1
|
Sahoo B, Snyder AC. Neural Dynamics in Extrastriate Cortex Underlying False Alarms. J Neurosci 2025; 45:e1733242025. [PMID: 40164510 PMCID: PMC12079754 DOI: 10.1523/jneurosci.1733-24.2025] [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: 09/11/2024] [Revised: 01/14/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025] Open
Abstract
The unfolding of neural population activity can be described as a dynamical system. Stability in the latent dynamics that characterize neural population activity has been linked with consistency in animal behavior, such as motor control or value-based decision-making. However, whether such characteristics of neural dynamics can explain visual perceptual behavior is not well understood. To study this, we recorded V4 populations in two male monkeys engaged in a non-match-to-sample visual change-detection task that required sustained engagement. We measured how the stability in the latent dynamics in V4 might affect monkeys' perceptual behavior. Specifically, we reasoned that unstable sensory neural activity around dynamic attractor boundaries may make animals susceptible to taking incorrect actions when withholding action would have been correct ("false alarms"). We made three key discoveries: (1) greater stability was associated with longer trial sequences; (2) false alarm rate decreased (and response times slowed) when neural dynamics were more stable; and (3) low stability predicted false alarms on a single-trial level, and this relationship depended on the position of the neural activity within the state space, consistent with the latent neural state approaching an attractor boundary. Our results suggest the same outward false alarm behavior can be attributed to two different potential strategies that can be disambiguated by examining neural stability: (1) premeditated false alarms that might lead to greater stability in population dynamics and faster response time and (2) false alarms due to unstable sensory activity consistent with misperception.
Collapse
Affiliation(s)
- Bikash Sahoo
- Brain & Cognitive Sciences, University of Rochester, Rochester, NY 14627
| | | |
Collapse
|
2
|
Han A, Yermalovich A, Najia MAT, Pearson DS, Fujiwara Y, Bolgov M, Kubaczka C, North TE, Lundin V, Orkin S, Daley GQ. RNA Editors Sculpt the Transcriptome During Terminal Erythropoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647020. [PMID: 40236006 PMCID: PMC11996565 DOI: 10.1101/2025.04.03.647020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Selective RNA degradation during terminal erythropoiesis results in a globin-rich transcriptome in mature erythrocytes, but the specific RNA decay pathways remain unknown. We found that deficiency of the terminal uridylyl transferase enzyme Zcchc6 and the 3'-5' exoribonuclease Dis3l2 in mouse models led to fetal and perinatal reticulocytosis, an accumulation of RNA-rich precursors of terminal erythroid cells, suggesting their crucial roles in terminal red cell differentiation. Notably, knockout embryos exhibited persistent high-level expression of Hbb-bh1 globin, the ortholog of human fetal γ- globin. Perturbation of the Zcchc6-Dis3l2 pathway in mice engineered to express the human β-globin locus likewise increased γ -globin levels in fetal erythroid cells, suggesting that globin switching entails post-transcriptional mechanisms of mRNA destabilization in addition to transcriptional down-regulation. We cultured human hematopoietic stem and progenitor cells (HSPCs), performed CRISPR/Cas9-mediated knockout of ZCCHC6 and DIS3L2, and observed accumulation of RNA and elevated γ-globin levels in terminal erythroid cells. Our findings reveal a conserved role for the ZCCHC6/DIS3L2 RNA editors in terminal erythropoiesis and demonstrate a post-transcriptional mechanism for γ- globin gene switching, advancing research into in vitro erythrocyte generation and γ- globin stabilization to ameliorate hemoglobinopathies.
Collapse
|
3
|
Li S, Jun S, Kim Y, Gogineni A, Lee F, Kim CH, Kim UK, Peng AW, Shin JB. A Tonotopic Regulatory Axis Governing Isoform-Specific MYO7A Expression in Cochlear Hair Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647241. [PMID: 40236169 PMCID: PMC11996477 DOI: 10.1101/2025.04.04.647241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
1. Myo7a , a gene mutated in Usher syndrome and non-syndromic deafness, encodes an unconventional myosin essential for hair cell function. Our previous work revealed that cochlear hair cells express distinct Myo7a isoforms with distinct spatial and cell type-specific patterns. The canonical isoform ( Myo7a-C ) and a novel isoform ( Myo7a-N ) are co-expressed in outer hair cells (OHCs) but exhibit opposing tonotopic gradients, while inner hair cells (IHCs) primarily express Myo7a-C . These isoforms arise from distinct transcriptional start sites, indicating separate regulatory inputs. Here, we identify an intronic cis-regulatory element, EnhancerA , essential for tonotopically graded Myo7a expression. EnhancerA deletion reduces MYO7A protein levels, disrupts hair bundle morphogenesis, alters OHC mechanotransduction, and leads to hair cell degeneration and hearing loss. We further identify SIX2, a tonotopically expressed transcription factor that may interact with EnhancerA to regulate Myo7a-N in OHCs. These findings define a cis-trans regulatory axis critical for isoform-specific Myo7a expression and cochlear function.
Collapse
|
4
|
Gourdine JPF, Pacentine IV, Barstad A, Dahl EM, Gregory WT, Wolfe AJ, Sysoeva TA, Karstens L. Complex carbohydrates catabolism capacity of bladder microbiota inhabiting healthy and overactive female bladders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646827. [PMID: 40236069 PMCID: PMC11996462 DOI: 10.1101/2025.04.02.646827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Overactive bladder syndrome (OAB) is a poorly understood symptom complex that affects 40% of females over the age of 40, with clinical features including urinary urgency and incontinence. In addition to inflammation, oxidative stress, nerve damage and reduced blood flow, alterations in the urinary microbiome (urobiome), specifically in bladder bacterial diversity, have been reported to be associated with OAB. Bladder bacteria are members of the urobiome along with viruses, archaea, fungi, and protozoans. The urobiome metabolism, particularly in relationship to host complex sugars (glycans), has been investigated recently in terms of glycosaminoglycan (GAG) utilization. Nevertheless, other urinary free oligosaccharides (FOS) have not yet been explored in both OAB and urobiome contexts. Similarly, a comprehensive search of microbial genes involved in host glycan metabolism in the bladder of adult females with or without OAB has not yet been reported. In this study, we investigated urinary FOS by mass spectrometry in women without OAB (asymptomatic controls), with OAB without incontinence (dry OAB), or with OAB with incontinence (wet OAB or urgency urinary incontinence, UUI). We also questioned the ability of commensal bladder bacteria to digest these FOS and other glycans, using bioinformatic tools to query publicly available bladder genomes isolated from affected and unaffected adult females to identify genes that encode polysaccharide lyases (PL) and glycoside hydrolases (GH). Our results show that FOS are present in a similar level in affected and unaffected controls with a few exceptions: ten FOS were found to differ between the OAB dry groups and either the control (four) or UUI (six) groups. Our results indicate that bladder microbiota from adult females both with and without OAB have the genetic capacity to digest host glycans and dietary sugars with subtle differences. Bladder bacteria isolated from females with OAB possess more GH/PL genes for host mucins, whereas bladder bacteria from controls possess more GH/PL genes for GAG digestion. In the control group, specifically, the genus Streptococcus possessed genes for the PL8 and GH88 enzymes, known to be involved in host GAG digestion. These novel bioinformatic data can enable future biochemical exploration of the urobiome's metabolism toward specific host glycans, such as GAGs, mucins O-glycans and N-glycans.
