2023; 43:5918-5935. [PMID: 37507231 PMCID: PMC10436691 DOI: 10.1523/jneurosci.0193-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/09/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The ventromedial hypothalamus (VMH) is a functionally heterogeneous nucleus critical for systemic energy, glucose, and lipid balance. We showed previously that the metabotropic glutamate receptor 5 (mGluR5) plays essential roles regulating excitatory and inhibitory transmission in SF1+ neurons of the VMH and facilitating glucose and lipid homeostasis in female mice. Although mGluR5 is also highly expressed in VMH astrocytes in the mature brain, its role there influencing central metabolic circuits is unknown. In contrast to the glucose intolerance observed only in female mice lacking mGluR5 in VMH SF1 neurons, selective depletion of mGluR5 in VMH astrocytes enhanced glucose tolerance without affecting food intake or body weight in both adult female and male mice. The improved glucose tolerance was associated with elevated glucose-stimulated insulin release. Astrocytic mGluR5 male and female mutants also exhibited reduced adipocyte size and increased sympathetic tone in gonadal white adipose tissue. Diminished excitatory drive and synaptic inputs onto VMH Pituitary adenylate cyclase-activating polypeptide (PACAP+) neurons and reduced activity of these cells during acute hyperglycemia underlie the observed changes in glycemic control. These studies reveal an essential role of astrocytic mGluR5 in the VMH regulating the excitatory drive onto PACAP+ neurons and activity of these cells facilitating glucose homeostasis in male and female mice.SIGNIFICANCE STATEMENT Neuronal circuits within the VMH play chief roles in the regulation of whole-body metabolic homeostasis. It remains unclear how astrocytes influence neurotransmission in this region to facilitate energy and glucose balance control. Here, we explored the role of the metabotropic glutamate receptor, mGluR5, using a mouse model with selective depletion of mGluR5 from VMH astrocytes. We show that astrocytic mGluR5 critically regulates the excitatory drive and activity of PACAP-expressing neurons in the VMH to control glucose homeostasis in both female and male mice. Furthermore, mGluR5 in VMH astrocytes influences adipocyte size and sympathetic tone in white adipose tissue. These studies provide novel insight toward the importance of hypothalamic astrocytes participating in central circuits regulating peripheral metabolism.
Collapse
Affiliation(s)
- Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| | - Maribel Rios
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
28
|
Gaspari S, Labouèbe G, Picard A, Berney X, Rodriguez Sanchez‐Archidona A, Thorens B. Tmem117 in AVP neurons regulates the counterregulatory response to hypoglycemia. EMBO Rep 2023; 24:e57344. [PMID: 37314252 PMCID: PMC10398655 DOI: 10.15252/embr.202357344] [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: 04/14/2023] [Revised: 05/21/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
The counterregulatory response to hypoglycemia (CRR), which ensures a sufficient glucose supply to the brain, is an essential survival function. It is orchestrated by incompletely characterized glucose-sensing neurons, which trigger a coordinated autonomous and hormonal response that restores normoglycemia. Here, we investigate the role of hypothalamic Tmem117, identified in a genetic screen as a regulator of CRR. We show that Tmem117 is expressed in vasopressin magnocellular neurons of the hypothalamus. Tmem117 inactivation in these neurons increases hypoglycemia-induced vasopressin secretion leading to higher glucagon secretion in male mice, and this effect is estrus cycle phase dependent in female mice. Ex vivo electrophysiological analysis, in situ hybridization, and in vivo calcium imaging reveal that Tmem117 inactivation does not affect the glucose-sensing properties of vasopressin neurons but increases ER stress, ROS production, and intracellular calcium levels accompanied by increased vasopressin production and secretion. Thus, Tmem117 in vasopressin neurons is a physiological regulator of glucagon secretion, which highlights the role of these neurons in the coordinated response to hypoglycemia.
Collapse
Affiliation(s)
- Sevasti Gaspari
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Gwenaël Labouèbe
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Alexandre Picard
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Xavier Berney
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | | | - Bernard Thorens
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
29
|
Tu L, Bean JC, He Y, Liu H, Yu M, Liu H, Zhang N, Yin N, Han J, Scarcelli NA, Conde KM, Wang M, Li Y, Feng B, Gao P, Cai ZL, Fukuda M, Xue M, Tong Q, Yang Y, Liao L, Xu J, Wang C, He Y, Xu Y. Anoctamin 4 channel currents activate glucose-inhibited neurons in the mouse ventromedial hypothalamus during hypoglycemia. J Clin Invest 2023; 133:e163391. [PMID: 37261917 PMCID: PMC10348766 DOI: 10.1172/jci163391] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 05/30/2023] [Indexed: 06/03/2023] Open
Abstract
Glucose is the basic fuel essential for maintenance of viability and functionality of all cells. However, some neurons - namely, glucose-inhibited (GI) neurons - paradoxically increase their firing activity in low-glucose conditions and decrease that activity in high-glucose conditions. The ionic mechanisms mediating electric responses of GI neurons to glucose fluctuations remain unclear. Here, we showed that currents mediated by the anoctamin 4 (Ano4) channel are only detected in GI neurons in the ventromedial hypothalamic nucleus (VMH) and are functionally required for their activation in response to low glucose. Genetic disruption of the Ano4 gene in VMH neurons reduced blood glucose and impaired counterregulatory responses during hypoglycemia in mice. Activation of VMHAno4 neurons increased food intake and blood glucose, while chronic inhibition of VMHAno4 neurons ameliorated hyperglycemia in a type 1 diabetic mouse model. Finally, we showed that VMHAno4 neurons represent a unique orexigenic VMH population and transmit a positive valence, while stimulation of neurons that do not express Ano4 in the VMH (VMHnon-Ano4) suppress feeding and transmit a negative valence. Together, our results indicate that the Ano4 channel and VMHAno4 neurons are potential therapeutic targets for human diseases with abnormal feeding behavior or glucose imbalance.
