1
|
Ben-Simon Y, Hooper M, Narayan S, Daigle TL, Dwivedi D, Way SW, Oster A, Stafford DA, Mich JK, Taormina MJ, Martinez RA, Opitz-Araya X, Roth JR, Alexander JR, Allen S, Amster A, Arbuckle J, Ayala A, Baker PM, Bakken TE, Barcelli T, Barta S, Bendrick J, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chance RK, Chavan S, Clark M, Colbert K, Collman F, Daniel S, Departee M, DiValentin P, Donadio N, Dotson N, Egdorf T, Fliss T, Gabitto M, Garcia J, Gary A, Gasperini M, Gloe J, Goldy J, Gore BB, Graybuck L, Greisman N, Haeseleer F, Halterman C, Haradon Z, Hastings SD, Helback O, Ho W, Hockemeyer D, Huang C, Huff S, Hunker A, Johansen N, Jones D, Juneau Z, Kalmbach B, Kannan M, Khem S, Kussick E, Kutsal R, Larsen R, Lee C, Lee AY, Leibly M, Lenz GH, Li S, Liang E, Lusk N, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mollenkopf T, Moosman S, Morin E, Newman D, Ng L, Ngo K, Omstead V, Otto S, Oyama A, Pena N, Pham T, Phillips E, Pom CA, Potekhina L, Ransford S, et alBen-Simon Y, Hooper M, Narayan S, Daigle TL, Dwivedi D, Way SW, Oster A, Stafford DA, Mich JK, Taormina MJ, Martinez RA, Opitz-Araya X, Roth JR, Alexander JR, Allen S, Amster A, Arbuckle J, Ayala A, Baker PM, Bakken TE, Barcelli T, Barta S, Bendrick J, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chance RK, Chavan S, Clark M, Colbert K, Collman F, Daniel S, Departee M, DiValentin P, Donadio N, Dotson N, Egdorf T, Fliss T, Gabitto M, Garcia J, Gary A, Gasperini M, Gloe J, Goldy J, Gore BB, Graybuck L, Greisman N, Haeseleer F, Halterman C, Haradon Z, Hastings SD, Helback O, Ho W, Hockemeyer D, Huang C, Huff S, Hunker A, Johansen N, Jones D, Juneau Z, Kalmbach B, Kannan M, Khem S, Kussick E, Kutsal R, Larsen R, Lee C, Lee AY, Leibly M, Lenz GH, Li S, Liang E, Lusk N, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mollenkopf T, Moosman S, Morin E, Newman D, Ng L, Ngo K, Omstead V, Otto S, Oyama A, Pena N, Pham T, Phillips E, Pom CA, Potekhina L, Ransford S, Ray PL, Rette D, Reynoldson C, Rimorin C, Rocha D, Ruiz A, Sanchez REA, Sawyer L, Sedeno-Cortes A, Sevigny JP, Shapovalova N, Shepard N, Shulga L, Sigler AR, Siverts L, Soliman S, Somasundaram S, Staats B, Stewart K, Szelenyi E, Tieu M, Trader C, Tran A, van Velthoven CTJ, Walker M, Wang Y, Weed N, Wirthlin M, Wood T, Wynalda B, Yao Z, Zhou T, Ariza J, Dee N, Reding M, Ronellenfitch K, Mufti S, Sunkin SM, Smith KA, Esposito L, Waters J, Thyagarajan B, Yao S, Lein ES, Zeng H, Levi BP, Ngai J, Ting JT, Tasic B. A suite of enhancer AAVs and transgenic mouse lines for genetic access to cortical cell types. Cell 2025; 188:3045-3064.e23. [PMID: 40403729 DOI: 10.1016/j.cell.2025.05.002] [Show More Authors] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/25/2025] [Accepted: 05/01/2025] [Indexed: 05/24/2025]
Abstract
The mammalian cortex is comprised of cells classified into types according to shared properties. Defining the contribution of each cell type to the processes guided by the cortex is essential for understanding its function in health and disease. We use transcriptomic and epigenomic cortical cell-type taxonomies from mouse and human to define marker genes and putative enhancers and create a large toolkit of transgenic lines and enhancer adeno-associated viruses (AAVs) for selective targeting of cortical cell populations. We report creation and evaluation of fifteen transgenic driver lines, two reporter lines, and >1,000 different enhancer AAV vectors covering most subclasses of cortical cells. The tools reported here have been made publicly available, and along with the scaled process of tool creation, evaluation, and modification, they will enable diverse experimental strategies toward understanding mammalian cortex and brain function.
Collapse
Affiliation(s)
- Yoav Ben-Simon
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Marcus Hooper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Sharon W Way
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Aaron Oster
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Jada R Roth
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Shona Allen
- University of California, Berkeley, Berkeley, CA 94720, USA
| | - Adam Amster
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Joel Arbuckle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Angela Ayala
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Pamela M Baker
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Tyler Barcelli
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Stuard Barta
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | | | - Jessica Bowlus
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Krissy Brouner
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brittny Casian
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Anish B Chakka
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Sakshi Chavan
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Clark
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kaity Colbert
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tim Fliss
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Jazmin Garcia
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lucas Graybuck
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Noah Greisman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Zeb Haradon
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Olivia Helback
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Sydney Huff
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Avery Hunker
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Danielle Jones
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zoe Juneau
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Madhav Kannan
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shannon Khem
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Emily Kussick
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rana Kutsal
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachael Larsen
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Angus Y Lee
- University of California, Berkeley, Berkeley, CA 94720, USA
| | - Madison Leibly
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Garreck H Lenz
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Su Li
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Nicholas Lusk
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Jessica Malloy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jocelin Malone
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jose Melchor
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Skyler Moosman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Elyse Morin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Dakota Newman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Sven Otto
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Alana Oyama
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nick Pena
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | | | - Shea Ransford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Patrick L Ray
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Dean Rette
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Dana Rocha
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Augustin Ruiz
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Noah Shepard
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ana R Sigler
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Sherif Soliman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kaiya Stewart
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Eric Szelenyi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Cameron Trader
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Alex Tran
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Miranda Walker
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Yimin Wang
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Toren Wood
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brooke Wynalda
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeanelle Ariza
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Melissa Reding
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Susan M Sunkin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Luke Esposito
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - John Ngai
- University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
2
|
He J, Phan BN, Kerkhoff WG, Alikaya A, Hong T, Brull OR, Fredericks JM, Sedorovitz M, Srinivasan C, Leone MJ, Wirfel OM, Brown A, Dauby S, Tittle RK, Lin MK, Hooks BM, Bostan AC, Gharbawie OA, Byrne LC, Pfenning AR, Stauffer WR. Machine learning identification of enhancers in the rhesus macaque genome. Neuron 2025; 113:1548-1561.e8. [PMID: 40403706 DOI: 10.1016/j.neuron.2025.04.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/28/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025]
Abstract
Nonhuman primate (NHP) neuroanatomy and cognitive complexity make NHPs ideal models to study human neurobiology and disease. However, NHP circuit-function investigations are limited by the availability of molecular reagents that are effective in NHPs. This calls for reagent development approaches that prioritize NHPs. Therefore, we derived enhancers from the NHP genome. We defined cell-type-specific open chromatin regions (OCRs) in single-cell data from rhesus macaques. We trained machine-learning models to rank those OCRs according to their potential as cell-type-specific enhancers for cells in the dorsolateral prefrontal cortex (DLPFC). We packaged the top-ranked layer-3-pyramidal-neuron enhancer into AAV and injected it into the macaque DLPFC. Expression was mostly restricted to layers 2 and 3 and confirmed with light-driven activation of channelrhodopsin. These results provide a crucial tool for studying the causal functions of DLPFC and provide a roadmap for optimized gene delivery in primates.
Collapse
Affiliation(s)
- Jing He
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - BaDoi N Phan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA; University of Pittsburgh-Carnegie Mellon University Medical Scientist Training Program, Pittsburgh, PA 15213, USA
| | - Willa G Kerkhoff
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Aydin Alikaya
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tao Hong
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Program in Neural Computation, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Olivia R Brull
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - J Megan Fredericks
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Morgan Sedorovitz
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Chaitanya Srinivasan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Michael J Leone
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA; University of Pittsburgh-Carnegie Mellon University Medical Scientist Training Program, Pittsburgh, PA 15213, USA
| | - Olivia M Wirfel
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ashley Brown
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Samuel Dauby
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rachel K Tittle
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Meng K Lin
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Andreea C Bostan
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Omar A Gharbawie
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Andreas R Pfenning
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | - William R Stauffer
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
3
|
Hunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, Ayala A, Baker PM, Barcelli T, Barta S, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chong P, Clark M, Colbert K, Daniel S, Dawe T, Departee M, DiValentin P, Donadio NP, Dotson NI, Dwivedi D, Egdorf T, Fliss T, Gary A, Goldy J, Grasso C, Groce EL, Gudsnuk K, Han W, Haradon Z, Hastings S, Helback O, Ho WV, Huang C, Johnson T, Jones DL, Juneau Z, Kenney J, Leibly M, Li S, Liang E, Loeffler H, Lusk NA, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mich JK, Moosman S, Morin E, Naidoo R, Newman D, Ngo K, Nguyen K, Oster AL, Ouellette B, Oyama AA, Pena N, Pham T, Phillips E, Pom C, Potekhina L, Ransford S, Ray PL, Reding M, Rette DF, Reynoldson C, Rimorin C, Sigler AR, Rocha DB, Ronellenfitch K, et alHunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, Ayala A, Baker PM, Barcelli T, Barta S, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chong P, Clark M, Colbert K, Daniel S, Dawe T, Departee M, DiValentin P, Donadio NP, Dotson NI, Dwivedi D, Egdorf T, Fliss T, Gary A, Goldy J, Grasso C, Groce EL, Gudsnuk K, Han W, Haradon Z, Hastings S, Helback O, Ho WV, Huang C, Johnson T, Jones DL, Juneau Z, Kenney J, Leibly M, Li S, Liang E, Loeffler H, Lusk NA, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mich JK, Moosman S, Morin E, Naidoo R, Newman D, Ngo K, Nguyen K, Oster AL, Ouellette B, Oyama AA, Pena N, Pham T, Phillips E, Pom C, Potekhina L, Ransford S, Ray PL, Reding M, Rette DF, Reynoldson C, Rimorin C, Sigler AR, Rocha DB, Ronellenfitch K, Ruiz A, Sawyer L, Sevigny JP, Shapovalova NV, Shepard N, Shulga L, Soliman S, Staats B, Taormina MJ, Tieu M, Wang Y, Wilkes J, Wood T, Zhou T, Williford A, Dee N, Mollenkopf T, Ng L, Esposito L, Kalmbach BE, Yao S, Ariza J, Collman F, Mufti S, Smith K, Waters J, Ersing I, Patrick M, Zeng H, Lein ES, Kojima Y, Horwitz G, Owen SF, Levi BP, Daigle TL, Tasic B, Bakken TE, Ting JT. Enhancer AAV toolbox for accessing and perturbing striatal cell types and circuits. Neuron 2025; 113:1507-1524.e17. [PMID: 40403704 DOI: 10.1016/j.neuron.2025.04.035] [Show More Authors] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/17/2025] [Accepted: 04/30/2025] [Indexed: 05/24/2025]
Abstract
We present an enhancer-AAV toolbox for accessing and perturbing striatal cell types and circuits. Best-in-class vectors were curated for accessing major striatal neuron populations including medium spiny neurons (MSNs), direct- and indirect-pathway MSNs, Sst-Chodl, Pvalb-Pthlh, and cholinergic interneurons. Specificity was evaluated by multiple modes of molecular validation, by three different routes of virus delivery, and with diverse transgene cargos. Importantly, we provide detailed information necessary to achieve reliable cell-type-specific labeling under different experimental contexts. We demonstrate direct pathway circuit-selective optogenetic perturbation of behavior and multiplex labeling of striatal interneuron types for targeted analysis of cellular features. Lastly, we show conserved in vivo activity for exemplary MSN enhancers in rats and macaques. This collection of striatal enhancer AAVs offers greater versatility compared to available transgenic lines and can readily be applied for cell type and circuit studies in diverse mammalian species beyond the mouse model.