Collapse
|
5
|
Koh M, Anselmi F, Kaushalya SK, Hernandez DE, Bast WG, Villar PS, Chae H, Davis MB, Teja SS, Qu Z, Gradinaru V, Gupta P, Banerjee A, Albeanu DF. Axially decoupled photo-stimulation and two photon readout ( ADePT) for mapping functional connectivity of neural circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639992. [PMID: 40161637 PMCID: PMC11952351 DOI: 10.1101/2025.02.24.639992] [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: 04/02/2025]
Abstract
All optical physiology in vivo provides a conduit for investigating the function of neural circuits in 3-D. Here, we report a new strategy for flexible, axially-decoupled photo-stimulation and two photon readout (ADePT) of neuronal activity. To achieve axially-contained widefield optogenetic patterned stimulation, we couple a digital micro-mirror device illuminated by a solid-state laser with a motorized holographic diffuser. In parallel, we use multiphoton imaging of neural activity across different z-planes. We use ADePT to analyze the excitatory and inhibitory functional connectivity of the mouse early olfactory system. Specifically, we control the activity of individual input glomeruli on the olfactory bulb surface, and map the ensuing responses of output mitral and tufted cell bodies in deeper layers. This approach identifies cohorts of sister mitral and tufted cells, whose firing is driven by the same parent glomerulus, and also reveals their differential inhibition by other glomeruli. In addition, selective optogenetic activation of glomerular GABAergic/dopaminergic (DAT+) interneurons triggers dense, but spatially heterogeneous suppression of mitral and tufted cell baseline activity and odor responses, further demonstrating specificity in the inhibitory olfactory bulb connectivity. In summary, ADePT enables high-throughput functional connectivity mapping in optically accessible brain regions.
Collapse
Affiliation(s)
- Matthew Koh
- CSHL School for Biological Sciences
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | | | | | | | | | - Pablo S. Villar
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Honggoo Chae
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Martin B. Davis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Sadhu Sai Teja
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Zhe Qu
- California Institute of Technology, Pasadena, CA, 91125
| | | | - Priyanka Gupta
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Arkarup Banerjee
- CSHL School for Biological Sciences
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Dinu F. Albeanu
- CSHL School for Biological Sciences
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| |
Collapse
|
6
|
Ho 何鎮宇 ECY, Newton AJH, Urdapilleta E, Dura-Bernal S, Truccolo W. Downmodulation of Potassium Conductances Induces Epileptic Seizures in Cortical Network Models Via Multiple Synergistic Factors. J Neurosci 2025; 45:e1909232025. [PMID: 39880680 PMCID: PMC11949479 DOI: 10.1523/jneurosci.1909-23.2025] [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: 10/09/2023] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Voltage-gated potassium conductances g K play a critical role not only in normal neural function, but also in many neurological disorders and related therapeutic interventions. In particular, in an important animal model of epileptic seizures, 4-aminopyridine (4-AP) administration is thought to induce seizures by reducing g K in cortex and other brain areas. Interestingly, 4-AP has also been useful in the treatment of neurological disorders such as multiple sclerosis and spinal cord injury, where it is thought to improve action potential propagation in axonal fibers. Here, we examined g K downmodulation in biophysical models of cortical networks that included different neuron types organized in layers, potassium diffusion in interstitial and larger extracellular spaces, and glial buffering. Our findings are fourfold. First, g K downmodulation in pyramidal and fast-spiking inhibitory interneurons led to differential effects, making the latter much more likely to enter depolarization block. Second, both neuron types showed an increase in the duration and amplitude of action potentials, with more pronounced effects in pyramidal neurons. Third, a sufficiently strong g K reduction dramatically increased network synchrony, resulting in seizure-like dynamics. Fourth, we hypothesized that broader action potentials were likely to not only improve their propagation, as in 4-AP therapeutic uses, but also to increase synaptic coupling. Notably, graded-synapses incorporating this effect further amplified network synchronization and seizure-like dynamics. Overall, our findings elucidate different effects that g K downmodulation may have in cortical networks, explaining its potential role in both pathological neural dynamics and therapeutic applications.