Collapse
Affiliation(s)
- Longlong Tu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Jonathan C. Bean
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Yang He
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Hailan Liu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Meng Yu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Hesong Liu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Nan Zhang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Na Yin
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Junying Han
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Nikolas A. Scarcelli
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Kristine M. Conde
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Mengjie Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Yongxiang Li
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Bing Feng
- Brain glycemic and metabolism control department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Peiyu Gao
- Brain glycemic and metabolism control department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Zhao-Lin Cai
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Makoto Fukuda
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yongjie Yang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chunmei Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Yanlin He
- Brain glycemic and metabolism control department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Yong Xu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Hwang E, Portillo B, Grose K, Fujikawa T, Williams KW. Exercise-induced hypothalamic neuroplasticity: Implications for energy and glucose metabolism. Mol Metab 2023; 73:101745. [PMID: 37268247 PMCID: PMC10326746 DOI: 10.1016/j.molmet.2023.101745] [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: 11/11/2022] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Neuroplasticity refers to the brain's ability to undergo functional and structural changes in response to diverse challenges. Converging evidence supports the notion that exercise serves as a metabolic challenge, triggering the release of multiple factors both in the periphery and within the brain. These factors actively contribute to plasticity in the brain, and in turn, regulate energy and glucose metabolism. SCOPE OF REVIEW The primary focus of this review is to explore the impact of exercise-induced plasticity in the brain on metabolic homeostasis, with an emphasis on the role of the hypothalamus in this process. Additionally, the review provides an overview of various factors induced by exercise that contribute to energy balance and glucose metabolism. Notably, these factors exert their effects, at least in part, through actions within the hypothalamus and more broadly in the central nervous system. MAJOR CONCLUSIONS Exercise elicits both transient and sustained changes in metabolism, accompanied by changes in neural activity within specific brain regions. Importantly, the contribution of exercise-induced plasticity and the underlying mechanisms by which neuroplasticity influences the effects of exercise are not well understood. Recent work has begun to overcome this gap in knowledge by examining the complex interactions of exercise-induced factors which alter neural circuit properties to influence metabolism.
Collapse
Affiliation(s)
- Eunsang Hwang
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Bryan Portillo
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Teppei Fujikawa
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
31
|
Jeong S, Shin W, Park M, Lee JU, Lim Y, Noh K, Lee JH, Jun YW, Kwak M, Cheon J. Hydrogel Magnetomechanical Actuator Nanoparticles for Wireless Remote Control of Mechanosignaling In Vivo. NANO LETTERS 2023; 23:5227-5235. [PMID: 37192537 PMCID: PMC10614426 DOI: 10.1021/acs.nanolett.3c01207] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
As a new enabling nanotechnology tool for wireless, target-specific, and long-distance stimulation of mechanoreceptors in vivo, here we present a hydrogel magnetomechanical actuator (h-MMA) nanoparticle. To allow both deep-tissue penetration of input signals and efficient force generation, h-MMA integrates a two-step transduction mechanism that converts magnetic anisotropic energy to thermal energy within its magnetic core (i.e., Zn0.4Fe2.6O4 nanoparticle cluster) and then to mechanical energy to induce the surrounding polymer (i.e., pNiPMAm) shell contraction, finally delivering forces to activate targeted mechanoreceptors. We show that h-MMAs enable on-demand modulation of Notch signaling in both fluorescence reporter cell lines and a xenograft mouse model, demonstrating its utility as a powerful in vivo perturbation approach for mechanobiology interrogation in a minimally invasive and untethered manner.
Collapse
Affiliation(s)
- Sumin Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Mansoo Park
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung-uk Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Yongjun Lim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Young-wook Jun
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minsuk Kwak
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
32
|
Karatum O, Han M, Erdogan ET, Karamursel S, Nizamoglu S. Physical mechanisms of emerging neuromodulation modalities. J Neural Eng 2023; 20:031001. [PMID: 37224804 DOI: 10.1088/1741-2552/acd870] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/24/2023] [Indexed: 05/26/2023]
Abstract
One of the ultimate goals of neurostimulation field is to design materials, devices and systems that can simultaneously achieve safe, effective and tether-free operation. For that, understanding the working mechanisms and potential applicability of neurostimulation techniques is important to develop noninvasive, enhanced, and multi-modal control of neural activity. Here, we review direct and transduction-based neurostimulation techniques by discussing their interaction mechanisms with neurons via electrical, mechanical, and thermal means. We show how each technique targets modulation of specific ion channels (e.g. voltage-gated, mechanosensitive, heat-sensitive) by exploiting fundamental wave properties (e.g. interference) or engineering nanomaterial-based systems for efficient energy transduction. Overall, our review provides a detailed mechanistic understanding of neurostimulation techniques together with their applications toin vitro, in vivo, and translational studies to guide the researchers toward developing more advanced systems in terms of noninvasiveness, spatiotemporal resolution, and clinical applicability.