Collapse
Affiliation(s)
| | | | - Gursajan Gill
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Jacqueline L Bendrick
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Sharon W Way
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Sven Otto
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Avalon Amaya
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Adam Amster
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Angela Ayala
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Pam M Baker
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Stuard Barta
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | | - Peter Chong
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dawe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Fliss
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Conor Grasso
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Erin L Groce
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Warren Han
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Zeb Haradon
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Sam Hastings
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Windy V Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tye Johnson
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Zoe Juneau
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jaimie Kenney
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Su Li
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jose Melchor
- Allen Institute for Brain Science, Seattle, WA, USA
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Elyse Morin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Robyn Naidoo
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Ben Ouellette
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Nick Pena
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | | | - Dean F Rette
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Dana B Rocha
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Noah Shepard
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yimin Wang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Josh Wilkes
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Toren Wood
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ali Williford
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Brian E Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA; Department of Neurobiology & Biophysics, University of Washington, Seattle, WA, USA
| | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA; Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Yoshiko Kojima
- Department of Otolaryngology, Head and Neck Surgery, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, Seattle, WA, USA
| | - Greg Horwitz
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, Seattle, WA, USA
| | - Scott F Owen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA, USA; Department of Neurobiology & Biophysics, University of Washington, Seattle, WA, USA
| | | | | | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA; Department of Neurobiology & Biophysics, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, Seattle, WA, USA.
| |
Collapse
|
4
|
Furlanis E, Dai M, Leyva Garcia B, Tran T, Vergara J, Pereira A, Gorissen BL, Wills S, Vlachos A, Hairston A, Dwivedi D, Du S, McMahon J, Huang S, Morabito A, Vazquez A, Kim S, Lee AT, Chang EF, Razzaq T, Qazi A, Vargish G, Yuan X, Caccavano A, Hunt S, Chittajallu R, McLean N, Hewitt L, Paranzino E, Rice H, Cummins AC, Plotnikova A, Mohanty A, Tangen AC, Shin JH, Azadi R, Eldridge MAG, Alvarez VA, Averbeck BB, Alyahyay M, Vallejo TR, Soheib M, Vattino LG, MacGregor CP, Chatain CP, Banks E, Olah VJ, Naskar S, Hill S, Liebergall S, Badiani R, Hyde L, Hanley E, Xu Q, Allaway KC, Goldberg EM, Rowan MJM, Nowakowski TJ, Lee S, Favuzzi E, Kaeser PS, Sjulson L, Batista-Brito R, Takesian AE, Ibrahim LA, Iqbal A, Pelkey KA, McBain CJ, Dimidschstein J, Fishell G, Wang Y. An enhancer-AAV toolbox to target and manipulate distinct interneuron subtypes. Neuron 2025; 113:1525-1547.e15. [PMID: 40403705 DOI: 10.1016/j.neuron.2025.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/18/2025] [Accepted: 05/01/2025] [Indexed: 05/24/2025]
Abstract
In recent years, we and others have identified a number of enhancers that, when incorporated into rAAV vectors, can restrict the transgene expression to particular neuronal populations. Yet, viral tools to access and manipulate specific neuronal subtypes are still limited. Here, we performed systematic analysis of single-cell genomic data to identify enhancer candidates for each of the telencephalic interneuron subtypes. We established a set of enhancer-AAV tools that are highly specific for distinct cortical interneuron populations and striatal cholinergic interneurons. These enhancers, when used in the context of different effectors, can target (fluorescent proteins), observe activity (GCaMP), and manipulate (opto-genetics) specific neuronal subtypes. We also validated our enhancer-AAV tools across species. Thus, we provide the field with a powerful set of tools to study neural circuits and functions and to develop precise and targeted therapy.
Collapse
Affiliation(s)
- Elisabetta Furlanis
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Min Dai
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brenda Leyva Garcia
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thien Tran
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Josselyn Vergara
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ana Pereira
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bram L Gorissen
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sara Wills
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ariel Hairston
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Deepanjali Dwivedi
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah Du
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Justin McMahon
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuhan Huang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Annunziato Morabito
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Arenski Vazquez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Soyoun Kim
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anthony T Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Adam Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Lauren Hewitt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Emily Paranzino
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Haley Rice
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Anya Plotnikova
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Arya Mohanty
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Anne Claire Tangen
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Jung Hoon Shin
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Reza Azadi
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Mark A G Eldridge
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Veronica A Alvarez
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Bruno B Averbeck
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-4415, USA
| | - Mansour Alyahyay
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Tania Reyes Vallejo
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Mohammed Soheib
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Lucas G Vattino
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Cathryn P MacGregor
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Carolina Piletti Chatain
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
| | - Emmie Banks
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Viktor Janos Olah
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shovan Naskar
- Unit of Functional Neural Circuit, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophie Hill
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sophie Liebergall
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rohan Badiani
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lili Hyde
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ella Hanley
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qing Xu
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia; Genomics & Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Kathryn C Allaway
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ethan M Goldberg
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew J M Rowan
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Soohyun Lee
- Unit of Functional Neural Circuit, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emilia Favuzzi
- Department of Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
| | - Pascal S Kaeser
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Lucas Sjulson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Renata Batista-Brito
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anne E Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Leena A Ibrahim
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | | | - Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Yating Wang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
5
|
Kussick E, Johansen N, Taskin N, Chowdhury A, Quinlan MA, Fraser A, Clark AG, Wynalda B, Martinez R, Groce EL, Reding M, Liang E, Shulga L, Huang C, Casper T, Clark M, Ho W, Gao Y, van Velthoven CTJ, Sobieski C, Ferrer R, Berg MR, Curtis BC, English C, Day JC, Fortuna MG, Donadio N, Newman D, Yao S, Chakka AB, Goldy J, Torkelson A, Guzman JB, Chakrabarty R, Nguy B, Guilford N, Pham TH, Wright V, Ronellenfitch K, Naidoo R, Kenney J, Williford A, Ramakrishnan C, Drinnenberg A, Gudsnuk K, Thyagarajan B, Smith KA, Dee N, Deisseroth K, Zeng H, Yao Z, Tasic B, Levi BP, Hodge R, Bakken TE, Lein ES, Ting JT, Daigle TL. Enhancer AAVs for targeting spinal motor neurons and descending motor pathways in rodents and macaque. Cell Rep 2025:115730. [PMID: 40403722 DOI: 10.1016/j.celrep.2025.115730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/24/2025] Open
Abstract
Experimental access to cell types within the mammalian spinal cord is severely limited by the availability of genetic tools. To enable access to spinal motor neurons (SMNs) and SMN subtypes, we generated single-cell multiome datasets from mouse and macaque spinal cords and discovered putative enhancers for each neuronal population. We cloned these enhancers into adeno-associated viral vectors driving a reporter fluorophore and functionally screened them in the mouse. We extensively characterized the most promising candidate enhancers in rat and macaque and developed an optimized pan-SMN enhancer virus. Additionally, we generated derivative viruses expressing iCre297T recombinase or ChR2-EYFP for labeling and functional studies, and we created a single vector with combined enhancer elements to achieve simultaneous labeling of layer 5 extratelencephalic projecting neurons and SMNs. This unprecedented SMN toolkit will enable future investigations of cell type function across species and potential therapeutic interventions for human neurodegenerative diseases.
Collapse
Affiliation(s)
- Emily Kussick
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Alex Fraser
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Andrew G Clark
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brooke Wynalda
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Erin L Groce
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Melissa Reding
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Clark
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Rebecca Ferrer
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Melissa R Berg
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Britni C Curtis
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Chris English
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Jesse C Day
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | | | | | - Dakota Newman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Amy Torkelson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Beagen Nguy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Vonn Wright
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Robyn Naidoo
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jaimie Kenney
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ali Williford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305 USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Washington National Primate Research Center, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
6
|
Johansen NJ, Kempynck N, Zemke NR, Somasundaram S, De Winter S, Hooper M, Dwivedi D, Lohia R, Wehbe F, Li B, Abaffyová D, Armand EJ, De Man J, Ekşi EC, Hecker N, Hulselmans G, Konstantakos V, Mauduit D, Mich JK, Partel G, Daigle TL, Levi BP, Zhang K, Tanaka Y, Gillis J, Ting JT, Ben-Simon Y, Miller J, Ecker JR, Ren B, Aerts S, Lein ES, Tasic B, Bakken TE. Evaluating methods for the prediction of cell-type-specific enhancers in the mammalian cortex. CELL GENOMICS 2025:100879. [PMID: 40403730 DOI: 10.1016/j.xgen.2025.100879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/03/2025] [Accepted: 04/17/2025] [Indexed: 05/24/2025]
Abstract
Identifying cell-type-specific enhancers is critical for developing genetic tools to study the mammalian brain. We organized the "Brain Initiative Cell Census Network (BICCN) Challenge: Predicting Functional Cell Type-Specific Enhancers from Cross-Species Multi-Omics" to evaluate machine learning and feature-based methods for nominating enhancer sequences targeting mouse cortical cell types. Methods were assessed using in vivo data from hundreds of adeno-associated virus (AAV)-packaged, retro-orbitally delivered enhancers. Open chromatin was the strongest predictor of functional enhancers, while sequence models improved prediction of non-functional enhancers and identified cell-type-specific transcription factor codes to inform in silico enhancer design. This challenge establishes a benchmark for enhancer prioritization and highlights computational and molecular features critical for identifying functional cortical enhancers, advancing efforts to map and manipulate gene regulation in the mammalian cortex.
Collapse
Affiliation(s)
| | - Niklas Kempynck
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Nathan R Zemke
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Seppe De Winter
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Marcus Hooper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ruchi Lohia
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Fabien Wehbe
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Bocheng Li
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Darina Abaffyová
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Ethan J Armand
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Julie De Man
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Eren Can Ekşi
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Nikolai Hecker
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Gert Hulselmans
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Vasilis Konstantakos
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - David Mauduit
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Gabriele Partel
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kai Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yoshiaki Tanaka
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Jesse Gillis
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Yoav Ben-Simon
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeremy Miller
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Joseph R Ecker
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bing Ren
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stein Aerts
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | |
Collapse
|
7
|
Matuszek Z, Brown BL, Yrigollen CM, Keiser MS, Davidson BL. Current trends in gene therapy to treat inherited disorders of the brain. Mol Ther 2025; 33:1988-2014. [PMID: 40181540 DOI: 10.1016/j.ymthe.2025.03.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
Gene therapy development, re-engineering, and application to patients hold promise to revolutionize medicine, including therapies for disorders of the brain. Advances in delivery modalities, expression regulation, and improving safety profiles are of critical importance. Additionally, each inherited disorder has its own unique characteristics as to regions and cell types impacted and the temporal dynamics of that impact that are essential for the design of therapeutic design strategies. Here, we review the current state of the art in gene therapies for inherited brain disorders, summarizing key considerations for vector delivery, gene addition, gene silencing, gene editing, and epigenetic editing. We provide examples from animal models, human cell lines, and, where possible, clinical trials. This review also highlights the various tools available to researchers for basic research questions and discusses our views on the current limitations in the field.
Collapse
Affiliation(s)
- Zaneta Matuszek
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Brandon L Brown
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carolyn M Yrigollen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Megan S Keiser
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Chai H, Huang X, Xiong G, Huang J, Pels KK, Meng L, Han J, Tang D, Pan G, Deng L, Xiao Q, Wang X, Zhang M, Banecki K, Plewczynski D, Wei CL, Ruan Y. Tri-omic single-cell mapping of the 3D epigenome and transcriptome in whole mouse brains throughout the lifespan. Nat Methods 2025; 22:994-1007. [PMID: 40301621 DOI: 10.1038/s41592-025-02658-7] [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: 11/14/2024] [Accepted: 03/13/2025] [Indexed: 05/01/2025]
Abstract
Exploring the genomic basis of transcriptional programs has been a long-standing research focus. Here we report a single-cell method, ChAIR, to map chromatin accessibility, chromatin interactions and RNA expression simultaneously. After validating in cultured cells, we applied ChAIR to whole mouse brains and delineated the concerted dynamics of epigenome, three-dimensional (3D) genome and transcriptome during maturation and aging. In particular, gene-centric chromatin interactions and open chromatin states provided 3D epigenomic mechanism underlying cell-type-specific transcription and revealed spatially resolved specificity. Importantly, the composition of short-range and ultralong chromatin contacts in individual cells is remarkably correlated with transcriptional activity, open chromatin state and genome folding density. This genomic property, along with associated cellular properties, differs in neurons and non-neuronal cells across different anatomic regions throughout the lifespan, implying divergent nuclear mechano-genomic mechanisms at play in brain cells. Our results demonstrate ChAIR's robustness in revealing single-cell 3D epigenomic states of cell-type-specific transcription in complex tissues.
Collapse
Affiliation(s)
- Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xingyu Huang
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Guangzhou Xiong
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jiaxiang Huang
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Katarzyna Karolina Pels
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Lingyun Meng
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jin Han
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Dongmei Tang
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Guanjing Pan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Liang Deng
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Qin Xiao
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xiaotao Wang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Shanghai Key Laboratory of Reproduction and Development, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Krzysztof Banecki
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Chia-Lin Wei
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Furlanis E, Dai M, Garcia BL, Tran T, Vergara J, Pereira A, Gorissen BL, Wills S, Vlachos A, Hairston A, Dwivedi D, Du S, McMahon J, Huang S, Morabito A, Vazquez A, Kim S, Lee AT, Chang EF, Razzaq T, Qazi A, Vargish G, Yuan X, Caccavano A, Hunt S, Chittajallu R, McLean N, Hewitt L, Paranzino E, Rice H, Cummins AC, Plotnikova A, Mohanty A, Tangen AC, Shin JH, Azadi R, Eldridge MAG, Alvarez VA, Averbeck BB, Alyahyay M, Vallejo TR, Soheib M, Vattino LG, MacGregor CP, Chatain CP, Banks E, Olah VJ, Naskar S, Hill S, Liebergall S, Badiani R, Hyde L, Hanley E, Xu Q, Allaway KC, Goldberg EM, Rowan MJM, Nowakowski TJ, Lee S, Favuzzi E, Kaeser PS, Sjulson L, Batista-Brito R, Takesian AE, Ibrahim LA, Iqbal A, Pelkey KA, McBain CJ, Dimidschstein J, Fishell G, Wang Y. An enhancer-AAV toolbox to target and manipulate distinct interneuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603924. [PMID: 39091835 PMCID: PMC11291062 DOI: 10.1101/2024.07.17.603924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, we and others have identified a number of enhancers that, when incorporated into rAAV vectors, can restrict the transgene expression to particular neuronal populations. Yet, viral tools to access and manipulate specific neuronal subtypes are still limited. Here, we performed systematic analysis of single cell genomic data to identify enhancer candidates for each of the telencephalic interneuron subtypes. We established a set of enhancer-AAV tools that are highly specific for distinct cortical interneuron populations and striatal cholinergic interneurons. These enhancers, when used in the context of different effectors, can target (fluorescent proteins), observe activity (GCaMP) and manipulate (opto-genetics) specific neuronal subtypes. We also validated our enhancer-AAV tools across species. Thus, we provide the field with a powerful set of tools to study neural circuits and functions and to develop precise and targeted therapy.