Collapse
Affiliation(s)
- Ernest C Y Ho 何鎮宇
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
| | - Adam J H Newton
- Department of Physiology and Pharmacology, State University of New York (SUNY), Downstate Health Sciences University, Brooklyn, New York 11203
| | - Eugenio Urdapilleta
- Centro Atómico Bariloche and Instituto Balseiro, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, R8402AGP Bariloche, Río Negro, Argentina
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York (SUNY), Downstate Health Sciences University, Brooklyn, New York 11203
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York 10962
| | - Wilson Truccolo
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
7
|
Moss EH, Tantry EK, Le E, Chin PS, Ambrosi P, Brandel-Ankrapp KL, Arenkiel BR. Distinct Patterns of PV and SST GABAergic Neuronal Activity in the Basal Forebrain during Olfactory-Guided Behavior in Mice. J Neurosci 2025; 45:e0200242025. [PMID: 39965928 PMCID: PMC11949486 DOI: 10.1523/jneurosci.0200-24.2025] [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: 02/06/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
Sensory perception relies on the flexible detection and interpretation of stimuli across variable contexts, conditions, and behavioral states. The basal forebrain (BF) is a hub for behavioral state regulation, supplying dense cholinergic and GABAergic projections to various brain regions involved in sensory processing. Of GABAergic neurons in the BF, parvalbumin (PV) and somatostatin (SST) subtypes serve opposing roles toward regulating behavioral states. To elucidate the role of BF circuits in sensory-guided behavior, we investigated GABAergic signaling dynamics during odor-guided decision-making in male and female mice. We used fiber photometry to record cell type-specific BF activity during an odor discrimination task and correlated temporal patterns of PV and SST neuronal activity with olfactory task performance. We found that while both PV-expressing and SST-expressing GABAergic neurons were excited during trial initiation, PV neurons were selectively suppressed by reward, whereas SST neurons were excited. Notably, chemogenetic inhibition of BF SST neurons modestly altered decision bias to favor reward seeking, while optogenetic inhibition of BF PV neurons during odor presentations improved discrimination accuracy. Together, these results suggest that the bidirectional activity of GABAergic BF neuron subtypes distinctly influence perception and decision-making during olfactory-guided behavior.
Collapse
Affiliation(s)
- Elizabeth H Moss
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 97030
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
| | - Evelyne K Tantry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 97030
| | - Elaine Le
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 97030
| | - Pey-Shyuan Chin
- Neuroscience Graduate Program, Baylor College of Medicine, Houston, Texas 97030
| | - Priscilla Ambrosi
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239
| | | | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 97030
| |
Collapse
|
8
|
Olalde I, Altena E, Bourgeois Q, Fokkens H, Amkreutz L, Deguilloux MF, Fichera A, Flas D, Gandini F, Kegler JF, Kootker LM, Leijnse K, Kooijmans LL, Lauwerier R, Miller R, Molthof H, Noiret P, Raemaekers DCM, Rivollat M, Smits L, Stewart JR, Anscher TT, Toussaint M, Callan K, Cheronet O, Frost T, Iliev L, Mah M, Micco A, Oppenheimer J, Patterson I, Qiu L, Soos G, Workman JN, Edwards CJ, Lazaridis I, Mallick S, Patterson N, Rohland N, Richards MB, Pinhasi R, Haak W, Pala M, Reich D. Long-term hunter-gatherer continuity in the Rhine-Meuse region was disrupted by local formation of expansive Bell Beaker groups. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644985. [PMID: 40196638 PMCID: PMC11974744 DOI: 10.1101/2025.03.24.644985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The first phase of the ancient DNA revolution painted a broad-brush picture of European Holocene prehistory, whereby 6500-4000 BCE, farmers descending from western Anatolians mixed with local hunter-gatherers resulting in 70-100% ancestry turnover, then 3000-2500 BCE people associated with the Corded Ware complex spread steppe ancestry into north-central Europe. We document an exception to this pattern in the wider Rhine-Meuse area in communities in the wetlands, riverine areas, and coastal areas of the western and central Netherlands, Belgium and western Germany, where we assembled genome-wide data for 109 people 8500-1700 BCE. Here, a distinctive population with high hunter-gatherer ancestry (~50%) persisted up to three thousand years later than in continental European regions, reflecting limited incorporation of females of Early European Farmer ancestry into local communities. In the western Netherlands, the arrival of the Corded Ware complex was also exceptional: lowland individuals from settlements adopting Corded Ware pottery had hardly any steppe ancestry, despite a characteristic early Corded Ware Y-chromosome. The limited influx may reflect the unique ecology of the region's river-dominated landscapes, which were not amenable to wholesale adoption of the early Neolithic type of farming introduced by Linearbandkeramik, making it possible for previously established groups to thrive, and creating a persistent but permeable boundary that allowed transfer of ideas and low-level gene flow. This changed with the formation-through-mixture of Bell Beaker using populations ~2500 BCE by fusion of local Rhine-Meuse people (9-17%) and Corded Ware associated migrants of both sexes. Their expansion from the Rhine-Meuse region then had a disruptive impact across a much wider part of northwest Europe, including Britain where its arrival was the main source of a 90-100% replacement of local Neolithic peoples.
Collapse
Affiliation(s)
- Iñigo Olalde
- BIOMICs Research Group, Department of Zoology and Animal Cell Biology, University of the Basque Country UPV/ EHU, Vitoria-Gasteiz, Spain
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Ikerbasque-Basque Foundation of Science, Bilbao, Spain
| | - Eveline Altena
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Harry Fokkens
- Faculty of Archaeology, Leiden University, Leiden, The Netherlands
| | - Luc Amkreutz
- Faculty of Archaeology, Leiden University, Leiden, The Netherlands
- National Museum of Antiquities, Leiden, The Netherlands
| | | | - Alessandro Fichera
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Damien Flas
- Service de Préhistoire, Université de Liège, Liège, Belgium
- LAMPEA, Aix-Marseille Université, Aix-en-Provence, France
| | - Francesca Gandini
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Jan F Kegler
- Department of Archaeology, Ostfriesische Landschaft, Aurich, Germany
| | - Lisette M Kootker
- Faculty of Science, Department of Earth Sciences - Isotope Archaeology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | - Roel Lauwerier
- Formerly Cultural Heritage Agency of the Netherlands, Ministry of Education, Culture and Science, The Netherlands
| | - Rebecca Miller
- Service de Préhistoire, Université de Liège, Liège, Belgium
| | | | - Pierre Noiret
- Service de Préhistoire, Université de Liège, Liège, Belgium
| | - Daan C M Raemaekers
- University of Groningen, Groningen Institute of Archaeology (GIA), Groningen, The Netherlands
| | - Maïté Rivollat
- PACEA UMR 5199, University of Bordeaux, Pessac, France
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- ArcheOs, Research Laboratory for Biological Anthropology, Department of Archaeology, University of Ghent, Ghent, Belgium
| | - Liesbeth Smits
- Faculty of Humanities, University of Amsterdam, Amsterdam, The Netherlands
| | - John R Stewart