Collapse
Affiliation(s)
- Onuralp Karatum
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
| | - Mertcan Han
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
| | - Ezgi Tuna Erdogan
- Department of Physiology, Koc University School of Medicine, Istanbul 34450, Turkey
| | - Sacit Karamursel
- Department of Physiology, Koc University School of Medicine, Istanbul 34450, Turkey
| | - Sedat Nizamoglu
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
- Department of Biomedical Science and Engineering, Koc University, Istanbul 34450, Turkey
| |
Collapse
|
33
|
Chen X, Gong Y, Chen W. Advanced Temporally-Spatially Precise Technologies for On-Demand Neurological Disorder Intervention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207436. [PMID: 36929323 PMCID: PMC10190591 DOI: 10.1002/advs.202207436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Indexed: 05/18/2023]
Abstract
Temporal-spatial precision has attracted increasing attention for the clinical intervention of neurological disorders (NDs) to mitigate adverse effects of traditional treatments and achieve point-of-care medicine. Inspiring steps forward in this field have been witnessed in recent years, giving the credit to multi-discipline efforts from neurobiology, bioengineering, chemical materials, artificial intelligence, and so on, exhibiting valuable clinical translation potential. In this review, the latest progress in advanced temporally-spatially precise clinical intervention is highlighted, including localized parenchyma drug delivery, precise neuromodulation, as well as biological signal detection to trigger closed-loop control. Their clinical potential in both central and peripheral nervous systems is illustrated meticulously related to typical diseases. The challenges relative to biosafety and scaled production as well as their future perspectives are also discussed in detail. Notably, these intelligent temporally-spatially precision intervention systems could lead the frontier in the near future, demonstrating significant clinical value to support billions of patients plagued with NDs.
Collapse
Affiliation(s)
- Xiuli Chen
- Department of Pharmacology, School of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology430030WuhanChina
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology430030WuhanChina
| | - Yusheng Gong
- Department of Pharmacology, School of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology430030WuhanChina
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology430030WuhanChina
| | - Wei Chen
- Department of Pharmacology, School of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology430030WuhanChina
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and Technology430030WuhanChina
| |
Collapse
|
34
|
Thakkar P, Pauza AG, Murphy D, Paton JFR. Carotid body: an emerging target for cardiometabolic co-morbidities. Exp Physiol 2023; 108:661-671. [PMID: 36999224 PMCID: PMC10988524 DOI: 10.1113/ep090090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/03/2023] [Indexed: 04/01/2023]
Abstract
NEW FINDINGS What is the topic of this review? Regarding the global metabolic syndrome crisis, this review focuses on common mechanisms for high blood sugar and high blood pressure. Connections are made between the homeostatic regulation of blood pressure and blood sugar and their dysregulation to reveal signalling mechanisms converging on the carotid body. What advances does it highlight? The carotid body plays a major part in the generation of excessive sympathetic activity in diabetes and also underpins diabetic hypertension. As treatment of diabetic hypertension is notoriously difficult, we propose that novel receptors within the carotid body may provide a novel treatment strategy. ABSTRACT The maintenance of glucose homeostasis is obligatory for health and survival. It relies on peripheral glucose sensing and signalling between the brain and peripheral organs via hormonal and neural responses that restore euglycaemia. Failure of these mechanisms causes hyperglycaemia or diabetes. Current anti-diabetic medications control blood glucose but many patients remain with hyperglycemic condition. Diabetes is often associated with hypertension; the latter is more difficult to control in hyperglycaemic conditions. We ask whether a better understanding of the regulatory mechanisms of glucose control could improve treatment of both diabetes and hypertension when they co-exist. With the involvement of the carotid body (CB) in glucose sensing, metabolic regulation and control of sympathetic nerve activity, we consider the CB as a potential treatment target for both diabetes and hypertension. We provide an update on the role of the CB in glucose sensing and glucose homeostasis. Physiologically, hypoglycaemia stimulates the release of hormones such as glucagon and adrenaline, which mobilize or synthesize glucose; however, these counter-regulatory responses were markedly attenuated after denervation of the CBs in animals. Also, CB denervation prevents and reverses insulin resistance and glucose intolerance. We discuss the CB as a metabolic regulator (not just a sensor of blood gases) and consider recent evidence of novel 'metabolic' receptors within the CB and putative signalling peptides that may control glucose homeostasis via modulation of the sympathetic nervous system. The evidence presented may inform future clinical strategies in the treatment of patients with both diabetes and hypertension, which may include the CB.