Collapse
|
10
|
Schünemann KD, Hattingh RM, Verhoog MB, Yang D, Bak AV, Peter S, van Loo KMJ, Wolking S, Kronenberg-Versteeg D, Weber Y, Schwarz N, Raimondo JV, Melvill R, Tromp SA, Butler JT, Höllig A, Delev D, Wuttke TV, Kampa BM, Koch H. Comprehensive analysis of human dendritic spine morphology and density. J Neurophysiol 2025; 133:1086-1102. [PMID: 40013734 DOI: 10.1152/jn.00622.2024] [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: 01/03/2025] [Revised: 01/14/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
Dendritic spines, small protrusions on neuronal dendrites, play a crucial role in brain function by changing shape and size in response to neural activity. So far, in-depth analysis of dendritic spines in human brain tissue is lacking. This study presents a comprehensive analysis of human dendritic spine morphology and density using a unique dataset from human brain tissue from 27 patients (8 females, 19 males, aged 18-71 yr) undergoing tumor or epilepsy surgery at three neurosurgery sites. We used acute slices and organotypic brain slice cultures to examine dendritic spines, classifying them into the three main morphological subtypes: mushroom, thin, and stubby, via three-dimensional (3-D) reconstruction using ZEISS arivis Pro software. A deep learning model, trained on 39 diverse datasets, automated spine segmentation and 3-D reconstruction, achieving a 74% F1-score and reducing processing time by over 50%. We show significant differences in spine density by sex, dendrite type, and tissue condition. Females had higher spine densities than males, and apical dendrites were denser in spines than basal ones. Acute tissue showed higher spine densities compared with cultured human brain tissue. With time in culture, mushroom spines decreased, whereas stubby and thin spine percentages increased, particularly from 7-9 to 14 days in vitro, reflecting potential synaptic plasticity changes. Our study underscores the importance of using human brain tissue to understand unique synaptic properties and shows that integrating deep learning with traditional methods enables efficient large-scale analysis, revealing key insights into sex- and tissue-specific dendritic spine dynamics relevant to neurological diseases.NEW & NOTEWORTHY This study presents a dataset of nearly 4,000 morphologically reconstructed human dendritic spines across different ages, gender, and tissue conditions. The dataset was further used to evaluate a deep learning algorithm for three-dimensional spine reconstruction, offering a scalable method for semiautomated spine analysis across various tissues and microscopy setups. The findings enhance understanding of human neurology, indicating potential connections between spine morphology, brain function, and the mechanisms of neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Kerstin D Schünemann
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Roxanne M Hattingh
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Matthijs B Verhoog
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Danqing Yang
- Institute of Neuroscience and Medicine 10, Research Center Juelich, Juelich, Germany
| | - Aniella V Bak
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sabrina Peter
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Karen M J van Loo
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan Wolking
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Deborah Kronenberg-Versteeg
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Yvonne Weber
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Joseph V Raimondo
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Roger Melvill
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Sean A Tromp
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - James T Butler
- Neuroscience Institute, University of Cape Town, Cape Town,South Africa
- Division of Neurosurgery, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Anke Höllig
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nürnberg, Erlangen, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Björn M Kampa
- Systems Neurophysiology, Institute of Biology II, RWTH Aachen University, Aachen, Germany
- JARA BRAIN Institute of Neuroscience and Medicine (INM-10), Research Center Juelich, Juelich, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
11
|
Kamaguchi R, Amemori S, Amemori KI, Osakada F. Bridge protein-mediated viral targeting of cells expressing endogenous μ-opioid G protein-coupled receptors in the mouse and monkey brain. Neurosci Res 2025; 213:35-50. [PMID: 39954866 DOI: 10.1016/j.neures.2025.02.007] [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: 01/07/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
Targeting specific cell types is essential for understanding their functional roles in the brain. Although genetic approaches enable cell-type-specific targeting in animals, their application to higher mammalian species, such as nonhuman primates, remains challenging. Here, we developed a nontransgenic method using bridge proteins to direct viral vectors to cells endogenously expressing μ-opioid receptors (MORs), a G protein-coupled receptor. The bridge protein comprises the avian viral receptor TVB, the MOR ligand β-endorphin (βed), and an interdomain linker. EnvB-enveloped viruses bind to the TVB component, followed by the interaction of βed with MORs, triggering viral infection in MOR-expressing cells. We optimized the secretion signals, domain arrangements, and interdomain linkers of the bridge proteins to maximize viral targeting efficiency and specificity. Alternative configurations incorporating different ligands and viral receptors also induced viral infection in MOR-expressing cells. The optimized βed-f2-TVB bridge protein with EnvB-pseudotyped lentiviruses induced infection in MOR-expressing cells in the striatum of mice and monkeys. An intersectional approach combining βed-f2-TVB with a neuron-specific promoter refined cell-type specificity. This study establishes the foundation for the rational bridge protein design and the feasibility of targeting G protein-coupled receptors beyond tyrosine kinase receptors, thereby expanding targetable cell types in the brain and throughout the body.
Collapse
Affiliation(s)
- Riki Kamaguchi
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Satoko Amemori
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan; Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Ken-Ichi Amemori
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan; Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Research Institute for Quantum and Chemical Innovation, Institutes of Innovation for Future Society, Nagoya University, Nagoya, Japan; PRESTO/CREST, Japan Science and Technology Agency (JST), Saitama, Japan.
| |
Collapse
|
12
|
Kussick E, Johansen N, Taskin N, Chowdhury A, Quinlan MA, Fraser A, Clark AG, Wynalda B, Martinez R, Groce EL, Reding M, Liang E, Shulga L, Huang C, Casper T, Clark M, Ho W, Gao Y, van Velthoven CTJ, Sobieski C, Ferrer R, Berg MR, Curtis BC, English C, Day JC, Fortuna MG, Donadio N, Newman D, Yao S, Chakka AB, Goldy J, Torkelson A, Guzman JB, Chakrabarty R, Nguy B, Guilford N, Pham TH, Wright V, Ronellenfitch K, Naidoo R, Kenney J, Williford A, Ramakrishnan C, Drinnenberg A, Gudsnuk K, Thyagarajan B, Smith KA, Dee N, Deisseroth K, Zeng H, Yao Z, Tasic B, Levi BP, Hodge R, Bakken TE, Lein ES, Ting JT, Daigle TL. Enhancer AAVs for targeting spinal motor neurons and descending motor pathways in rodents and macaque. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.30.605864. [PMID: 39131318 PMCID: PMC11312589 DOI: 10.1101/2024.07.30.605864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Experimental access to cell types within the mammalian spinal cord is severely limited by the availability of genetic tools. To enable access to lower motor neurons (LMNs) and LMN subtypes, we generated single cell multiome datasets from mouse and macaque spinal cords and discovered putative enhancers for each neuronal population. We cloned these enhancers into adeno-associated viral vectors (AAVs) driving a reporter fluorophore and functionally screened them in mouse. We extensively characterized the most promising candidate enhancers in rat and macaque and developed an optimized pan LMN enhancer virus. Additionally, we generated derivative viruses expressing iCre297T recombinase or ChR2-EYFP for labeling and functional studies, and we created a single vector with combined enhancer elements to achieve simultaneous labeling of layer 5 extratelencephalic projecting (ET) neurons and LMNs. This unprecedented LMN toolkit will enable future investigations of cell type function across species and potential therapeutic interventions for human neurodegenerative diseases.
Collapse
|
13
|
Johansen NJ, Kempynck N, Zemke NR, Somasundaram S, De Winter S, Hooper M, Dwivedi D, Lohia R, Wehbe F, Li B, Abaffyová D, Armand EJ, De Man J, Eksi EC, Hecker N, Hulselmans G, Konstantakos V, Mauduit D, Mich JK, Partel G, Daigle TL, Levi BP, Zhang K, Tanaka Y, Gillis J, Ting JT, Ben-Simon Y, Miller J, Ecker JR, Ren B, Aerts S, Lein ES, Tasic B, Bakken TE. Evaluating Methods for the Prediction of Cell Type-Specific Enhancers in the Mammalian Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.21.609075. [PMID: 39229027 PMCID: PMC11370467 DOI: 10.1101/2024.08.21.609075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Identifying cell type-specific enhancers in the brain is critical to building genetic tools for investigating the mammalian brain. Computational methods for functional enhancer prediction have been proposed and validated in the fruit fly and not yet the mammalian brain. We organized the 'Brain Initiative Cell Census Network (BICCN) Challenge: Predicting Functional Cell Type-Specific Enhancers from Cross-Species Multi-Omics' to assess machine learning and feature-based methods designed to nominate enhancer DNA sequences to target cell types in the mouse cortex. Methods were evaluated based on in vivo validation data from hundreds of cortical cell type-specific enhancers that were previously packaged into individual AAV vectors and retro-orbitally injected into mice. We find that open chromatin was a key predictor of functional enhancers, and sequence models improved prediction of non-functional enhancers that can be deprioritized as opposed to pursued for in vivo testing. Sequence models also identified cell type-specific transcription factor codes that can guide designs of in silico enhancers. This community challenge establishes a benchmark for enhancer prioritization algorithms and reveals computational approaches and molecular information that are crucial for identifying functional enhancers in mammalian cortical cell types. The results of this challenge bring us closer to understanding the complex gene regulatory landscape of the mammalian cortex and to designing more efficient genetic tools to target cortical cell types.
Collapse
Affiliation(s)
- Nelson J Johansen
- Allen Institute for Brain Science, Seattle, WA 98109
- These authors contributed equally
| | - Niklas Kempynck
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
- These authors contributed equally
| | - Nathan R Zemke
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | | | - Seppe De Winter
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Marcus Hooper
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Ruchi Lohia
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Fabien Wehbe
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Bocheng Li
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Darina Abaffyová
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Ethan J Armand
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093
| | - Julie De Man
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Eren Can Eksi
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Nikolai Hecker
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Gert Hulselmans
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Vasilis Konstantakos
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - David Mauduit
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Gabriele Partel
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | | | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Kai Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yoshiaki Tanaka
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Jesse Gillis
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | | | - Jeremy Miller
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Joseph R Ecker
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Bing Ren
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Stein Aerts
- VIB Center for AI & Computational Biology, VIB-KU Leuven Center for Brain and Disease Research & KU Leuven Department of Human Genetics, Leuven, Belgium
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Trygve E Bakken
- Allen Institute for Brain Science, Seattle, WA 98109
- Lead contact
| |
Collapse
|
14
|
Coughlin GM, Borsos M, Barcelona BH, Appling N, Mayfield AMH, Mackey ED, Eser RA, Jackson CR, Chen X, Kumar SR, Gradinaru V. Spatial genomics of AAV vectors reveals mechanism of transcriptional crosstalk that enables targeted delivery of large genetic cargo. Nat Biotechnol 2025:10.1038/s41587-025-02565-4. [PMID: 40113953 DOI: 10.1038/s41587-025-02565-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 01/17/2025] [Indexed: 03/22/2025]
Abstract
Cell-type-specific regulatory elements such as enhancers can direct expression of recombinant adeno-associated viruses (AAVs) to specific cell types, but this approach is limited by the relatively small packaging capacity of AAVs. In this study, we used spatial genomics to show that transcriptional crosstalk between individual AAV genomes provides a general method for cell-type-specific expression of large cargo by separating distally acting regulatory elements into a second AAV genome. We identified and profiled transcriptional crosstalk in AAV genomes carrying 11 different enhancers active in mouse brain. We developed spatial genomics methods to identify and localize AAV genomes and their concatemeric forms in cultured cells and in tissue, and we demonstrate here that transcriptional crosstalk is dependent upon concatemer formation. Finally, we leveraged transcriptional crosstalk to drive expression of a 3.2-kb Cas9 cargo in a cell-type-specific manner with systemically administered engineered AAVs, and we demonstrate AAV-delivered, minimally invasive, cell-type-specific gene editing in wild-type mice that recapitulates known disease phenotypes.