- Faculty of Science and Technology, Bournemouth University, Poole, Dorset, UK
| | | | - Michel Toussaint
- ArcheOs, Research Laboratory for Biological Anthropology, Department of Archaeology, University of Ghent, Ghent, Belgium
| | - Kim Callan
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Olivia Cheronet
- Department of Evolutionary Anthropology, University of Vienna, Vienna, Austria
| | - Trudi Frost
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lora Iliev
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Matthew Mah
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam Micco
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Iris Patterson
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lijun Qiu
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Gregory Soos
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - J Noah Workman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Iosif Lazaridis
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Swapan Mallick
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | | | - Nadin Rohland
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Martin B Richards
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Ron Pinhasi
- Department of Evolutionary Anthropology, University of Vienna, Vienna, Austria
- Human Evolution and Archaeological Sciences, University of Vienna, Vienna, Austria
| | - Wolfgang Haak
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Maria Pala
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - David Reich
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
9
|
Iturria-Medina Y, Poole VN, Zammit AR, Yu L, Tasaki S, Hong JH, Lopes KDP, Batalha C, Ridwan AR, Vialle RA, Sanchez-Rodriguez L, Geddes MR, Abadir P, Ortlund E, De Jager P, Menon V, Beeri MS, Buchman AS, Levin Y, Morgenstern D, Schneider JA, Daouk RK, Wyss-Coray T, Seyfried NT, Arfanakis K, Rosa-Neto P, Wang Y, Bennett DA. Translating the Post-Mortem Brain Multi-Omics Molecular Taxonomy of Alzheimer's Dementia to Living Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644323. [PMID: 40196602 PMCID: PMC11974700 DOI: 10.1101/2025.03.20.644323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Alzheimer's disease (AD) dementia is characterized by significant molecular and phenotypic heterogeneity, which confounds its mechanistic understanding, diagnosis, and effective treatment. In this study, we harness the most comprehensive dataset of paired ante-mortem blood omics, clinical, psychological, and post-mortem brain multi-omics data and neuroimaging to extensively characterize and translate the molecular taxonomy of AD dementia to living individuals. First, utilizing a comprehensive integration of eight complementary molecular layers from brain multi-omics data (N = 1,189), we identified three distinct molecular AD dementia subtypes exhibiting strong associations with cognitive decline, sex, psychological traits, brain morphology, and characterized by specific cellular and molecular drivers involving immune, vascular, and oligodendrocyte precursor cells. Next, in a significant translational effort, we developed predictive models to convert these advanced brain-derived molecular profiles (AD dementia pseudotimes and subtypes) into blood-, MRI- and psychological traits-based markers. The translation results underscore both the promise of these models and the opportunities for further enhancement. Our findings enhance the understanding of AD heterogeneity, underscore the value of multi-scale molecular approaches for elucidating causal mechanisms, and lay the groundwork for the development of novel therapies in living persons that target multi-level brain molecular subtypes of AD dementia.
Collapse
Affiliation(s)
- Yasser Iturria-Medina
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Victoria N. Poole
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Andrea R. Zammit
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Lei Yu
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Shinya Tasaki
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Joon Hwan Hong
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Katia de Paiva Lopes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Caio Batalha
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Abdur Raquib Ridwan
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ricardo A. Vialle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Lazaro Sanchez-Rodriguez
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Maiya Rachel Geddes
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Peter Abadir
- Johns Hopkins University School of Medicine, Baltimore, USA
| | - Eric Ortlund
- Department of Biochemistry at Emory University School of Medicine, Atlanta, USA
| | - Philip De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michal Schnaider Beeri
- Kreiger Klein Alzheimer’s Research Center, Brain Health Institute, Rutgers Health, NJ, USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Yishai Levin
- Israel National Center for Personalized Medicine at Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- Israel National Center for Personalized Medicine at Weizmann Institute of Science, Rehovot, Israel
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montreal Neurological Institute, Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Douglas Hospital Research Centre - Centre intégré universitaire de santé et services sociaux de l’Ouest-de-l’Île-de-Montréal, Verdun, Quebec, Canada
- The Peter O’Donnell Jr. Brain Institute (OBI), University of Texas Southwestern Medical Centre (UTSW). Dallas, TX, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| |
Collapse
|
10
|
Kim DW, Duncan LH, Xu J, Chang M, Sørensen SS, Terrillion CE, Kanold PO, Place E, Blackshaw S. Decoding Gene Networks Controlling Hypothalamic and Prethalamic Neuron Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632449. [PMID: 39829936 PMCID: PMC11741371 DOI: 10.1101/2025.01.10.632449] [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: 01/22/2025]
Abstract
Neuronal subtypes derived from the embryonic hypothalamus and prethalamus regulate many essential physiological processes, yet the gene regulatory networks controlling their development remain poorly understood. Using single-cell RNA- and ATAC-sequencing, we analyzed mouse hypothalamic and prethalamic development from embryonic day 11 to postnatal day 8, profiling 660,000 cells in total. This identified key transcriptional and chromatin dynamics driving regionalization, neurogenesis, and differentiation. This identified multiple distinct neural progenitor populations, as well as gene regulatory networks that control their spatial and temporal identities, and their terminal differentiation into major neuronal subtypes. Integrating these results with large-scale genome-wide association study data, we identified a central role for transcription factors controlling supramammillary hypothalamic development in a broad range of metabolic and cognitive traits. Recurring cross-repressive regulatory relationships were observed between transcription factors that induced prethalamic and tuberal hypothalamic identity on the one hand and mammillary and supramammillary hypothalamic identity on the other. In postnatal animals, Dlx1/2 was found to severely disrupt GABAergic neuron specification in both the hypothalamus and prethalamus, resulting in a loss of inhibition of thalamic neurons, hypersensitivity to cold, and behavioral hyperactivity. By identifying core gene regulatory networks controlling the specification and differentiation of major hypothalamic and prethalamic neuronal cell types, this study provides a roadmap for future efforts aimed at preventing and treating a broad range of homeostatic and cognitive disorders.