Collapse
Affiliation(s)
- Pratik Thakkar
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Audrys G. Pauza
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| | - Julian F. R. Paton
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
35
|
Central Nervous System Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
36
|
Romero G, Park J, Koehler F, Pralle A, Anikeeva P. Modulating cell signalling in vivo with magnetic nanotransducers. NATURE REVIEWS. METHODS PRIMERS 2022; 2:92. [PMID: 38111858 PMCID: PMC10727510 DOI: 10.1038/s43586-022-00170-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 12/20/2023]
Abstract
Weak magnetic fields offer nearly lossless transmission of signals within biological tissue. Magnetic nanomaterials are capable of transducing magnetic fields into a range of biologically relevant signals in vitro and in vivo. These nanotransducers have recently enabled magnetic control of cellular processes, from neuronal firing and gene expression to programmed apoptosis. Effective implementation of magnetically controlled cellular signalling relies on careful tailoring of magnetic nanotransducers and magnetic fields to the responses of the intended molecular targets. This primer discusses the versatility of magnetic modulation modalities and offers practical guidelines for selection of appropriate materials and field parameters, with a particular focus on applications in neuroscience. With recent developments in magnetic instrumentation and nanoparticle chemistries, including those that are commercially available, magnetic approaches promise to empower research aimed at connecting molecular and cellular signalling to physiology and behaviour in untethered moving subjects.
Collapse
Affiliation(s)
- Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jimin Park
- Department of Materials Science and Engineering, 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
| | - Florian Koehler
- 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 Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnd Pralle
- Department of Physics, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | - Polina Anikeeva
- Department of Materials Science and Engineering, 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
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
37
|
Peters A, Sprengell M, Kubera B. The principle of 'brain energy on demand' and its predictive power for stress, sleep, stroke, obesity and diabetes. Neurosci Biobehav Rev 2022; 141:104847. [PMID: 36067964 DOI: 10.1016/j.neubiorev.2022.104847] [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: 01/08/2022] [Revised: 08/10/2022] [Accepted: 08/26/2022] [Indexed: 12/01/2022]
Abstract
Does the brain actively draw energy from the body when needed? There are different schools of thought regarding energy metabolism. In this study, the various theoretical models are classified into one of two categories: (1) conceptualizations of the brain as being purely passively supplied, which we call 'P-models,' and (2) models understanding the brain as not only passively receiving energy but also actively procuring energy for itself on demand, which we call 'A-models.' One prominent example of such theories making use of an A-model is the selfish-brain theory. The ability to make predictions was compared between the A- and P-models. A-models were able to predict and coherently explain all data examined, which included stress, sleep, caloric restriction, stroke, type-1-diabetes mellitus, obesity, and type-2-diabetes, whereas the predictions of P-models failed in most cases. The strength of the evidence supporting A-models is based on the coherence of accurate predictions across a spectrum of metabolic states. The theory test conducted here speaks to a brain that pulls its energy from the body on-demand.
Collapse
Affiliation(s)
- Achim Peters
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany.
| | - Marie Sprengell
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | - Britta Kubera
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| |
Collapse
|
38
|
Stefanov BA, Mansouri M, Charpin-El Hamri G, Fussenegger M. Sunlight-Controllable Biopharmaceutical Production for Remote Emergency Supply of Directly Injectable Therapeutic Proteins. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202566. [PMID: 36084222 DOI: 10.1002/smll.202202566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Biopharmaceutical manufacturing requires specialized facilities and a long-range cold supply chain for the delivery of the therapeutics to patients. In order to produce biopharmaceuticals in locations lacking such infrastructure, a production process is designed that utilizes the trigger-inducible release of large quantities of a stored therapeutic protein from engineered endocrine cells within minutes to generate a directly injectable saline solution of the protein. To illustrate the versatility of this approach, it is shown that not only insulin, but also glucagon-like peptide 1 (GLP-1), nanoluciferase (NLuc), and the model biopharmaceutical erythropoietin (EPO) can be trigger-inducibly released, even when using biologically inactive insulin as a carrier. The facilitating beta cells are engineered with a controllable TRPV1-mediated Ca2+ influx that induces the fusion of cytoplasmic storage vesicles with the membrane, leading to the release of the stored protein. When required, the growth medium is exchanged for saline solution, and the system is stimulated with the small molecule capsaicin, with a hand-warming pack, or simply by using sunlight. Injection of insulin saline solution obtained in this way into a type-1 diabetes mouse model results in the regulation of blood glucose levels. It is believed that this system will be readily adaptable to deliver various biopharmaceutical proteins at remote locations.