Collapse
Affiliation(s)
- Gerard M Coughlin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Máté Borsos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bre'Anna H Barcelona
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Nathan Appling
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Acacia M H Mayfield
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Elisha D Mackey
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rana A Eser
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Cameron R Jackson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
15
|
Hunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, Ayala A, Baker PM, Barcelli T, Barta S, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Clark M, Colbert K, Daniel S, Dawe T, Departee M, DiValentin P, Donadio NP, Dotson NI, Dwivedi D, Egdorf T, Fliss T, Gary A, Goldy J, Grasso C, Groce EL, Gudsnuk K, Han W, Haradon Z, Hastings S, Helback O, Ho WV, Huang C, Johnson T, Jones DL, Juneau Z, Kenney J, Leibly M, Li S, Liang E, Loeffler H, Lusk NA, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mich JK, Moosman S, Morin E, Naidoo R, Newman D, Ngo K, Nguyen K, Oster AL, Ouellette B, Oyama AA, Pena N, Pham T, Phillips E, Pom C, Potekhina L, Ransford S, Reding M, Rette DF, Reynoldson C, Rimorin C, Sigler AR, Rocha DB, Ronellenfitch K, Ruiz A, Sawyer L, et alHunker AC, Wirthlin ME, Gill G, Johansen NJ, Hooper M, Omstead V, Vargas S, Lerma MN, Taskin N, Weed N, Laird WD, Bishaw YM, Bendrick JL, Gore BB, Ben-Simon Y, Opitz-Araya X, Martinez RA, Way SW, Thyagarajan B, Otto S, Sanchez REA, Alexander JR, Amaya A, Amster A, Arbuckle J, Ayala A, Baker PM, Barcelli T, Barta S, Bertagnolli D, Bielstein C, Bishwakarma P, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Clark M, Colbert K, Daniel S, Dawe T, Departee M, DiValentin P, Donadio NP, Dotson NI, Dwivedi D, Egdorf T, Fliss T, Gary A, Goldy J, Grasso C, Groce EL, Gudsnuk K, Han W, Haradon Z, Hastings S, Helback O, Ho WV, Huang C, Johnson T, Jones DL, Juneau Z, Kenney J, Leibly M, Li S, Liang E, Loeffler H, Lusk NA, Madigan Z, Malloy J, Malone J, McCue R, Melchor J, Mich JK, Moosman S, Morin E, Naidoo R, Newman D, Ngo K, Nguyen K, Oster AL, Ouellette B, Oyama AA, Pena N, Pham T, Phillips E, Pom C, Potekhina L, Ransford S, Reding M, Rette DF, Reynoldson C, Rimorin C, Sigler AR, Rocha DB, Ronellenfitch K, Ruiz A, Sawyer L, Sevigny J, Shapovalova NV, Shepard N, Shulga L, Soliman S, Staats B, Taormina MJ, Tieu M, Wang Y, Wilkes J, Wood T, Zhou T, Williford A, Dee N, Mollenkopf T, Ng L, Esposito L, Kalmbach B, Yao S, Ariza J, Collman F, Mufti S, Smith K, Waters J, Ersing I, Patrick M, Zeng H, Lein ES, Kojima Y, Horwitz G, Owen SF, Levi BP, Daigle TL, Tasic B, Bakken TE, Ting JT. Enhancer AAV toolbox for accessing and perturbing striatal cell types and circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.27.615553. [PMID: 39386678 PMCID: PMC11463465 DOI: 10.1101/2024.09.27.615553] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
We present an enhancer AAV toolbox for accessing and perturbing striatal cell types and circuits. Best-in-class vectors were curated for accessing major striatal neuron populations including medium spiny neurons (MSNs), direct and indirect pathway MSNs, as well as Sst-Chodl, Pvalb-Pthlh, and cholinergic interneurons. Specificity was evaluated by multiple modes of molecular validation, three different routes of virus delivery, and with diverse transgene cargos. Importantly, we provide detailed information necessary to achieve reliable cell type specific labeling under different experimental contexts. We demonstrate direct pathway circuit-selective optogenetic perturbation of behavior and multiplex labeling of striatal interneuron types for targeted analysis of cellular features. Lastly, we show conserved in vivo activity for exemplary MSN enhancers in rat and macaque. This collection of striatal enhancer AAVs offers greater versatility compared to available transgenic lines and can readily be applied for cell type and circuit studies in diverse mammalian species beyond the mouse model.
Collapse
Affiliation(s)
| | | | - Gursajan Gill
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA
| | | | | | | | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | - Sven Otto
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Avalon Amaya
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Adam Amster
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Pam M Baker
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Tim Dawe
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA
| | - Tim Fliss
- Allen Institute for Brain Science, Seattle, WA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA
| | - Conor Grasso
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | | | - Warren Han
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Zeb Haradon
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Windy V Ho
- Allen Institute for Brain Science, Seattle, WA
| | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA
| | - Tye Johnson
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | - Zoe Juneau
- Allen Institute for Brain Science, Seattle, WA
| | | | | | - Su Li
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | - John K Mich
- Allen Institute for Brain Science, Seattle, WA
| | | | - Elyse Morin
- Allen Institute for Brain Science, Seattle, WA
| | - Robyn Naidoo
- Allen Institute for Neural Dynamics, Seattle, WA
| | | | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | - Nick Pena
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | | | | | | | | | - Yimin Wang
- Allen Institute for Brain Science, Seattle, WA
| | - Josh Wilkes
- Allen Institute for Neural Dynamics, Seattle, WA
| | - Toren Wood
- Allen Institute for Brain Science, Seattle, WA
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA
| | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA
| | | | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
| | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurological Surgery, University of Washington, Seattle, WA
| | - Yoshiko Kojima
- Department of Otolaryngology, Head and Neck Surgery, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| | - Greg Horwitz
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| | - Scott F Owen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
| | | | | | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA
- Washington National Primate Research Center, Seattle, WA
| |
Collapse
|
16
|
Mich JK, Ryu J, Wei AD, Gore BB, Guo R, Bard AM, Martinez RA, Luber EM, Liu J, Bishaw YM, Christian RJ, Oliveira LM, Miranda N, Ramirez JM, Ting JT, Lein ES, Levi BP, Kalume FK. Interneuron-specific dual-AAV SCN1A gene replacement corrects epileptic phenotypes in mouse models of Dravet syndrome. Sci Transl Med 2025; 17:eadn5603. [PMID: 40106582 DOI: 10.1126/scitranslmed.adn5603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Dravet syndrome (DS) is a severe developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10 to 20% rate of premature death. Most patients with DS harbor loss-of-function mutations in one copy of SCN1A, which encodes the voltage-gated sodium channel (NaV)1.1 alpha subunit and has been associated with inhibitory neuron dysfunction. Here, we generated a split-intein form of SCN1A and used a dual-vector delivery approach to circumvent adeno-associated virus (AAV) packaging limitations. In addition, we applied previously developed enhancer technology to produce an interneuron-specific gene replacement therapy for DS, called DLX2.0-SCN1A. The split-intein SCN1A vectors produced full-length NaV1.1 protein, and functional sodium channels were recorded in HEK293 cells in vitro. Administration of dual DLX2.0-SCN1A AAVs to wild-type mice produced full-length, reconstituted human protein by Western blot and telencephalic interneuron-specific and dose-dependent NaV1.1 expression by immunohistochemistry. These vectors also conferred strong dose-dependent protection against postnatal mortality and seizures in Scn1afl/+;Meox2-Cre and Scn1a+/R613X DS mouse models. Injection of single or dual DLX2.0-SCN1A AAVs into wild-type mice did not result in increased mortality, weight loss, or gliosis as measured by immunohistochemistry. In contrast, expression of SCN1A in all neurons driven by the human SYNAPSIN I promoter caused an adverse effect marked by increased mortality in the preweaning period, before disease onset. These findings demonstrate proof of concept that interneuron-specific AAV-mediated SCN1A gene replacement can rescue DS phenotypes in mouse models and suggest that it could be a therapeutic approach for patients with DS.
Collapse
Affiliation(s)
- John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jiyun Ryu
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Aguan D Wei
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rong Guo
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Angela M Bard
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Emily M Luber
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jiatai Liu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Luiz M Oliveira
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Jan-Marino Ramirez
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Franck K Kalume
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|
17
|
Jaeger ECB, Vijatovic D, Deryckere A, Zorin N, Nguyen AL, Ivanian G, Woych J, Arnold RC, Gurrola AO, Shvartsman A, Barbieri F, Toma FA, Cline HT, Shay TF, Kelley DB, Yamaguchi A, Shein-Idelson M, Tosches MA, Sweeney LB. Adeno-associated viral tools to trace neural development and connectivity across amphibians. Dev Cell 2025; 60:794-812.e6. [PMID: 39603234 PMCID: PMC12068381 DOI: 10.1016/j.devcel.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Amphibians, by virtue of their phylogenetic position, provide invaluable insights on nervous system evolution, development, and remodeling. The genetic toolkit for amphibians, however, remains limited. Recombinant adeno-associated viral vectors (AAVs) are a powerful alternative to transgenesis for labeling and manipulating neurons. Although successful in mammals, AAVs have never been shown to transduce amphibian cells efficiently. We screened AAVs in three amphibian species-the frogs Xenopus laevis and Pelophylax bedriagae and the salamander Pleurodeles waltl-and identified at least two AAV serotypes per species that transduce neurons. In developing amphibians, AAVs labeled groups of neurons generated at the same time during development. In the mature brain, AAVrg retrogradely traced long-range projections. Our study introduces AAVs as a tool for amphibian research, establishes a generalizable workflow for AAV screening in new species, and expands opportunities for cross-species comparisons of nervous system development, function, and evolution.
Collapse
Affiliation(s)
- Eliza C B Jaeger
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - David Vijatovic
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Astrid Deryckere
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Nikol Zorin
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Akemi L Nguyen
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Georgiy Ivanian
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jamie Woych
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Rebecca C Arnold
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Arik Shvartsman
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | | | - Florina A Toma
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Hollis T Cline
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Darcy B Kelley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ayako Yamaguchi
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Mark Shein-Idelson
- Department of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Lora B Sweeney
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
18
|
Franco-Enzástiga Ú, Inturi NN, Natarajan K, Mwirigi JM, Mazhar K, Schlachetzki JC, Schumacher M, Price TJ. Epigenomic landscape of the human dorsal root ganglion: sex differences and transcriptional regulation of nociceptive genes. Pain 2025; 166:614-630. [PMID: 39928726 PMCID: PMC11819886 DOI: 10.1097/j.pain.0000000000003508] [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: 07/17/2024] [Accepted: 10/14/2024] [Indexed: 02/12/2025]
Abstract
ABSTRACT Cell states are influenced by the regulation of gene expression orchestrated by transcription factors capable of binding to accessible DNA regions. To uncover if sex differences exist in chromatin accessibility in the human dorsal root ganglion (hDRG), where nociceptive neurons innervating the body are found, we performed bulk and spatial assays for transposase-accessible chromatin technology followed by sequencing (ATAC-seq) from organ donors without a history of chronic pain. Using bulk ATAC-seq, we detected abundant sex differences in the hDRG. In women, differentially accessible regions (DARs) mapped mostly to the X chromosome, whereas in men, they mapped to autosomal genes. Hormone-responsive transcription factor binding motifs such as EGR1/3 were abundant within DARs in women, while JUN, FOS, and other activating protein 1 factor motifs were enriched in men, suggesting a higher activation state of cells compared with women. These observations were consistent with spatial ATAC-seq data. Furthermore, we validated that EGR1 expression is biased to female hDRG using RNAscope. In neurons, spatial ATAC-seq revealed higher chromatin accessibility in GABAergic, glutamatergic, and interferon-related genes in women and in Ca2+-signaling-related genes in men. Strikingly, XIST, responsible for inactivating 1 X chromosome by compacting it and maintaining at the periphery of the nucleus, was found to be highly dispersed in female neuronal nuclei. This is likely related to the higher chromatin accessibility in X in female hDRG neurons observed using both ATAC-seq approaches. We have documented baseline epigenomic sex differences in the hDRG which provide important descriptive information to test future hypotheses.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Nikhil N. Inturi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Khadijah Mazhar
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Johannes C.M. Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, California, 94143 USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
19
|
Garcia FJ, Heiman M. Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases. Mol Neurodegener 2025; 20:13. [PMID: 39881338 PMCID: PMC11780804 DOI: 10.1186/s13024-025-00799-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: 11/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Many diseases and disorders of the nervous system suffer from a lack of adequate therapeutics to halt or slow disease progression, and to this day, no cure exists for any of the fatal neurodegenerative diseases. In part this is due to the incredible diversity of cell types that comprise the brain, knowledge gaps in understanding basic mechanisms of disease, as well as a lack of reliable strategies for delivering new therapeutic modalities to affected areas. With the advent of single cell genomics, it is now possible to interrogate the molecular characteristics of diverse cell populations and their alterations in diseased states. More recently, much attention has been devoted to cell populations that have historically been difficult to profile with bulk single cell technologies. In particular, cell types that comprise the cerebrovasculature have become increasingly better characterized in normal and neurodegenerative disease contexts. In this review, we describe the current understanding of cerebrovasculature structure, function, and cell type diversity and its role in the mechanisms underlying various neurodegenerative diseases. We focus on human and mouse cerebrovasculature studies and discuss both origins and consequences of cerebrovascular dysfunction, emphasizing known cell type-specific vulnerabilities in neuronal and cerebrovascular cell populations. Lastly, we highlight how novel insights into cerebrovascular biology have impacted the development of modern therapeutic approaches and discuss outstanding questions in the field.