Collapse
Affiliation(s)
- Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Leighton H. Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jenny Xu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minzi Chang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara Sejer Sørensen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chantelle E. Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick O. Kanold
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elsie Place
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Rainwater RR, Azevedo-Pouly AC, Waldrip ZJ, Hicks BH, Callais NA, Koss B, Storey AJ, Burdine L, Burdine MS. DNA-PKcs governs LAT-dependent signaling in CD4 + and CD8 + T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641745. [PMID: 40161607 PMCID: PMC11952348 DOI: 10.1101/2025.03.06.641745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Formation of the immune synapse (IS) following T cell antigen recognition includes recruitment of the Linker for Activation of T cells (LAT). Once at the IS, LAT tyrosines are phosphorylated allowing it to serve as a scaffold for formation of the "signalosome", a multiprotein complex that drives TCR signaling. Here, we show that upon T cell activation, DNA dependent protein kinase catalytic subunit (DNA-PKcs) interacts with LAT and localizes to the IS. Inhibition of DNA-PKcs diminishes LAT localization at the IS. We identified two LAT serines phosphorylated by DNA-PKcs, S224 and S241, that impact LAT tyrosine phosphorylation, protein binding, and cytokine production. Using our mouse model designed to delete DNA-PKcs expression within mature CD4 + or CD8 + T cells, we show loss of DNA-PKcs results in T cells unable to control tumor growth or induce allogeneic graft rejection. These data bring to the forefront DNA-PKcs as a pivotal protein in T cell function.
Collapse
|
12
|
Salamzade R, Kottapalli A, Kalan LR. skDER & CiDDER: two scalable approaches for microbial genome dereplication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.09.27.559801. [PMID: 38045253 PMCID: PMC10690176 DOI: 10.1101/2023.09.27.559801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
An abundance of microbial genomes have been sequenced in the past two decades. For fundamental comparative genomic investigations, where the goal is to determine the major gain and loss events shaping the pangenome of a species, it is often unnecessary and computationally onerous to include all available genomes in studies. In addition, over-representation of specific lineages due to sampling and sequencing bias can have undesired effects on evolutionary analyses. To assist users with genomic dereplication, selecting a subset of representative genomes, for downstream comparative genomic investigations, we developed skDER & CiDDER (https://github.com/raufs/skDER). skDER combines recent advances to efficiently estimate average nucleotide identity (ANI) between thousands of microbial genomes with two efficient algorithms for genomic dereplication. Further, CiDDER implements an approach whereby protein clusters are determined across all genomes and genomes are iteratively selected as representatives until a user-defined saturation of the total protein space is achieved. To support ease of use, several auxiliary functionalities are implemented within the two programs, including arguments to: (i) test the number of representative genomes resulting from a variety of clustering parameters, (ii) automate downloading of genomes belonging to a bacterial species or genus by name, (iii) cluster non-representative genomes to their closest representative genomes, and (iv) automatically filter predicted plasmids and phages prior to dereplication. We further assess the effects of filtering mobile genetic elements (MGEs) on ANI and alignment fraction (AF) estimates between pairs of genomes and find that MGEs tend to slightly deflate both metrics in one species.
Collapse
Affiliation(s)
- Rauf Salamzade
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Aamuktha Kottapalli
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Lindsay R Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
13
|
Hong N, Vargo SM, Hatanaka G, Gong Z, Stanis N, Zhou J, Belloir T, Wang RK, Bair W, Chamanzar M, Yazdan-Shahmorad A. Multimodal optical imaging and modulation through Smart Dura in non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640384. [PMID: 40093178 PMCID: PMC11908230 DOI: 10.1101/2025.02.27.640384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
A multimodal neural interface integrating electrical and optical functionalities is a promising tool for recording and manipulating neuronal activity, providing multiscale information with enhanced spatiotemporal resolution. However, most technologies for multimodal implementation are limited in their applications to small animal models and lack the ability to translate to larger brains, such as non-human primates (NHPs). Recently, we have developed a large-scale neural interface for NHPs, Smart Dura, which enables electrophysiological recordings and high optical accessibility. In this paper, we demonstrate the multimodal applications of Smart Dura in NHPs by combining with multiphoton imaging, optical coherence tomography angiography (OCTA), and intrinsic signal optical imaging (ISOI), as well as optical manipulations such as photothrombotic lesioning and optogenetics. Through the transparent Smart Dura, we could obtain fluorescence images down to 200 μm and 550 μm depth using two-photon and three-photon microscopy, respectively. Integrated with simultaneous electrophysiology using the Smart Dura, we could also assess vascular and neural dynamics with OCTA and ISOI, induce ischemic stroke, and apply optogenetic neuromodulation over a wide coverage area of 20 mm diameter. This multimodal interface enables comprehensive investigations of brain dynamics in NHPs, advancing translational neurotechnology for human applications.
Collapse
Affiliation(s)
- Nari Hong
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Sergio Montalvo Vargo
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Gaku Hatanaka
- Washington National Primate Research Center, Seattle, WA, 98195, USA
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Zhaoyu Gong
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Noah Stanis
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Jasmine Zhou
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Tiphaine Belloir
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Ruikang K Wang
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Wyeth Bair
- Washington National Primate Research Center, Seattle, WA, 98195, USA
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Maysamreza Chamanzar
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Carnegie Mellon Neuroscience Institute, Pittsburgh, PA, 15213, USA
| | - Azadeh Yazdan-Shahmorad
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, 98195, USA
- Weill Neurohub
| |
Collapse
|
14
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [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] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
15
|
Hamlin V, Ansaf H, Heffern R, Williams-Simon PA, King EG. Multiple methods for assessing learning and memory in Drosophila melanogaster demonstrates the highly complex, context-dependent genetic underpinnings of cognitive traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640179. [PMID: 40060392 PMCID: PMC11888412 DOI: 10.1101/2025.02.26.640179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Learning and memory are fundamental for an individual to be able to respond to changing stimuli in their environment. Between individuals we see variation in their ability to perform learning and memory tasks, however, it is still largely unknown what genetic factors may impact this variability. To gain better insight to the genetic components impacting variation in learning and memory, we use recombinant inbred lines (RILs) from the Drosophila synthetic population resource (DSPR), a multiparent mapping population exhibiting natural variation in many traits. Using a reward based associative learning and memory assay, we trained flies to associate an odor with a sucrose reward under starvation condition and measured olfactory learning and memory ability in y-mazes for 50 DSPR RILs. While we do not find significant QTLs for olfactory learning or memory, we found suggestive regions that may be contributing to variability in performance when trained to different odors. We provide evidence that performance with specific odors should be considered different phenotypes and introduce new methods for analysis for olfactory y-maze assays with multiple decision points. Additionally, we compare our data to previously collected place learning and memory data to show there is limited correlation in performance outcomes.