Collapse
Affiliation(s)
- Bozhidar-Adrian Stefanov
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Ghislaine Charpin-El Hamri
- Département Génie Biologique, Institut Universitaire de Technologie, Villeurbanne, Cedex F-69622, France
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
- Faculty of Life Science, University of Basel, Basel, 4058, Switzerland
| |
Collapse
|
39
|
Wang Q, Zhang B, Stutz B, Liu ZW, Horvath TL, Yang X. Ventromedial hypothalamic OGT drives adipose tissue lipolysis and curbs obesity. SCIENCE ADVANCES 2022; 8:eabn8092. [PMID: 36044565 PMCID: PMC9432828 DOI: 10.1126/sciadv.abn8092] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/14/2022] [Indexed: 05/31/2023]
Abstract
The ventromedial hypothalamus (VMH) is known to regulate body weight and counterregulatory response. However, how VMH neurons regulate lipid metabolism and energy balance remains unknown. O-linked β-d-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), catalyzed by O-GlcNAc transferase (OGT), is considered a cellular sensor of nutrients and hormones. Here, we report that genetic ablation of OGT in VMH neurons inhibits neuronal excitability. Mice with VMH neuron-specific OGT deletion show rapid weight gain, increased adiposity, and reduced energy expenditure, without significant changes in food intake or physical activity. The obesity phenotype is associated with adipocyte hypertrophy and reduced lipolysis of white adipose tissues. In addition, OGT deletion in VMH neurons down-regulates the sympathetic activity and impairs the sympathetic innervation of white adipose tissues. These findings identify OGT in the VMH as a homeostatic set point that controls body weight and underscore the importance of the VMH in regulating lipid metabolism through white adipose tissue-specific innervation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bichen Zhang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Bernardo Stutz
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L. Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaoyong Yang
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
40
|
Rawlinson S, Reichenbach A, Clarke RE, Nuñez-Iglesias J, Dempsey H, Lockie SH, Andrews ZB. In Vivo Photometry Reveals Insulin and 2-Deoxyglucose Maintain Prolonged Inhibition of VMH Vglut2 Neurons in Male Mice. Endocrinology 2022; 163:6631280. [PMID: 35788848 DOI: 10.1210/endocr/bqac095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Indexed: 11/19/2022]
Abstract
The ventromedial hypothalamic (VMH) nucleus is a well-established hub for energy and glucose homeostasis. In particular, VMH neurons are thought to be important for initiating the counterregulatory response to hypoglycemia, and ex vivo electrophysiology and immunohistochemistry data indicate a clear role for VMH neurons in sensing glucose concentration. However, the temporal response of VMH neurons to physiologically relevant changes in glucose availability in vivo has been hampered by a lack of available tools for measuring neuronal activity over time. Since the majority of neurons within the VMH are glutamatergic and can be targeted using the vesicular glutamate transporter Vglut2, we expressed cre-dependent GCaMP7s in Vglut2 cre mice and examined the response profile of VMH to intraperitoneal injections of glucose, insulin, and 2-deoxyglucose (2DG). We show that reduced available glucose via insulin-induced hypoglycemia and 2DG-induced glucoprivation, but not hyperglycemia induced by glucose injection, inhibits VMH Vglut2 neuronal population activity in vivo. Surprisingly, this inhibition was maintained for at least 45 minutes despite prolonged hypoglycemia and initiation of a counterregulatory response. Thus, although VMH stimulation, via pharmacological, electrical, or optogenetic approaches, is sufficient to drive a counterregulatory response, our data suggest VMH Vglut2 neurons are not the main drivers required to do so, since VMH Vglut2 neuronal population activity remains suppressed during hypoglycemia and glucoprivation.
Collapse
Affiliation(s)
- Sasha Rawlinson
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Alex Reichenbach
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Rachel E Clarke
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Juan Nuñez-Iglesias
- Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Harry Dempsey
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Sarah H Lockie
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
41
|
Litke R, Vicari J, Huang BT, Gonzalez D, Grimaldi N, Sharma O, Ma G, Shapiro L, Yoon Y, Kellner C, Mobbs C. Diets, genes, and drugs that increase lifespan and delay age-related diseases: Role of nutrient-sensing neurons and Creb-binding protein. Pharmacol Biochem Behav 2022; 219:173428. [PMID: 35868565 DOI: 10.1016/j.pbb.2022.173428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 06/23/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022]
Abstract
Discovery of interventions that delay or minimize age-related diseases is arguably the major goal of aging research. Conversely discovery of interventions based on phenotypic screens have often led to further elucidation of pathophysiological mechanisms. Although most hypotheses to explain lifespan focus on cell-autonomous processes, increasing evidence suggests that in multicellular organisms, neurons, particularly nutrient-sensing neurons, play a determinative role in lifespan and age-related diseases. For example, protective effects of dietary restriction and inactivation of insulin-like signaling increase lifespan and delay age-related diseases dependent on Creb-binding protein in GABA neurons, and Nrf2/Skn1 in just 2 nutrient-sensing neurons in C. elegans. Screens for drugs that increase lifespan also indicate that such drugs are predominantly active through neuronal signaling. Our own screens also indicate that neuroactive drugs also delay pathology in an animal model of Alzheimer's Disease, as well as inhibit cytokine production implicated in driving many age-related diseases. The most likely mechanism by which nutrient-sensing neurons influence lifespan and the onset of age-related diseases is by regulating metabolic architecture, particularly the relative rate of glycolysis vs. alternative metabolic pathways such as ketone and lipid metabolism. These results suggest that neuroactive compounds are a most promising class of drugs to delay or minimize age-related diseases.