Collapse
Affiliation(s)
- Francisco J Garcia
- The Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
20
|
Liu Y, McDaniel JA, Chen C, Yang L, Kipcak A, Savier EL, Erisir A, Cang J, Campbell JN. Co-Conservation of Synaptic Gene Expression and Circuitry in Collicular Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634521. [PMID: 39896595 PMCID: PMC11785205 DOI: 10.1101/2025.01.23.634521] [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
The superior colliculus (SC), a midbrain sensorimotor hub, is anatomically and functionally similar across vertebrates, but how its cell types have evolved is unclear. Using single-nucleus transcriptomics, we compared the SC's molecular and cellular organization in mice, tree shrews, and humans. Despite over 96 million years of evolutionary divergence, we identified ~30 consensus neuronal subtypes, including Cbln2+ neurons that form the SC-pulvinar circuit in mice and tree shrews. Synapse-related genes were among the most conserved, unlike neocortex, suggesting co-conservation of synaptic genes and circuitry. In contrast, cilia-related genes diverged significantly across species, highlighting the potential importance of the neuronal primary cilium in SC evolution. Additionally, we identified a novel inhibitory SC neuron in tree shrews and humans but not mice. Our findings reveal that the SC has evolved by conserving neuron subtypes, synaptic genes, and circuitry, while diversifying ciliary gene expression and an inhibitory neuron subtype.
Collapse
Affiliation(s)
- Yuanming Liu
- Department of Biology, Charlottesville, VA 22904, USA
| | - John A McDaniel
- Department of Psychology University of Virginia, Charlottesville, VA 22904, USA
| | - Chen Chen
- Department of Psychology University of Virginia, Charlottesville, VA 22904, USA
| | - Lu Yang
- Department of Biology, Charlottesville, VA 22904, USA
| | - Arda Kipcak
- Department of Psychology University of Virginia, Charlottesville, VA 22904, USA
| | | | - Alev Erisir
- Department of Psychology University of Virginia, Charlottesville, VA 22904, USA
| | - Jianhua Cang
- Department of Biology, Charlottesville, VA 22904, USA
- Department of Psychology University of Virginia, Charlottesville, VA 22904, USA
| | - John N Campbell
- Department of Biology, Charlottesville, VA 22904, USA
- Lead Contact
| |
Collapse
|
21
|
Hunker AC, Mich JK, Taskin N, Torkelson A, Pham T, Bertagnolli D, Chakka AB, Chakrabarty R, Donadio NP, Ferrer R, Gasperini M, Goldy J, Guzman JB, Jin K, Khem S, Kutsal R, Lalanne JB, Martinez RA, Newman D, Pena N, Shapovalova NV, Weed N, Zhou T, Yao S, Shendure J, Smith KA, Lein ES, Tasic B, Levi BP, Ting JT. Technical and biological sources of noise confound multiplexed enhancer AAV screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633018. [PMID: 39868122 PMCID: PMC11760716 DOI: 10.1101/2025.01.14.633018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cis -acting regulatory enhancer elements are valuable tools for gaining cell type-specific genetic access. Leveraging large chromatin accessibility atlases, putative enhancer sequences can be identified and deployed in adeno-associated virus (AAV) delivery platforms. However, a significant bottleneck in enhancer AAV discovery is charting their detailed expression patterns in vivo , a process that currently requires gold-standard one-by-one testing. Here we present a barcoded multiplex strategy for screening enhancer AAVs at cell type resolution using single cell RNA sequencing and taxonomy mapping. We executed a proof-of-concept study using a small pool of validated enhancer AAVs expressing in a variety of neuronal and non-neuronal cell types across the mouse brain. Unexpectedly, we encountered substantial technical and biological noise including chimeric packaging products, necessitating development of novel techniques to accurately deconvolve enhancer expression patterns. These results underscore the need for improved methods to mitigate noise and highlight the complexity of enhancer AAV biology in vivo .
Collapse
|
22
|
Lalanne JB, Mich JK, Huynh C, Hunker AC, McDiarmid TA, Levi BP, Ting JT, Shendure J. Extensive length and homology dependent chimerism in pool-packaged AAV libraries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632594. [PMID: 39868341 PMCID: PMC11761685 DOI: 10.1101/2025.01.14.632594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Adeno-associated viruses (AAVs) have emerged as the foremost gene therapy delivery vehicles due to their versatility, durability, and safety profile. Here we demonstrate extensive chimerism, manifesting as pervasive barcode swapping, among complex AAV libraries that are packaged as a pool. The observed chimerism is length- and homology-dependent but capsid-independent, in some cases affecting the majority of packaged AAV genomes. These results have implications for the design and deployment of functional AAV libraries in both research and clinical settings.
Collapse
Affiliation(s)
- Jean-Benoît Lalanne
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Chau Huynh
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | | | - Troy A McDiarmid
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| |
Collapse
|
23
|
Matthews EA, Russ JB, Qian Y, Zhao S, Thompson P, Methani M, Vestal ML, Josh Huang Z, Southwell DG. RNA-programmable cell type monitoring and manipulation in the human cortex with CellREADR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626590. [PMID: 39677799 PMCID: PMC11642864 DOI: 10.1101/2024.12.03.626590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Reliable and systematic experimental access to diverse cell types is necessary for understanding the neural circuit organization, function, and pathophysiology of the human brain. Methods for targeting human neural populations are scarce and currently center around identifying and engineering transcriptional enhancers and viral capsids. Here we demonstrate the utility of CellREADR, a programmable RNA sensor-effector technology that couples cellular RNA sensing to effector protein translation, for accessing, monitoring, and manipulating specific neuron types in ex vivo human cortical tissues. We designed CellREADR constructs to target two distinct human neuron types, CALB2+ (calretinin) GABAergic interneurons and FOXP2+ (forkhead box protein P2) glutamatergic projection neurons, and validated cell targeting using histological, electrophysiological, and transcriptomic methods. CellREADR-mediated expression of optogenetic effectors and genetically-encoded calcium indicators allowed us to manipulate and monitor these neuronal populations in cortical microcircuits. We further demonstrate that AAV-based CellREADR and enhancer vectors can be jointly used to target different subpopulations in the same preparation. By demonstrating specific, reliable, and programmable experimental access to targeted cell types, our results highlight CellREADR's potential for studying human neural circuits and treating brain disorders with cell type resolution.
Collapse
Affiliation(s)
- Elizabeth A. Matthews
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Jeffrey B. Russ
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Pediatrics, Division of Neurology, Duke University School of Medicine, Durham, NC USA
| | - Yongjun Qian
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Current affiliation: College of Future technology, Peking-Tsinghua Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Advanced Center of RNA Biology (BEACON), Peking University, China
| | - Shengli Zhao
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Peyton Thompson
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Muhib Methani
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Matthew L. Vestal
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Current affiliation: Department of Neurosurgery, Dartmouth University, Dartmouth, MA USA
| | - Z. Josh Huang
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC USA
| | - Derek G. Southwell
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC USA
| |
Collapse
|
24
|
Nagy MA, Price S, Wang K, Gill S, Ren E, Jayne L, Pajak V, Deighan S, Liu B, Lu X, Diallo A, Lo SC, Kleiman R, Henderson C, Suh J, Griffith EC, Greenberg ME, Hrvatin S. Cis-regulatory elements driving motor neuron-selective viral payload expression within the mammalian spinal cord. Proc Natl Acad Sci U S A 2024; 121:e2418024121. [PMID: 39602276 PMCID: PMC11626145 DOI: 10.1073/pnas.2418024121] [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: 09/10/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Spinal motor neuron (MN) dysfunction is the cause of a number of clinically significant movement disorders. Despite the recent approval of gene therapeutics targeting these MN-related disorders, there are no viral delivery mechanisms that achieve MN-restricted transgene expression. In this study, chromatin accessibility profiling of genetically defined mouse MNs was used to identify candidate cis-regulatory elements (CREs) capable of driving MN-selective gene expression. Subsequent testing of these candidates identified two CREs that confer MN-selective gene expression in the spinal cord as well as reduced off-target expression in dorsal root ganglia. Within one of these candidate elements, we identified a compact core transcription factor (TF)-binding region that drives MN-selective gene expression. Finally, we demonstrated that selective spinal cord expression driven by this mouse CRE is preserved in non-human primates. These findings suggest that cell-type-selective viral reagents in which cell-type-selective CREs drive restricted gene expression will be valuable research tools in mice and other mammalian species, with potentially significant therapeutic value in humans.
Collapse
Affiliation(s)
- M. Aurel Nagy
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, MA02115
| | - Spencer Price
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Kristina Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Department of Immunology, Harvard Medical School, Boston, MA02115
| | - Stanley Gill
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Erika Ren
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Lorna Jayne
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Victoria Pajak
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Sarah Deighan
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | | | | | | | | | | | | | - Eric C. Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | - Sinisa Hrvatin
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
25
|
Andrews JP, Geng J, Voitiuk K, Elliott MAT, Shin D, Robbins A, Spaeth A, Wang A, Li L, Solis D, Keefe MG, Sevetson JL, Rivera de Jesús JA, Donohue KC, Larson HH, Ehrlich D, Auguste KI, Salama S, Sohal V, Sharf T, Haussler D, Cadwell CR, Schaffer DV, Chang EF, Teodorescu M, Nowakowski TJ. Multimodal evaluation of network activity and optogenetic interventions in human hippocampal slices. Nat Neurosci 2024; 27:2487-2499. [PMID: 39548326 DOI: 10.1038/s41593-024-01782-5] [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: 08/24/2023] [Accepted: 08/30/2024] [Indexed: 11/17/2024]
Abstract
Seizures are made up of the coordinated activity of networks of neurons, suggesting that control of neurons in the pathologic circuits of epilepsy could allow for control of the disease. Optogenetics has been effective at stopping seizure-like activity in non-human disease models by increasing inhibitory tone or decreasing excitation, although this effect has not been shown in human brain tissue. Many of the genetic means for achieving channelrhodopsin expression in non-human models are not possible in humans, and vector-mediated methods are susceptible to species-specific tropism that may affect translational potential. Here we demonstrate adeno-associated virus-mediated, optogenetic reductions in network firing rates of human hippocampal slices recorded on high-density microelectrode arrays under several hyperactivity-provoking conditions. This platform can serve to bridge the gap between human and animal studies by exploring genetic interventions on network activity in human brain tissue.
Collapse
Affiliation(s)
- John P Andrews
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jinghui Geng
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Kateryna Voitiuk
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Matthew A T Elliott
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - David Shin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Ash Robbins
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Alex Spaeth
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Lin Li
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel Solis
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Matthew G Keefe
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica L Sevetson
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, USA
| | | | - Kevin C Donohue
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - H Hanh Larson
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Drew Ehrlich
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Computational Media, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Kurtis I Auguste
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sofie Salama
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, USA
| | - Vikaas Sohal
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tal Sharf
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - David Haussler
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - David V Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, California, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute (IGI), University of California, Berkeley, Berkeley, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Mircea Teodorescu
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA.
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA.
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA.
| | - Tomasz Jan Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Artemyev V, Gubaeva A, Paremskaia AI, Dzhioeva AA, Deviatkin A, Feoktistova SG, Mityaeva O, Volchkov PY. Synthetic Promoters in Gene Therapy: Design Approaches, Features and Applications. Cells 2024; 13:1963. [PMID: 39682712 PMCID: PMC11640742 DOI: 10.3390/cells13231963] [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: 10/24/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Gene therapy is a promising approach to the treatment of various inherited diseases, but its development is complicated by a number of limitations of the natural promoters used. The currently used strong ubiquitous natural promoters do not allow for the specificity of expression, while natural tissue-specific promoters have lowactivity. These limitations of natural promoters can be addressed by creating new synthetic promoters that achieve high levels of tissue-specific target gene expression. This review discusses recent advances in the development of synthetic promoters that provide a more precise regulation of gene expression. Approaches to the design of synthetic promoters are reviewed, including manual design and bioinformatic methods using machine learning. Examples of successful applications of synthetic promoters in the therapy of hereditary diseases and cancer are presented, as well as prospects for their clinical use.