Collapse
Affiliation(s)
- Victoria Hamlin
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | - Huda Ansaf
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | - Reiley Heffern
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| | | | - Elizabeth G King
- Division of Biological Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
16
|
Perrine WG, Sauer EL, Love AC, Morris A, Novotny J, DuRant SE. A high-lipid diet leads to greater pathology and lower tolerance during infection. J Exp Biol 2025; 228:JEB249541. [PMID: 39886835 PMCID: PMC11925396 DOI: 10.1242/jeb.249541] [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: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Altered food landscapes contribute importantly to wildlife disease dynamics and may play a critical role in host heterogeneity in disease outcomes through changes in host diet composition. We explored the effects of dietary macronutrient composition on disease pathology and feeding behavior of canaries (Serinus canaria domestica) infected with Mycoplasma gallisepticum (MG). In the first experiment, we provided canaries with isocaloric diets composed of identical ingredients that varied in macronutrient content (high protein or high lipid) then MG- or sham-inoculated birds. In the second experiment, we offered both diets to canaries before and after MG or sham inoculation. In experiment one, high-protein diet birds consumed more food than high-lipid diet birds and experienced a more pronounced decrease in food intake after infection. High-protein diet birds were more tolerant to MG infection, exhibiting reduced pathology when compared with high-lipid diet birds, despite the two treatments having similar levels of MG-specific antibodies and MG loads. When birds had access to both diets, they consumed more of the high-protein diet and experienced pathology for less time than lipid- or protein-restricted birds. These results highlight that macronutrient makeup of the diet can shape vertebrate host tolerance and pathology, which has direct implications for host-pathogen transmission dynamics.
Collapse
Affiliation(s)
- Weston G. Perrine
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Erin L. Sauer
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Ashley C. Love
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
- Department of Biology, Miami University, Miami, OH 45056, USA
| | - Ashley Morris
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Johnathan Novotny
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Sarah E. DuRant
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
17
|
Lalli JL, Bortvin AN, McCoy RC, Werling DM. A T2T-CHM13 recombination map and globally diverse haplotype reference panel improves phasing and imputation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639687. [PMID: 40060455 PMCID: PMC11888259 DOI: 10.1101/2025.02.24.639687] [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: 03/20/2025]
Abstract
The T2T-CHM13 complete human reference genome contains ~200 Mb of newly resolved sequence, improving read mapping and variant calling compared to GRCh38. However, the benefits of using complete reference genomes in other contexts are unclear. Here, we present a reference T2T-CHM13 recombination map and phased haplotype panel derived from 3202 samples from the 1000 Genomes Project (1KGP). Using published long-read based assemblies as a reference-neutral ground truth, we compared our T2T-CHM13 1KGP panel to the previously released GRCh38 1KGP phased callset. We find that alignment to T2T-CHM13 resulted in 38% fewer assembly-discordant genotypes and 16% fewer switch errors. The largest gains in panel accuracy are observed on chromosome X and in the regions flanking disease-causing CNVs. Simons Genome Diversity Project samples were more accurately imputed when using the T2T-CHM13 panel. Our study demonstrates that use of a T2T-native phased haplotype panel improves statistical phasing and imputation for samples from diverse human populations.
Collapse
Affiliation(s)
- Joseph L Lalli
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, United States
| | - Andrew N Bortvin
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
- These authors jointly supervised this work
| | - Donna M Werling
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, United States
- These authors jointly supervised this work
| |
Collapse
|
18
|
Tassara FJ, Barella M, Simó L, Folgueira Serrao MM, Rodríguez-Caron M, Ispizua JI, Ellisman MH, de la Iglesia HO, Ceriani MF, Gargiulo J. Single Objective Light Sheet Microscopy allows high-resolution in vivo brain imaging of Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.06.622263. [PMID: 39574646 PMCID: PMC11580930 DOI: 10.1101/2024.11.06.622263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2025]
Abstract
In vivo imaging of dynamic sub-cellular brain structures in Drosophila melanogaster is key to understanding several phenomena in neuroscience. However, its implementation has been hindered by a trade-off between spatial resolution, speed, photobleaching, phototoxicity, and setup complexity required to access the specific target regions of the small brain of Drosophila . Here, we present a single objective light-sheet microscope, customized for in vivo imaging of adult flies and optimized for maximum resolution. With it, we imaged the axonal projections of small lateral ventral neurons (known as s-LNvs) in intact adult flies. We imaged the plasma membrane, mitochondria, and dense-core vesicles with high spatial resolution up to 370 nm, ten times lower photobleaching than confocal microscopy, lower invasiveness and complexity in sample mounting than alternative light-sheet technologies, and without relying on phototoxic pulsed infrared lasers. This unique set of features paves the way for new long-term, dynamic studies in the brains of living flies.
Collapse
|
19
|
Ghose SL, Eisen JA. Skin microbiomes of frogs vary among individuals and body regions, revealing differences that reflect known patterns of chytrid infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636728. [PMID: 39975414 PMCID: PMC11839087 DOI: 10.1101/2025.02.05.636728] [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: 02/21/2025]
Abstract
The amphibian skin microbiome is an important line of defense against pathogens including the deadly chytrid fungus, Batrachochytrium dendrobatidis (Bd). Intra-species variation in disease susceptibility and intra-individual variation in infection distribution across the skin, therefore, may relate to differences in skin microbiomes. However, characterization of microbiome variation within and among amphibian individuals is needed. We utilized 16S rRNA gene amplicon sequencing to compare microbiomes of ten body regions from nine captive R. sierrae individuals and their tank environments. While frogs harbored distinct microbial communities compared to their tank environments, tank identity was associated with more variation in frog microbiomes than individual frog identity. Within individuals, we detected differences between microbiomes of body regions where Bd infection would be expected compared to regions that infrequently experience infection. Notably, the bacterial families Burkholderiaceae (phylum Proteobacteria) and Rubritaleaceae (phylum Verrucomicrobia) were dominant on frog skin, and the relative abundances of undescribed members of these families were important to describing differences among and within individuals. Two undescribed Burkholderiaceae taxa were found to be putatively Bd-inhibitory, and both showed higher relative abundance on body regions where Bd infection is often localized. These findings highlight the importance of considering intrapopulation and intraindividual heterogeneities, which could provide insights relevant to predicting localized interactions with pathogens.