Collapse
Affiliation(s)
- Rachel Litke
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America.
| | - James Vicari
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Bik Tzu Huang
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Damian Gonzalez
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Nicholas Grimaldi
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Ojee Sharma
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Gang Ma
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Lila Shapiro
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - YoneJung Yoon
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Christopher Kellner
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| | - Charles Mobbs
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, United States of America
| |
Collapse
|
42
|
Hampton RF, Jimenez-Gonzalez M, Stanley SA. Unravelling innervation of pancreatic islets. Diabetologia 2022; 65:1069-1084. [PMID: 35348820 PMCID: PMC9205575 DOI: 10.1007/s00125-022-05691-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/08/2022] [Indexed: 01/05/2023]
Abstract
The central and peripheral nervous systems play critical roles in regulating pancreatic islet function and glucose metabolism. Over the last century, in vitro and in vivo studies along with examination of human pancreas samples have revealed the structure of islet innervation, investigated the contribution of sympathetic, parasympathetic and sensory neural pathways to glucose control, and begun to determine how the structure and function of pancreatic nerves are disrupted in metabolic disease. Now, state-of-the art techniques such as 3D imaging of pancreatic innervation and targeted in vivo neuromodulation provide further insights into the anatomy and physiological roles of islet innervation. Here, we provide a summary of the published work on the anatomy of pancreatic islet innervation, its roles, and evidence for disordered islet innervation in metabolic disease. Finally, we discuss the possibilities offered by new technologies to increase our knowledge of islet innervation and its contributions to metabolic regulation.
Collapse
Affiliation(s)
- Rollie F Hampton
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
43
|
Staricoff EO, Evans ML. Recent advances in understanding hypothalamic control of defensive responses to hypoglycaemia. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 24:100353. [PMID: 39183767 PMCID: PMC11339540 DOI: 10.1016/j.coemr.2022.100353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Maintenance of normal blood glucose is important for survival. In particular, brain function is dependent on circulating glucose. In health, a series of powerful counterregulatory defences operate to prevent/limit hypoglycaemia. These defences are altered to varying degrees in diabetes and in particular, a subset of people with diabetes can develop profound deficits in these defences placing them at increased risk of suffering episodes of severe hypoglycaemia. Brain is an important controller of glucose homeostasis and developments in molecular techniques have allowed the neurocircuitry of a number of important centrally-controlled homeostatic processes such as energy balance, thirst and thermoregulation to be defined. This review describes how some of these advances have allowed a better understanding of the neuronal/brain ensembles which help protect against hypoglycaemia.
Collapse
Affiliation(s)
- Emily O. Staricoff
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, UK
| | - Mark L. Evans
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, UK
- Department of Medicine, Addenbrookes NIHR Biomedical Campus, Cambridge, UK
| |
Collapse
|
44
|
Tu L, Fukuda M, Tong Q, Xu Y. The ventromedial hypothalamic nucleus: watchdog of whole-body glucose homeostasis. Cell Biosci 2022; 12:71. [PMID: 35619170 PMCID: PMC9134642 DOI: 10.1186/s13578-022-00799-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
The brain, particularly the ventromedial hypothalamic nucleus (VMH), has been long known for its involvement in glucose sensing and whole-body glucose homeostasis. However, it is still not fully understood how the brain detects and responds to the changes in the circulating glucose levels, as well as brain-body coordinated control of glucose homeostasis. In this review, we address the growing evidence implicating the brain in glucose homeostasis, especially in the contexts of hypoglycemia and diabetes. In addition to neurons, we emphasize the potential roles played by non-neuronal cells, as well as extracellular matrix in the hypothalamus in whole-body glucose homeostasis. Further, we review the ionic mechanisms by which glucose-sensing neurons sense fluctuations of ambient glucose levels. We also introduce the significant implications of heterogeneous neurons in the VMH upon glucose sensing and whole-body glucose homeostasis, in which sex difference is also addressed. Meanwhile, research gaps have also been identified, which necessities further mechanistic studies in future.
Collapse
Affiliation(s)
- Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Makoto Fukuda
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
45
|
Ma H, He C, Li L, Gao P, Lu Z, Hu Y, Wang L, Zhao Y, Cao T, Cui Y, Zheng H, Yang G, Yan Z, Liu D, Zhu Z. TRPC5 deletion in the central amygdala antagonizes high-fat diet-induced obesity by increasing sympathetic innervation. Int J Obes (Lond) 2022; 46:1544-1555. [PMID: 35589963 DOI: 10.1038/s41366-022-01151-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/09/2022]
Abstract
Transient receptor potential channel 5 (TRPC5) is predominantly distributed in the brain, especially in the central amygdala (CeA), which is closely associated with pain and addiction. Although mounting evidence indicates that the CeA is related to energy homeostasis, the possible regulatory effect of TRPC5 in the CeA on metabolism remains unclear. Here, we reported that the expression of TRPC5 in the CeA of mice was increased under a high-fat diet (HFD). Specifically, the deleted TRPC5 protein in the CeA of mice using adeno-associated virus resisted HFD-induced weight gain, accompanied by increased food intake. Furthermore, the energy expenditure of CeA-specific TRPC5 deletion mice (TRPC5 KO) was elevated due to augmented white adipose tissue (WAT) browning and brown adipose tissue (BAT) activity. Mechanistically, deficiency of TRPC5 in the CeA boosted nonshivering thermogenesis under cold stimulation by stimulating sympathetic nerves, as the β3-adrenoceptor (Adrb3) antagonist SR59230A blocked the effect of TRPC5 KO on this process. In summary, TRPC5 deletion in the CeA alleviated the metabolic deterioration of mice fed a HFD, and these phenotypic improvements were correlated with the increased sympathetic distribution and activity of adipose tissue.