Collapse
Affiliation(s)
- Valentin Artemyev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
| | - Anna Gubaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Anastasiia Iu. Paremskaia
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Amina A. Dzhioeva
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
| | - Andrei Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Sofya G. Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Olga Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
| | - Pavel Yu. Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
- Moscow Clinical Scientific Center N.A. A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
27
|
Arokiaraj CM, Leone MJ, Kleyman M, Chamessian A, Noh MC, Phan BN, Lopes BC, Corrigan KA, Cherupally VK, Yeramosu D, Franusich ME, Podder R, Lele S, Shiers S, Kang B, Kennedy MM, Chen V, Chen Z, Mathys H, Dum RP, Lewis DA, Qadri Y, Price TJ, Pfenning AR, Seal RP. Spatial, transcriptomic, and epigenomic analyses link dorsal horn neurons to chronic pain genetic predisposition. Cell Rep 2024; 43:114876. [PMID: 39453813 PMCID: PMC11801220 DOI: 10.1016/j.celrep.2024.114876] [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] [Revised: 05/07/2024] [Accepted: 09/30/2024] [Indexed: 10/27/2024] Open
Abstract
Key mechanisms underlying chronic pain occur within the dorsal horn. Genome-wide association studies (GWASs) have identified genetic variants predisposed to chronic pain. However, most of these variants lie within regulatory non-coding regions that have not been linked to spinal cord biology. Here, we take a multi-species approach to determine whether chronic pain variants impact the regulatory genomics of dorsal horn neurons. First, we generate a large rhesus macaque single-nucleus RNA sequencing (snRNA-seq) atlas and integrate it with available human and mouse datasets to produce a single unified, species-conserved atlas of neuron subtypes. Cellular-resolution spatial transcriptomics in mouse shows the precise laminar location of these neuron subtypes, consistent with our analysis of neuron-subtype-selective markers in macaque. Using this cross-species framework, we generate a mouse single-nucleus open chromatin atlas of regulatory elements that shows strong and selective relationships between the neuron-subtype-specific chromatin regions and variants from major chronic pain GWASs.
Collapse
Affiliation(s)
- Cynthia M Arokiaraj
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael J Leone
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael Kleyman
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27708, USA; Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Myung-Chul Noh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - BaDoi N Phan
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Bettega C Lopes
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kelly A Corrigan
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Vijay Kiran Cherupally
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Deepika Yeramosu
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Michael E Franusich
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Riya Podder
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sumitra Lele
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Byungsoo Kang
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Meaghan M Kennedy
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Viola Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ziheng Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Hansruedi Mathys
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Richard P Dum
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yawar Qadri
- Department of Anesthesiology, Emory University, Atlanta, GA 30038, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas R Pfenning
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
28
|
Yang X, Woldemichael K, Hover J, Zhao S, Guo X, Qian Y, Gillis J, Huang ZJ. RNA programmable cell targeting and manipulation with CellREADR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625312. [PMID: 39651297 PMCID: PMC11623600 DOI: 10.1101/2024.11.26.625312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Methods that provide specific, easy, and scalable experimental access to animal cell types and cell states will have broad applications in biology and medicine. CellREADR - Cell access through RNA sensing by Endogenous ADAR (adenosine deaminase acting on RNA), is a programmable RNA sensor-actuator technology that couples the detection of a cell-defining RNA to the translation of an effector protein to monitor and manipulate the cell. The CellREADR RNA device consists of a 5' sensor region complementary to a cellular RNA and a 3' payload coding region; payload translation is gated by the removal of a STOP codon in the sensor region upon base pairing with the cognate cellular RNA through an ADAR-mediated A- to-I editing mechanism ubiquitous to metazoan cells. CellREADR thus highlights the potential for RNA-based monitoring and manipulation of animal cells in ways that are simple, versatile, and generalizable across tissues and species. Here, we describe a detailed protocol for implementing CellREADR experiments in cell cultures and in animals. The procedure includes sensor and payload design, cloning, validation and characterization in mammalian cell cultures. The in vivo animal protocol focuses on AAV-based delivery of CellREADR using brain tissue as examples. We describe current best practices, various experimental controls and trouble-shooting steps. Beginning from sensor design, the validation of RNA sensor-actuators in cell cultures can be completed in 2-3 weeks. The construction of AAV vectors and their in vivo validation and characterization will take additional 6-8 weeks.
Collapse
|
29
|
Lazaridis I, Crittenden JR, Ahn G, Hirokane K, Wickersham IR, Yoshida T, Mahar A, Skara V, Loftus JH, Parvataneni K, Meletis K, Ting JT, Hueske E, Matsushima A, Graybiel AM. Striosomes control dopamine via dual pathways paralleling canonical basal ganglia circuits. Curr Biol 2024; 34:5263-5283.e8. [PMID: 39447573 DOI: 10.1016/j.cub.2024.09.070] [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: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024]
Abstract
Balanced activity of canonical direct D1 and indirect D2 basal ganglia pathways is considered a core requirement for normal movement, and their imbalance is an etiologic factor in movement and neuropsychiatric disorders. We present evidence for a conceptually equivalent pair of direct D1 and indirect D2 pathways that arise from striatal projection neurons (SPNs) of the striosome compartment rather than from SPNs of the matrix, as do the canonical pathways. These striosomal D1 (S-D1) and D2 (S-D2) pathways target substantia nigra dopamine-containing neurons instead of basal ganglia motor output nuclei. They modulate movement with net effects opposite to those exerted by the canonical pathways: S-D1 is net inhibitory and S-D2 is net excitatory. The S-D1 and S-D2 circuits likely influence motivation for learning and action, complementing and reorienting canonical pathway modulation. A major conceptual reformulation of the classic direct-indirect pathway model of basal ganglia function is needed, as well as reconsideration of the effects of D2-targeting therapeutic drugs.
Collapse
Affiliation(s)
- Iakovos Lazaridis
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jill R Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gun Ahn
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kojiro Hirokane
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoko Yoshida
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ara Mahar
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vasiliki Skara
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Johnny H Loftus
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Krishna Parvataneni
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Jonathan T Ting
- Human Cell Types Department, Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Emily Hueske
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
30
|
Kerkhoff WG, Stauffer WR. Basal ganglia: Uniting circuit logic between matrix and striosome. Curr Biol 2024; 34:R1149-R1152. [PMID: 39561711 DOI: 10.1016/j.cub.2024.10.010] [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: 11/21/2024]
Abstract
A new study has identified a novel direct-indirect circuit architecture connecting the striosome compartment of the striatum with midbrain dopamine neurons. This circuit has the potential to integrate limbic and sensorimotor functions and to exert substantial control over biological reinforcement leaning.
Collapse
Affiliation(s)
- Willa G Kerkhoff
- Center for Neuroscience, Department of Neurobiology, The University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - William R Stauffer
- Center for Neuroscience, Department of Neurobiology, The University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
31
|
Chen X. Reimagining Cortical Connectivity by Deconstructing Its Molecular Logic into Building Blocks. Cold Spring Harb Perspect Biol 2024; 16:a041509. [PMID: 38621822 PMCID: PMC11529856 DOI: 10.1101/cshperspect.a041509] [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: 04/17/2024]
Abstract
Comprehensive maps of neuronal connectivity provide a foundation for understanding the structure of neural circuits. In a circuit, neurons are diverse in morphology, electrophysiology, gene expression, activity, and other neuronal properties. Thus, constructing a comprehensive connectivity map requires associating various properties of neurons, including their connectivity, at cellular resolution. A commonly used approach is to use the gene expression profiles as an anchor to which all other neuronal properties are associated. Recent advances in genomics and anatomical techniques dramatically improved the ability to determine and associate the long-range projections of neurons with their gene expression profiles. These studies revealed unprecedented details of the gene-projection relationship, but also highlighted conceptual challenges in understanding this relationship. In this article, I delve into the findings and the challenges revealed by recent studies using state-of-the-art neuroanatomical and transcriptomic techniques. Building upon these insights, I propose an approach that focuses on understanding the gene-projection relationship through basic features in gene expression profiles and projections, respectively, that associate with underlying cellular processes. I then discuss how the developmental trajectories of projections and gene expression profiles create additional challenges and necessitate interrogating the gene-projection relationship across time. Finally, I explore complementary strategies that, together, can provide a comprehensive view of the gene-projection relationship.
Collapse
Affiliation(s)
- Xiaoyin Chen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| |
Collapse
|
32
|
Gordon JA, Dzirasa K, Petzschner FH. The neuroscience of mental illness: Building toward the future. Cell 2024; 187:5858-5870. [PMID: 39423804 PMCID: PMC11490687 DOI: 10.1016/j.cell.2024.09.028] [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: 09/13/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
Mental illnesses arise from dysfunction in the brain. Although numerous extraneural factors influence these illnesses, ultimately, it is the science of the brain that will lead to novel therapies. Meanwhile, our understanding of this complex organ is incomplete, leading to the oft-repeated trope that neuroscience has yet to make significant contributions to the care of individuals with mental illnesses. This review seeks to counter this narrative, using specific examples of how neuroscientific advances have contributed to progress in mental health care in the past and how current achievements set the stage for further progress in the future.
Collapse
Affiliation(s)
- Joshua A Gordon
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA.
| | - Kafui Dzirasa
- Departments of Psychiatry and Behavioral Sciences, Neurology, and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | |
Collapse
|
33
|
Teo AYY, Squair JW, Courtine G, Skinnider MA. Best practices for differential accessibility analysis in single-cell epigenomics. Nat Commun 2024; 15:8805. [PMID: 39394227 PMCID: PMC11470024 DOI: 10.1038/s41467-024-53089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
Differential accessibility (DA) analysis of single-cell epigenomics data enables the discovery of regulatory programs that establish cell type identity and steer responses to physiological and pathophysiological perturbations. While many statistical methods to identify DA regions have been developed, the principles that determine the performance of these methods remain unclear. As a result, there is no consensus on the most appropriate statistical methods for DA analysis of single-cell epigenomics data. Here, we present a systematic evaluation of statistical methods that have been applied to identify DA regions in single-cell ATAC-seq (scATAC-seq) data. We leverage a compendium of scATAC-seq experiments with matching bulk ATAC-seq or scRNA-seq in order to assess the accuracy, bias, robustness, and scalability of each statistical method. The structure of our experiments also provides the opportunity to define best practices for the analysis of scATAC-seq data beyond DA itself. We leverage this understanding to develop an R package implementing these best practices.
Collapse
Affiliation(s)
- Alan Yue Yang Teo
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
- NeuroX Institute and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Jordan W Squair
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
- NeuroX Institute and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Gregoire Courtine
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
- NeuroX Institute and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Michael A Skinnider
- Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
- NeuroX Institute and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
34
|
Ben-Simon Y, Hooper M, Narayan S, Daigle T, Dwivedi D, Way SW, Oster A, Stafford DA, Mich JK, Taormina MJ, Martinez RA, Opitz-Araya X, Roth JR, Allen S, Ayala A, Bakken TE, Barcelli T, Barta S, Bendrick J, Bertagnolli D, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chance RK, Chavan S, Departee M, Donadio N, Dotson N, Egdorf T, Gabitto M, Garcia J, Gary A, Gasperini M, Goldy J, Gore BB, Graybuck L, Greisman N, Haeseleer F, Halterman C, Helback O, Hockemeyer D, Huang C, Huff S, Hunker A, Johansen N, Juneau Z, Kalmbach B, Khem S, Kussick E, Kutsal R, Larsen R, Lee C, Lee AY, Leibly M, Lenz GH, Liang E, Lusk N, Malone J, Mollenkopf T, Morin E, Newman D, Ng L, Ngo K, Omstead V, Oyama A, Pham T, Pom CA, Potekhina L, Ransford S, Rette D, Rimorin C, Rocha D, Ruiz A, Sanchez RE, Sedeno-Cortes A, Sevigny JP, Shapovalova N, Shulga L, Sigler AR, Siverts LA, Somasundaram S, Stewart K, Szelenyi E, Tieu M, Trader C, van Velthoven CT, Walker M, Weed N, Wirthlin M, Wood T, Wynalda B, Yao Z, Zhou T, Ariza J, Dee N, Reding M, et alBen-Simon Y, Hooper M, Narayan S, Daigle T, Dwivedi D, Way SW, Oster A, Stafford DA, Mich JK, Taormina MJ, Martinez RA, Opitz-Araya X, Roth JR, Allen S, Ayala A, Bakken TE, Barcelli T, Barta S, Bendrick J, Bertagnolli D, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chance RK, Chavan S, Departee M, Donadio N, Dotson N, Egdorf T, Gabitto M, Garcia J, Gary A, Gasperini M, Goldy J, Gore BB, Graybuck L, Greisman N, Haeseleer F, Halterman C, Helback O, Hockemeyer D, Huang C, Huff S, Hunker A, Johansen N, Juneau Z, Kalmbach B, Khem S, Kussick E, Kutsal R, Larsen R, Lee C, Lee AY, Leibly M, Lenz GH, Liang E, Lusk N, Malone J, Mollenkopf T, Morin E, Newman D, Ng L, Ngo K, Omstead V, Oyama A, Pham T, Pom CA, Potekhina L, Ransford S, Rette D, Rimorin C, Rocha D, Ruiz A, Sanchez RE, Sedeno-Cortes A, Sevigny JP, Shapovalova N, Shulga L, Sigler AR, Siverts LA, Somasundaram S, Stewart K, Szelenyi E, Tieu M, Trader C, van Velthoven CT, Walker M, Weed N, Wirthlin M, Wood T, Wynalda B, Yao Z, Zhou T, Ariza J, Dee N, Reding M, Ronellenfitch K, Mufti S, Sunkin SM, Smith KA, Esposito L, Waters J, Thyagarajan B, Yao S, Lein ES, Zeng H, Levi BP, Ngai J, Ting J, Tasic B. A suite of enhancer AAVs and transgenic mouse lines for genetic access to cortical cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.597244. [PMID: 38915722 PMCID: PMC11195086 DOI: 10.1101/2024.06.10.597244] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The mammalian cortex is comprised of cells classified into types according to shared properties. Defining the contribution of each cell type to the processes guided by the cortex is essential for understanding its function in health and disease. We used transcriptomic and epigenomic cortical cell type taxonomies from mouse and human to define marker genes and putative enhancers and created a large toolkit of transgenic lines and enhancer AAVs for selective targeting of cortical cell populations. We report evaluation of fifteen new transgenic driver lines, two new reporter lines, and >800 different enhancer AAVs covering most subclasses of cortical cells. The tools reported here as well as the scaled process of tool creation and modification enable diverse experimental strategies towards understanding mammalian cortex and brain function.