Collapse
Affiliation(s)
- Sonia L. Ghose
- Genome Center, University of California, Davis, CA, USA
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Jonathan A. Eisen
- Genome Center, University of California, Davis, CA, USA
- Department of Evolution and Ecology, University of California, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, CA, USA
| |
Collapse
|
20
|
O'Geen H, Mihalovits A, Brophy BD, Yang H, Miller MW, Lee CJ, Segal DJ, Tomkova M. De-novo DNA Methylation of Bivalent Promoters Induces Gene Activation through PRC2 Displacement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636872. [PMID: 39975160 PMCID: PMC11839071 DOI: 10.1101/2025.02.07.636872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Promoter DNA methylation is a key epigenetic mark, commonly associated with gene silencing. However, we noticed that a positive association between promoter DNA methylation and expression is surprisingly common in cancer. Here, we use hit-and-run CRISPR/dCas9 epigenome editing to evaluate how deposition of DNA methylation can regulate gene expression dependent on pre-existing chromatin environment. While the predominant effect of DNA methylation in non-bivalent promoters is gene repression, we show that in bivalent promoters this often leads to gene activation. We demonstrate that gain of DNA methylation leads to reduced MTF2 binding and eviction of H3K27me3, a repressive mark that guards bivalent genes against activation. Our cancer patient data analyses reveal that in cancer, this mechanism likely leads to activation of a large group of transcription factors regulating pluripotency, apoptosis, and senescence signalling. In conclusion, our study uncovers an activating role of DNA methylation in bivalent promoters, with broad implications for cancer and development.
Collapse
|
21
|
Herrera-Asmat O, Tong AB, Lin W, Kong T, Valle JRD, Guerra DG, Ebright YW, Ebright RH, Bustamante C. Pleomorphic effects of three small-molecule inhibitors on transcription elongation by Mycobacterium tuberculosis RNA polymerase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637008. [PMID: 39975155 PMCID: PMC11839117 DOI: 10.1101/2025.02.07.637008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The Mycobacterium tuberculosis RNA polymerase (MtbRNAP) is the target of the first-line anti-tuberculosis inhibitor rifampin, however, the emergence of rifampin resistance necessitates the development of new antibiotics. Here, we communicate the first single-molecule characterization of MtbRNAP elongation and its inhibition by three diverse small-molecule inhibitors: N(α)-aroyl-N-aryl-phenylalaninamide (D-IX216), streptolydigin (Stl), and pseudouridimycin (PUM) using high-resolution optical tweezers. Compared to Escherichia coli RNA polymerase (EcoRNAP), MtbRNAP transcribes more slowly, has similar mechanical robustness, and only weakly recognizes E. coli pause sequences. The three small-molecule inhibitors of MtbRNAP exhibit strikingly different effects on transcription elongation. In the presence of D-IX216, which inhibits RNAP active-center bridge-helix motions required for nucleotide addition, the enzyme exhibits transitions between slowly and super-slowly elongating inhibited states. Stl, which inhibits the RNAP trigger-loop motions also required for nucleotide addition, inhibits RNAP primarily by inducing pausing and backtracking. PUM, a nucleoside analog of UTP, in addition to acting as a competitive inhibitor, induces the formation of slowly elongating RNAP inhibited states. Our results indicate that the three classes of small-molecule inhibitors affect the enzyme in distinct ways and show that the combination of Stl and D-IX216, which both target the RNAP bridge helix, has a strong synergistic effect on the enzyme.
Collapse
Affiliation(s)
- Omar Herrera-Asmat
- Jason L. Choy Laboratory of Single-Molecule Biophysics, University of California, Berkeley, CA 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- Laboratorio de Moléculas Individuales, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Av Honorio Delgado 430, San Martin de Porras, Lima 15102, Peru
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, CA 94720
| | - Alexander B Tong
- Jason L. Choy Laboratory of Single-Molecule Biophysics, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, CA 94720
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Wenxia Lin
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, CA 94720
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518000, China
| | - Tiantian Kong
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518000, China
| | - Juan R Del Valle
- Department of Chemistry & Biochemistry, University of Notre Dame, IN 46556, USA
| | - Daniel G Guerra
- Laboratorio de Moléculas Individuales, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Av Honorio Delgado 430, San Martin de Porras, Lima 15102, Peru
| | - Yon W Ebright
- Waksman Institute, Rutgers University, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854
| | - Richard H Ebright
- Waksman Institute, Rutgers University, Piscataway, NJ 08854
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854
| | - Carlos Bustamante
- Jason L. Choy Laboratory of Single-Molecule Biophysics, University of California, Berkeley, CA 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, CA 94720
- Department of Chemistry, University of California, Berkeley, CA 94720
- Department of Physics, University of California, Berkeley, CA 94720
- Biophysics Graduate Group, University of California, Berkeley, CA 94720
- Kavli Energy Nanoscience Institute, University of California, Berkeley, CA 94720
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720
| |
Collapse
|
22
|
Najia MA, Jha DK, Zhang C, Laurent B, Kubaczka C, Markel A, Li C, Morris V, Tompkins A, Hensch L, Qin Y, Chapuy B, Huang YC, Morse M, Marunde MR, Vaidya A, Gillespie ZB, Howard SA, North TE, Dominguez D, Keogh MC, Schlaeger TM, Shi Y, Li H, Shipp MM, Blainey PC, Daley GQ. Heterochromatin fidelity is a therapeutic vulnerability in lymphoma and other human cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635709. [PMID: 39975048 PMCID: PMC11838449 DOI: 10.1101/2025.01.31.635709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Genes involved in the regulation of chromatin structure are frequently disrupted in cancer, contributing to an aberrant transcriptome and phenotypic plasticity. Yet, therapeutics targeting mutant forms of chromatin-modifying enzymes have yielded only modest clinical utility, underscoring the difficulty of targeting the epigenomic underpinnings of aberrant gene regulatory networks. Here, we sought to identify novel epigenetic vulnerabilities in diffuse large B-cell lymphoma (DLBCL). Through phenotypic screens and biochemical analysis, we demonstrated that inhibition of the H3K9 demethylases KDM4A and KDM4C elicits potent, subtype-agnostic cytotoxicity by antagonizing transcriptional networks associated with B-cell identity and epigenetically rewiring heterochromatin. KDM4 demethylases associated with the KRAB zinc finger ZNF587, and their enzymatic inhibition led to DNA replication stress and DNA damage-einduced cGAS-STING activation. Broad surveys of transcriptional data from patients also revealed KDM4 family dysregulation in several other cancer types. To explore this potential therapeutic avenue, we performed high-throughput small molecule screens with H3K9me3 nucleosome substrates and identified novel KDM4 demethylase inhibitors. AI-guided protein-ligand binding predictions suggested diverse modes of action for various small molecule hits. Our findings underscore the relevance of targeting fundamental transcriptional and epigenetic mechanisms for anti-cancer therapy.