Collapse
Affiliation(s)
- Huan Ma
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Chengkang He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Yingru Hu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Tingbing Cao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Yuanting Cui
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China. .,Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| |
Collapse
|
46
|
Zou L, Xu K, Tian H, Fang Y. Remote neural regulation mediated by nanomaterials. NANOTECHNOLOGY 2022; 33:272002. [PMID: 35442216 DOI: 10.1088/1361-6528/ac62b1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Neural regulation techniques play an essential role in the functional dissection of neural circuits and also the treatment of neurological diseases. Recently, a series of nanomaterials, including upconversion nanoparticles (UCNPs), magnetic nanoparticles (MNPs), and silicon nanomaterials (SNMs) that are responsive to remote optical or magnetic stimulation, have been applied as transducers to facilitate localized control of neural activities. In this review, we summarize the latest advances in nanomaterial-mediated neural regulation, especially in a remote and minimally invasive manner. We first give an overview of existing neural stimulation techniques, including electrical stimulation, transcranial magnetic stimulation, chemogenetics, and optogenetics, with an emphasis on their current limitations. Then we focus on recent developments in nanomaterial-mediated neural regulation, including UCNP-mediated fiberless optogenetics, MNP-mediated magnetic neural regulation, and SNM-mediated non-genetic neural regulation. Finally, we discuss the possibilities and challenges for nanomaterial-mediated neural regulation.
Collapse
Affiliation(s)
- Liang Zou
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ke Xu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
47
|
Del Sol-Fernández S, Martínez-Vicente P, Gomollón-Zueco P, Castro-Hinojosa C, Gutiérrez L, Fratila RM, Moros M. Magnetogenetics: remote activation of cellular functions triggered by magnetic switches. NANOSCALE 2022; 14:2091-2118. [PMID: 35103278 PMCID: PMC8830762 DOI: 10.1039/d1nr06303k] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/13/2021] [Indexed: 05/03/2023]
Abstract
During the last decade, the possibility to remotely control intracellular pathways using physical tools has opened the way to novel and exciting applications, both in basic research and clinical applications. Indeed, the use of physical and non-invasive stimuli such as light, electricity or magnetic fields offers the possibility of manipulating biological processes with spatial and temporal resolution in a remote fashion. The use of magnetic fields is especially appealing for in vivo applications because they can penetrate deep into tissues, as opposed to light. In combination with magnetic actuators they are emerging as a new instrument to precisely manipulate biological functions. This approach, coined as magnetogenetics, provides an exclusive tool to study how cells transform mechanical stimuli into biochemical signalling and offers the possibility of activating intracellular pathways connected to temperature-sensitive proteins. In this review we provide a critical overview of the recent developments in the field of magnetogenetics. We discuss general topics regarding the three main components for magnetic field-based actuation: the magnetic fields, the magnetic actuators and the cellular targets. We first introduce the main approaches in which the magnetic field can be used to manipulate the magnetic actuators, together with the most commonly used magnetic field configurations and the physicochemical parameters that can critically influence the magnetic properties of the actuators. Thereafter, we discuss relevant examples of magneto-mechanical and magneto-thermal stimulation, used to control stem cell fate, to activate neuronal functions, or to stimulate apoptotic pathways, among others. Finally, although magnetogenetics has raised high expectations from the research community, to date there are still many obstacles to be overcome in order for it to become a real alternative to optogenetics for instance. We discuss some controversial aspects related to the insufficient elucidation of the mechanisms of action of some magnetogenetics constructs and approaches, providing our opinion on important challenges in the field and possible directions for the upcoming years.
Collapse
Affiliation(s)
- Susel Del Sol-Fernández
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pablo Martínez-Vicente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pilar Gomollón-Zueco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Christian Castro-Hinojosa
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Lucía Gutiérrez
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Analítica, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Orgánica, Universidad de Zaragoza, C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
48
|
Papazoglou I, Lee JH, Cui Z, Li C, Fulgenzi G, Bahn YJ, Staniszewska-Goraczniak HM, Piñol RA, Hogue IB, Enquist LW, Krashes MJ, Rane SG. A distinct hypothalamus-to-β cell circuit modulates insulin secretion. Cell Metab 2022; 34:285-298.e7. [PMID: 35108515 PMCID: PMC8935365 DOI: 10.1016/j.cmet.2021.12.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/01/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
The central nervous system has long been thought to regulate insulin secretion, an essential process in the maintenance of blood glucose levels. However, the anatomical and functional connections between the brain and insulin-producing pancreatic β cells remain undefined. Here, we describe a functional transneuronal circuit connecting the hypothalamus to β cells in mice. This circuit originates from a subpopulation of oxytocin neurons in the paraventricular hypothalamic nucleus (PVNOXT), and it reaches the islets of the endocrine pancreas via the sympathetic autonomic branch to innervate β cells. Stimulation of PVNOXT neurons rapidly suppresses insulin secretion and causes hyperglycemia. Conversely, silencing of these neurons elevates insulin levels by dysregulating neuronal signaling and secretory pathways in β cells and induces hypoglycemia. PVNOXT neuronal activity is triggered by glucoprivation. Our findings reveal that a subset of PVNOXT neurons form functional multisynaptic circuits with β cells in mice to regulate insulin secretion, and their function is necessary for the β cell response to hypoglycemia.