Collapse
Affiliation(s)
- Yoav Ben-Simon
- Allen Institute for Brain Science, Seattle, WA 98109
- Equivalent contribution
| | - Marcus Hooper
- Allen Institute for Brain Science, Seattle, WA 98109
- Equivalent contribution
| | - Sujatha Narayan
- Allen Institute for Brain Science, Seattle, WA 98109
- Equivalent contribution
| | - Tanya Daigle
- Allen Institute for Brain Science, Seattle, WA 98109
- Equivalent contribution
| | | | - Sharon W. Way
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Aaron Oster
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - John K. Mich
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | - Jada R. Roth
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Shona Allen
- University of California, Berkeley, Berkeley, CA 94720
| | - Angela Ayala
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | - Stuard Barta
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | | | | | | | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | - Sakshi Chavan
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Jazmin Garcia
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Bryan B. Gore
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Noah Greisman
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | | | - Cindy Huang
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Sydney Huff
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Avery Hunker
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Zoe Juneau
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Shannon Khem
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Emily Kussick
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Rana Kutsal
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Angus Y. Lee
- University of California, Berkeley, Berkeley, CA 94720
| | | | | | | | - Nicholas Lusk
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | - Elyse Morin
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Dakota Newman
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Alana Oyama
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | - Shea Ransford
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Dean Rette
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Dana Rocha
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Augustin Ruiz
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | | | | | - Ana R. Sigler
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | - Kaiya Stewart
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Eric Szelenyi
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Toren Wood
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | | | - Luke Esposito
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA 98109
| | | | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Ed S. Lein
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Boaz P. Levi
- Allen Institute for Brain Science, Seattle, WA 98109
| | - John Ngai
- University of California, Berkeley, Berkeley, CA 94720
- Present affiliation: National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Jonathan Ting
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109
- Lead contact
| |
Collapse
|
35
|
Griffith EC, West AE, Greenberg ME. Neuronal enhancers fine-tune adaptive circuit plasticity. Neuron 2024; 112:3043-3057. [PMID: 39208805 PMCID: PMC11550865 DOI: 10.1016/j.neuron.2024.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Neuronal activity-regulated gene expression plays a crucial role in sculpting neural circuits that underpin adaptive brain function. Transcriptional enhancers are now recognized as key components of gene regulation that orchestrate spatiotemporally precise patterns of gene transcription. We propose that the dynamics of enhancer activation uniquely position these genomic elements to finely tune activity-dependent cellular plasticity. Enhancer specificity and modularity can be exploited to gain selective genetic access to specific cell states, and the precise modulation of target gene expression within restricted cellular contexts enabled by targeted enhancer manipulation allows for fine-grained evaluation of gene function. Mounting evidence also suggests that enduring stimulus-induced changes in enhancer states can modify target gene activation upon restimulation, thereby contributing to a form of cell-wide metaplasticity. We advocate for focused exploration of activity-dependent enhancer function to gain new insight into the mechanisms underlying brain plasticity and cognitive dysfunction.
Collapse
Affiliation(s)
- Eric C Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
36
|
Dembrow NC, Sawchuk S, Dalley R, Opitz-Araya X, Hudson M, Radaelli C, Alfiler L, Walling-Bell S, Bertagnolli D, Goldy J, Johansen N, Miller JA, Nasirova K, Owen SF, Parga-Becerra A, Taskin N, Tieu M, Vumbaco D, Weed N, Wilson J, Lee BR, Smith KA, Sorensen SA, Spain WJ, Lein ES, Perlmutter SI, Ting JT, Kalmbach BE. Areal specializations in the morpho-electric and transcriptomic properties of primate layer 5 extratelencephalic projection neurons. Cell Rep 2024; 43:114718. [PMID: 39277859 PMCID: PMC11488157 DOI: 10.1016/j.celrep.2024.114718] [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: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024] Open
Abstract
Large-scale analysis of single-cell gene expression has revealed transcriptomically defined cell subclasses present throughout the primate neocortex with gene expression profiles that differ depending upon neocortical region. Here, we test whether the interareal differences in gene expression translate to regional specializations in the physiology and morphology of infragranular glutamatergic neurons by performing Patch-seq experiments in brain slices from the temporal cortex (TCx) and motor cortex (MCx) of the macaque. We confirm that transcriptomically defined extratelencephalically projecting neurons of layer 5 (L5 ET neurons) include retrogradely labeled corticospinal neurons in the MCx and find multiple physiological properties and ion channel genes that distinguish L5 ET from non-ET neurons in both areas. Additionally, while infragranular ET and non-ET neurons retain distinct neuronal properties across multiple regions, there are regional morpho-electric and gene expression specializations in the L5 ET subclass, providing mechanistic insights into the specialized functional architecture of the primate neocortex.
Collapse
Affiliation(s)
- Nikolai C Dembrow
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA.
| | - Scott Sawchuk
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Mark Hudson
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | | | - Lauren Alfiler
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Scott F Owen
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Alejandro Parga-Becerra
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - David Vumbaco
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Julia Wilson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - William J Spain
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Steve I Perlmutter
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Jonathan T Ting
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Brian E Kalmbach
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
37
|
Federer F, Balsor J, Ingold A, Babcock DP, Dimidschstein J, Angelucci A. Laminar specificity and coverage of viral-mediated gene expression restricted to GABAergic interneurons and their parvalbumin subclass in marmoset primary visual cortex. eLife 2024; 13:RP97673. [PMID: 39297605 PMCID: PMC11412690 DOI: 10.7554/elife.97673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
In the mammalian neocortex, inhibition is important for dynamically balancing excitation and shaping the response properties of cells and circuits. The various computational functions of inhibition are thought to be mediated by different inhibitory neuron types, of which a large diversity exists in several species. Current understanding of the function and connectivity of distinct inhibitory neuron types has mainly derived from studies in transgenic mice. However, it is unknown whether knowledge gained from mouse studies applies to the non-human primate, the model system closest to humans. The lack of viral tools to selectively access inhibitory neuron types has been a major impediment to studying their function in the primate. Here, we have thoroughly validated and characterized several recently developed viral vectors designed to restrict transgene expression to GABAergic cells or their parvalbumin (PV) subtype, and identified two types that show high specificity and efficiency in marmoset V1. We show that in marmoset V1, AAV-h56D induces transgene expression in GABAergic cells with up to 91-94% specificity and 79% efficiency, but this depends on viral serotype and cortical layer. AAV-PHP.eB-S5E2 induces transgene expression in PV cells across all cortical layers with up to 98% specificity and 86-90% efficiency, depending on layer. Thus, these viral vectors are promising tools for studying GABA and PV cell function and connectivity in the primate cortex.
Collapse
Affiliation(s)
- Frederick Federer
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of UtahSalt Lake CityUnited States
| | - Justin Balsor
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of UtahSalt Lake CityUnited States
| | - Alexander Ingold
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of UtahSalt Lake CityUnited States
| | - David P Babcock
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of UtahSalt Lake CityUnited States
| | | | - Alessandra Angelucci
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of UtahSalt Lake CityUnited States
| |
Collapse
|
38
|
Haziza S, Chrapkiewicz R, Zhang Y, Kruzhilin V, Li J, Li J, Delamare G, Swanson R, Buzsáki G, Kannan M, Vasan G, Lin MZ, Zeng H, Daigle TL, Schnitzer MJ. Imaging high-frequency voltage dynamics in multiple neuron classes of behaving mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.607428. [PMID: 39185175 PMCID: PMC11343216 DOI: 10.1101/2024.08.15.607428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Fluorescent genetically encoded voltage indicators report transmembrane potentials of targeted cell-types. However, voltage-imaging instrumentation has lacked the sensitivity to track spontaneous or evoked high-frequency voltage oscillations in neural populations. Here we describe two complementary TEMPO voltage-sensing technologies that capture neural oscillations up to ~100 Hz. Fiber-optic TEMPO achieves ~10-fold greater sensitivity than prior photometry systems, allows hour-long recordings, and monitors two neuron-classes per fiber-optic probe in freely moving mice. With it, we uncovered cross-frequency-coupled theta- and gamma-range oscillations and characterized excitatory-inhibitory neural dynamics during hippocampal ripples and visual cortical processing. The TEMPO mesoscope images voltage activity in two cell-classes across a ~8-mm-wide field-of-view in head-fixed animals. In awake mice, it revealed sensory-evoked excitatory-inhibitory neural interactions and traveling gamma and 3-7 Hz waves in the visual cortex, and previously unreported propagation directions for hippocampal theta and beta waves. These technologies have widespread applications probing diverse oscillations and neuron-type interactions in healthy and diseased brains.
Collapse
Affiliation(s)
- Simon Haziza
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Radosław Chrapkiewicz
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Yanping Zhang
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Vasily Kruzhilin
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Jane Li
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Jizhou Li
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | | | - Rachel Swanson
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY 10016, USA
| | - György Buzsáki
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY 10016, USA
- Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA
| | - Madhuvanthi Kannan
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ganesh Vasan
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Z Lin
- Departments of Bioengineering & Pediatrics, Stanford University, Stanford CA 94305, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Mark J Schnitzer
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Lead contact
| |
Collapse
|
39
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
40
|
Lazaridis I, Crittenden JR, Ahn G, Hirokane K, Yoshida T, Wickersham IR, Mahar A, Skara V, Loftus JH, Parvataneni K, Meletis K, Ting JT, Hueske E, Matsushima A, Graybiel AM. Striosomes Target Nigral Dopamine-Containing Neurons via Direct-D1 and Indirect-D2 Pathways Paralleling Classic Direct-Indirect Basal Ganglia Systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596922. [PMID: 38915684 PMCID: PMC11195572 DOI: 10.1101/2024.06.01.596922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Balanced activity of canonical direct D1 and indirect D2 basal ganglia pathways is considered a core requirement for normal movement, and their imbalance is an etiologic factor in movement and neuropsychiatric disorders. We present evidence for a conceptually equivalent pair of direct-D1 and indirect-D2 pathways that arise from striatal projection neurons (SPNs) of the striosome compartment rather than from SPNs of the matrix, as do the canonical pathways. These S-D1 and S-D2 striosomal pathways target substantia nigra dopamine-containing neurons instead of basal ganglia motor output nuclei. They modulate movement oppositely to the modulation by the canonical pathways: S-D1 is inhibitory and S-D2 is excitatory. The S-D1 and S-D2 circuits likely influence motivation for learning and action, complementing and reorienting canonical pathway modulation. A major conceptual reformulation of the classic direct-indirect pathway model of basal ganglia function is needed, as well as reconsideration of the effects of D2-targeting therapeutic drugs.
Collapse
Affiliation(s)
- Iakovos Lazaridis
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Jill R. Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Gun Ahn
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Kojiro Hirokane
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Tomoko Yoshida
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Ian R. Wickersham
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Ara Mahar
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | | | - Johnny H. Loftus
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Krishna Parvataneni
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | | | - Jonathan T. Ting
- Human Cell Types Dept, Allen Institute for Brain Science, Seattle WA 98109, USA
- Department of Physiology and Biophysics, University of Washington, Seattle WA 98195, USA
| | - Emily Hueske
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| | - Ann M. Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences
| |
Collapse
|
41
|
Agmon A, Barth AL. A brief history of somatostatin interneuron taxonomy or: how many somatostatin subtypes are there, really? Front Neural Circuits 2024; 18:1436915. [PMID: 39091993 PMCID: PMC11292610 DOI: 10.3389/fncir.2024.1436915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024] Open
Abstract
We provide a brief (and unabashedly biased) overview of the pre-transcriptomic history of somatostatin interneuron taxonomy, followed by a chronological summary of the large-scale, NIH-supported effort over the last ten years to generate a comprehensive, single-cell RNA-seq-based taxonomy of cortical neurons. Focusing on somatostatin interneurons, we present the perspective of experimental neuroscientists trying to incorporate the new classification schemes into their own research while struggling to keep up with the ever-increasing number of proposed cell types, which seems to double every two years. We suggest that for experimental analysis, the most useful taxonomic level is the subdivision of somatostatin interneurons into ten or so "supertypes," which closely agrees with their more traditional classification by morphological, electrophysiological and neurochemical features. We argue that finer subdivisions ("t-types" or "clusters"), based on slight variations in gene expression profiles but lacking clear phenotypic differences, are less useful to researchers and may actually defeat the purpose of classifying neurons to begin with. We end by stressing the need for generating novel tools (mouse lines, viral vectors) for genetically targeting distinct supertypes for expression of fluorescent reporters, calcium sensors and excitatory or inhibitory opsins, allowing neuroscientists to chart the input and output synaptic connections of each proposed subtype, reveal the position they occupy in the cortical network and examine experimentally their roles in sensorimotor behaviors and cognitive brain functions.