Collapse
Affiliation(s)
- Ioannis Papazoglou
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA.
| | - Ji-Hyeon Lee
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Zhenzhong Cui
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Chia Li
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Gianluca Fulgenzi
- Neural Development Section, MCGP, CCR, NCI, NIH, Frederick, MD, USA; Department of Molecular and Clinical Sciences, Marche Polytechnic University, Ancona, Italy
| | - Young Jae Bahn
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | | | - Ramón A Piñol
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Ian B Hogue
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Lynn W Enquist
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michael J Krashes
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Sushil G Rane
- Diabetes, Endocrinology and Obesity Branch, NIDDK, NIH, Bethesda, MD, USA.
| |
Collapse
|
49
|
Yu Y, Payne C, Marina N, Korsak A, Southern P, García‐Prieto A, Christie IN, Baker RR, Fisher EMC, Wells JA, Kalber TL, Pankhurst QA, Gourine AV, Lythgoe MF. Remote and Selective Control of Astrocytes by Magnetomechanical Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104194. [PMID: 34927381 PMCID: PMC8867145 DOI: 10.1002/advs.202104194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/15/2021] [Indexed: 05/06/2023]
Abstract
Astrocytes play crucial and diverse roles in brain health and disease. The ability to selectively control astrocytes provides a valuable tool for understanding their function and has the therapeutic potential to correct dysfunction. Existing technologies such as optogenetics and chemogenetics require the introduction of foreign proteins, which adds a layer of complication and hinders their clinical translation. A novel technique, magnetomechanical stimulation (MMS), that enables remote and selective control of astrocytes without genetic modification is described here. MMS exploits the mechanosensitivity of astrocytes and triggers mechanogated Ca2+ and adenosine triphosphate (ATP) signaling by applying a magnetic field to antibody-functionalized magnetic particles that are targeted to astrocytes. Using purpose-built magnetic devices, the mechanosensory threshold of astrocytes is determined, a sub-micrometer particle for effective MMS is identified, the in vivo fate of the particles is established, and cardiovascular responses are induced in rats after particles are delivered to specific brainstem astrocytes. By eliminating the need for device implantation and genetic modification, MMS is a method for controlling astroglial activity with an improved prospect for clinical application than existing technologies.
Collapse
Affiliation(s)
- Yichao Yu
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Christopher Payne
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Alla Korsak
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Paul Southern
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
| | - Ana García‐Prieto
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Departamento Física Aplicada IUniversidad del País VascoBilbao48013Spain
| | - Isabel N. Christie
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Rebecca R. Baker
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Elizabeth M. C. Fisher
- Department of Neuromuscular DiseasesQueen Square Institute of NeurologyUniversity College LondonQueen SquareLondonWC1N 3BGUK
| | - Jack A. Wells
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Tammy L. Kalber
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Quentin A. Pankhurst
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
| | - Alexander V. Gourine
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Mark F. Lythgoe
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| |
Collapse
|
50
|
Liu X, Qiu F, Hou L, Wang X. Review of Noninvasive or Minimally Invasive Deep Brain Stimulation. Front Behav Neurosci 2022; 15:820017. [PMID: 35145384 PMCID: PMC8823253 DOI: 10.3389/fnbeh.2021.820017] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Brain stimulation is a critical technique in neuroscience research and clinical application. Traditional transcranial brain stimulation techniques, such as transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and deep brain stimulation (DBS) have been widely investigated in neuroscience for decades. However, TMS and tDCS have poor spatial resolution and penetration depth, and DBS requires electrode implantation in deep brain structures. These disadvantages have limited the clinical applications of these techniques. Owing to developments in science and technology, substantial advances in noninvasive and precise deep stimulation have been achieved by neuromodulation studies. Second-generation brain stimulation techniques that mainly rely on acoustic, electronic, optical, and magnetic signals, such as focused ultrasound, temporal interference, near-infrared optogenetic, and nanomaterial-enabled magnetic stimulation, offer great prospects for neuromodulation. This review summarized the mechanisms, development, applications, and strengths of these techniques and the prospects and challenges in their development. We believe that these second-generation brain stimulation techniques pave the way for brain disorder therapy.
Collapse
Affiliation(s)
- Xiaodong Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Fang Qiu
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lijuan Hou
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
- *Correspondence: Lijuan Hou Xiaohui Wang
| | - Xiaohui Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Lijuan Hou Xiaohui Wang
| |
Collapse
|