Collapse
Affiliation(s)
- Ariel Agmon
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Alison L. Barth
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
42
|
Franco-Enzástiga Ú, Inturi NN, Natarajan K, Mwirigi JM, Mazhar K, Schlachetzki JC, Schumacher M, Price TJ. Epigenomic landscape of the human dorsal root ganglion: sex differences and transcriptional regulation of nociceptive genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587047. [PMID: 38586055 PMCID: PMC10996669 DOI: 10.1101/2024.03.27.587047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Gene expression is influenced by chromatin architecture via controlled access of regulatory factors to DNA. To better understand gene regulation in the human dorsal root ganglion (hDRG) we used bulk and spatial transposase-accessible chromatin technology followed by sequencing (ATAC-seq). Using bulk ATAC-seq, we detected that in females diverse differentially accessible chromatin regions (DARs) mapped to the X chromosome and in males to autosomal genes. EGR1/3 and SP1/4 transcription factor binding motifs were abundant within DARs in females, and JUN, FOS and other AP-1 factors in males. To dissect the open chromatin profile in hDRG neurons, we used spatial ATAC-seq. The neuron cluster showed higher chromatin accessibility in GABAergic, glutamatergic, and interferon-related genes in females, and in Ca2+- signaling-related genes in males. Sex differences in transcription factor binding sites in neuron-proximal barcodes were consistent with the trends observed in bulk ATAC-seq data. We validated that EGR1 expression is biased to female hDRG compared to male. Strikingly, XIST, the long-noncoding RNA responsible for X inactivation, hybridization signal was found to be highly dispersed in the female neuronal but not non-neuronal nuclei suggesting weak X inactivation in female hDRG neurons. Our findings point to baseline epigenomic sex differences in the hDRG that likely underlie divergent transcriptional responses that determine mechanistic sex differences in pain.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Nikhil N. Inturi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Khadija Mazhar
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Johannes C.M. Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, California, 94143 USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
43
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [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: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
44
|
Kojima L, Seiriki K, Rokujo H, Nakazawa T, Kasai A, Hashimoto H. Optimization of AAV vectors for transactivator-regulated enhanced gene expression within targeted neuronal populations. iScience 2024; 27:109878. [PMID: 38799556 PMCID: PMC11126825 DOI: 10.1016/j.isci.2024.109878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/03/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Adeno-associated virus (AAV) vectors are potential tools for cell-type-selective gene delivery to the central nervous system. Although cell-type-specific enhancers and promoters have been identified for AAV systems, there is limited information regarding the effects of AAV genomic components on the selectivity and efficiency of gene expression. Here, we offer an alternative strategy to provide specific and efficient gene delivery to a targeted neuronal population by optimizing recombinant AAV genomic components, named TAREGET (TransActivator-Regulated Enhanced Gene Expression within Targeted neuronal populations). We established this strategy in oxytocinergic neurons and showed that the TAREGET enabled sufficient gene expression to label long-projecting axons in wild-type mice. Its application to other cell types, including serotonergic and dopaminergic neurons, was also demonstrated. These results demonstrate that optimization of AAV expression cassettes can improve the specificity and efficiency of cell-type-specific gene expression and that TAREGET can renew previously established cell-type-specific promoters with improved performance.
Collapse
Affiliation(s)
- Leo Kojima
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Rokujo
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takanobu Nakazawa
- Department of Bioscience, Tokyo University of Agriculture, Setagaya-ku, Tokyo 156-8502, Japan
| | - Atsushi Kasai
- Systems Neuropharmacology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
- Institute for Datability Science, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
45
|
Federer F, Balsor J, Ingold A, Babcock DP, Dimidschstein J, Angelucci A. Laminar specificity and coverage of viral-mediated gene expression restricted to GABAergic interneurons and their parvalbumin subclass in marmoset primary visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583998. [PMID: 38915672 PMCID: PMC11195055 DOI: 10.1101/2024.03.07.583998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In the mammalian neocortex, inhibition is important for dynamically balancing excitation and shaping the response properties of cells and circuits. The various computational functions of inhibition are thought to be mediated by different inhibitory neuron types of which a large diversity exists in several species. Current understanding of the function and connectivity of distinct inhibitory neuron types has mainly derived from studies in transgenic mice. However, it is unknown whether knowledge gained from mouse studies applies to the non-human primate, the model system closest to humans. The lack of viral tools to selectively access inhibitory neuron types has been a major impediment to studying their function in the primate. Here, we have thoroughly validated and characterized several recently-developed viral vectors designed to restrict transgene expression to GABAergic cells or their parvalbumin (PV) subtype, and identified two types that show high specificity and efficiency in marmoset V1. We show that in marmoset V1 AAV-h56D induces transgene expression in GABAergic cells with up to 91-94% specificity and 79% efficiency, but this depends on viral serotype and cortical layer. AAV-PHP.eB-S5E2 induces transgene expression in PV cells across all cortical layers with up to 98% specificity and 86-90% efficiency, depending on layer. Thus, these viral vectors are promising tools for studying GABA and PV cell function and connectivity in the primate cortex.
Collapse
Affiliation(s)
- Frederick Federer
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT
| | - Justin Balsor
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT
| | - Alexander Ingold
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT
- Present address: Department of Electrical Engineering and Computer Science, University of Utah, Salt Lake City, UT
| | - David P. Babcock
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT
- Present address: Stritch School of Medicine, Loyola University, Chicago, IL
| | | | - Alessandra Angelucci
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
46
|
Bredvik K, Liu C, Ryan TA. Characterization of β-Hydroxybutyrate as a Cell Autonomous Fuel for Active Excitatory and Inhibitory Neurons: β-Hydroxybutyrate as a Fuel for Active Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.08.598077. [PMID: 38895313 PMCID: PMC11185772 DOI: 10.1101/2024.06.08.598077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The ketogenic diet is an effective treatment for drug-resistant epilepsy, but the therapeutic mechanisms are poorly understood. Although ketones are able to fuel the brain, it is not known whether ketones are directly metabolized by neurons on a time scale sufficiently rapid to fuel the bioenergetic demands of sustained synaptic transmission. Here, we show that nerve terminals can use the ketone β-hydroxybutyrate in a cell- autonomous fashion to support neurotransmission in both excitatory and inhibitory nerve terminals and that this flexibility relies on Ca2+ dependent upregulation of mitochondrial metabolism. Using a genetically encoded ATP sensor, we show that inhibitory axons fueled by ketones sustain much higher ATP levels under steady state conditions than excitatory axons, but that the kinetics of ATP production following activity are slower when using ketones as fuel compared to lactate/pyruvate for both excitatory and inhibitory neurons.
Collapse
Affiliation(s)
- Kirsten Bredvik
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA, 10065
| | - Charmaine Liu
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
| |
Collapse
|
47
|
Lee AT, Chang EF, Paredes MF, Nowakowski TJ. Large-scale neurophysiology and single-cell profiling in human neuroscience. Nature 2024; 630:587-595. [PMID: 38898291 PMCID: PMC12049086 DOI: 10.1038/s41586-024-07405-0] [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: 07/06/2023] [Accepted: 04/09/2024] [Indexed: 06/21/2024]
Abstract
Advances in large-scale single-unit human neurophysiology, single-cell RNA sequencing, spatial transcriptomics and long-term ex vivo tissue culture of surgically resected human brain tissue have provided an unprecedented opportunity to study human neuroscience. In this Perspective, we describe the development of these paradigms, including Neuropixels and recent brain-cell atlas efforts, and discuss how their convergence will further investigations into the cellular underpinnings of network-level activity in the human brain. Specifically, we introduce a workflow in which functionally mapped samples of human brain tissue resected during awake brain surgery can be cultured ex vivo for multi-modal cellular and functional profiling. We then explore how advances in human neuroscience will affect clinical practice, and conclude by discussing societal and ethical implications to consider. Potential findings from the field of human neuroscience will be vast, ranging from insights into human neurodiversity and evolution to providing cell-type-specific access to study and manipulate diseased circuits in pathology. This Perspective aims to provide a unifying framework for the field of human neuroscience as we welcome an exciting era for understanding the functional cytoarchitecture of the human brain.
Collapse
Affiliation(s)
- Anthony T Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
48
|
Rubenstein JL, Nord AS, Ekker M. DLX genes and proteins in mammalian forebrain development. Development 2024; 151:dev202684. [PMID: 38819455 PMCID: PMC11190439 DOI: 10.1242/dev.202684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
The vertebrate Dlx gene family encode homeobox transcription factors that are related to the Drosophila Distal-less (Dll) gene and are crucial for development. Over the last ∼35 years detailed information has accrued about the redundant and unique expression and function of the six mammalian Dlx family genes. DLX proteins interact with general transcriptional regulators, and co-bind with other transcription factors to enhancer elements with highly specific activity in the developing forebrain. Integration of the genetic and biochemical data has yielded a foundation for a gene regulatory network governing the differentiation of forebrain GABAergic neurons. In this Primer, we describe the discovery of vertebrate Dlx genes and their crucial roles in embryonic development. We largely focus on the role of Dlx family genes in mammalian forebrain development revealed through studies in mice. Finally, we highlight questions that remain unanswered regarding vertebrate Dlx genes despite over 30 years of research.
Collapse
Affiliation(s)
- John L. Rubenstein
- UCSF Department of Psychiatry and Behavioral Sciences, Department of UCSF Weill Institute for Neurosciences, Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alex S. Nord
- Department of Neurobiology, Physiology, and Behavior and Department of Psychiatry and 20 Behavioral Sciences, Center for Neuroscience, University of California Davis, Davis, CA 95618, USA
| | - Marc Ekker
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
49
|
Maturana CJ. Engineered compact pan-neuronal promoter from Alphaherpesvirus LAP2 enhances target gene expression in the mouse brain and reduces tropism in the liver. Gene Ther 2024; 31:335-344. [PMID: 38012300 PMCID: PMC11090813 DOI: 10.1038/s41434-023-00430-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Small promoters capable of driving potent neuron-restricted gene expression are required to support successful brain circuitry and clinical gene therapy studies. However, converting large promoters into functional MiniPromoters, which can be used in vectors with limited capacity, remains challenging. In this study, we describe the generation of a novel version of alphaherpesvirus latency-associated promoter 2 (LAP2), which facilitates precise transgene expression exclusively in the neurons of the mouse brain while minimizing undesired targeting in peripheral tissues. Additionally, we aimed to create a compact neural promoter to facilitate packaging of larger transgenes. Our results revealed that MiniLAP2 (278 bp) drives potent transgene expression in all neurons in the mouse brain, with little to no expression in glial cells. In contrast to the native promoter, MiniLAP2 reduced tropism in the spinal cord and liver. No expression was detected in the kidney or skeletal muscle. In summary, we developed a minimal pan-neuronal promoter that drives specific and robust transgene expression in the mouse brain when delivered intravenously via AAV-PHP.eB vector. The use of this novel MiniPromoter may broaden the range of deliverable therapeutics and improve their safety and efficacy by minimizing the potential for off-target effects.
Collapse
Affiliation(s)
- Carola J Maturana
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
50
|
Yang L, Liu F, Hahm H, Okuda T, Li X, Zhang Y, Kalyanaraman V, Heitmeier MR, Samineni VK. Projection-TAGs enable multiplex projection tracing and multi-modal profiling of projection neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590975. [PMID: 38712231 PMCID: PMC11071495 DOI: 10.1101/2024.04.24.590975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Single-cell multiomic techniques have sparked immense interest in developing a comprehensive multi-modal map of diverse neuronal cell types and their brain wide projections. However, investigating the spatial organization, transcriptional and epigenetic landscapes of brain wide projection neurons is hampered by the lack of efficient and easily adoptable tools. Here we introduce Projection-TAGs, a retrograde AAV platform that allows multiplex tagging of projection neurons using RNA barcodes. By using Projection-TAGs, we performed multiplex projection tracing of the mouse cortex and high-throughput single-cell profiling of the transcriptional and epigenetic landscapes of the cortical projection neurons. Projection-TAGs can be leveraged to obtain a snapshot of activity-dependent recruitment of distinct projection neurons and their molecular features in the context of a specific stimulus. Given its flexibility, usability, and compatibility, we envision that Projection-TAGs can be readily applied to build a comprehensive multi-modal map of brain neuronal cell types and their projections.
Collapse
Affiliation(s)
- Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Fang Liu
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Hannah Hahm
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Takao Okuda
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Xiaoyue Li
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Yufen Zhang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Monique R. Heitmeier
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Vijay K. Samineni
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|