Collapse
Affiliation(s)
- Mohamad Ali Najia
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Deepak K. Jha
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Cheng Zhang
- Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55902, USA
| | - Benoit Laurent
- Division of Newborn Medicine and Epigenetics Program, Department of Medicine, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Caroline Kubaczka
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Arianna Markel
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Christopher Li
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Vivian Morris
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Allison Tompkins
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Luca Hensch
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Yue Qin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bjoern Chapuy
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
- Charité, University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Yu-Chung Huang
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Michael Morse
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | | | | | | | | | - Trista E. North
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Dominguez
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Thorsten M. Schlaeger
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
| | - Yang Shi
- Division of Newborn Medicine and Epigenetics Program, Department of Medicine, Boston Children’s Hospital, Boston, MA, 02115, USA
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hu Li
- Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55902, USA
| | - Margaret M. Shipp
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Paul C. Blainey
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - George Q. Daley
- Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115 USA
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
- Lead contact
| |
Collapse
|
23
|
Scott A, Paulson A, Prill C, Kermoade K, Newell B, Eckenwiler EA, Lemos JC, Richard JM. Ventral Pallidal GABAergic Neurons Drive Consumption in Male, But Not Female, Rats. eNeuro 2025; 12:ENEURO.0245-24.2025. [PMID: 39809537 PMCID: PMC11794971 DOI: 10.1523/eneuro.0245-24.2025] [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: 06/07/2024] [Revised: 11/05/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Food intake is controlled by multiple converging signals: hormonal signals that provide information about energy homeostasis and hedonic and motivational aspects of food and food cues that can drive nonhomeostatic or "hedonic" feeding. The ventral pallidum (VP) is a brain region implicated in the hedonic and motivational impact of food and food cues, as well as consumption of rewards. Disinhibition of VP neurons has been shown to generate intense hyperphagia, or overconsumption. While VP GABA neurons have been implicated in cue-elicited reward-seeking and motivation, the role of these neurons in the hyperphagia resulting from VP activation remains unclear. Here, we used designer receptors exclusively activated by designer drugs to activate VP GABA neurons in nonrestricted male and female rats during chow and sucrose consumption. We found that activation of VP GABA neurons increases consumption of chow and sucrose in male rats, but not female rats. Together, these findings suggest that activation of VP GABA neurons can stimulate consumption of routine or highly palatable rewards selectively in male rats.
Collapse
Affiliation(s)
- Alexandra Scott
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Anika Paulson
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Collin Prill
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Klaiten Kermoade
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
- Molecular Pharmacological and Therapeutics, University of Minnesota, Minneapolis, Minnesota
| | - Bailey Newell
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Elizabeth A Eckenwiler
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Julia C Lemos
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Jocelyn M Richard
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
24
|
Bertaccini GA, Casanellas I, Evans EL, Nourse JL, Dickinson GD, Liu G, Seal S, Ly AT, Holt JR, Wijerathne TD, Yan S, Hui EE, Lacroix JJ, Panicker MM, Upadhyayula S, Parker I, Pathak MM. Visualizing PIEZO1 Localization and Activity in hiPSC-Derived Single Cells and Organoids with HaloTag Technology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.22.573117. [PMID: 38187535 PMCID: PMC10769387 DOI: 10.1101/2023.12.22.573117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
PIEZO1 is critical to numerous physiological processes, transducing diverse mechanical stimuli into electrical and chemical signals. Recent studies underscore the importance of visualizing endogenous PIEZO1 activity and localization to understand its functional roles. To enable physiologically and clinically relevant studies on human PIEZO1, we genetically engineered human induced pluripotent stem cells (hiPSCs) to express a HaloTag fused to endogenous PIEZO1. Combined with advanced imaging, our chemogenetic platform allows precise visualization of PIEZO1 localization dynamics in various cell types. Furthermore, the PIEZO1-HaloTag hiPSC technology facilitates the non-invasive monitoring of channel activity across diverse cell types using Ca2+-sensitive HaloTag ligands, achieving temporal resolution approaching that of patch clamp electrophysiology. Finally, we used lightsheet imaging of hiPSC-derived neural organoids to achieve molecular scale imaging of PIEZO1 in three-dimensional tissue organoids. Our advances offer a novel platform for studying PIEZO1 mechanotransduction in human cells and tissues, with potential for elucidating disease mechanisms and targeted therapeutic development.
Collapse
Affiliation(s)
- Gabriella A Bertaccini
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Ignasi Casanellas
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Elizabeth L Evans
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - George D Dickinson
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Gaoxiang Liu
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Sayan Seal
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Alan T Ly
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Tharaka D Wijerathne
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shijun Yan
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Elliot E Hui
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Jerome J Lacroix
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Mitradas M Panicker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Srigokul Upadhyayula
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| |
Collapse
|
25
|
Lopez JA, McKeithen-Mead S, Shi H, Nguyen TH, Huang KC, Good BH. Abundance measurements reveal the balance between lysis and lysogeny in the human gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.614587. [PMID: 39386523 PMCID: PMC11463441 DOI: 10.1101/2024.09.27.614587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The human gut contains diverse communities of bacteriophage, whose interactions with the broader microbiome and potential roles in human health are only beginning to be uncovered. Here, we combine multiple types of data to quantitatively estimate gut phage population dynamics and lifestyle characteristics in human subjects. Unifying results from previous studies, we show that an average human gut contains a low ratio of phage particles to bacterial cells (~1:100), but a much larger ratio of phage genomes to bacterial genomes (~4:1), implying that most gut phage are effectively temperate (e.g., integrated prophage, phage-plasmids, etc.). By integrating imaging and sequencing data with a generalized model of temperate phage dynamics, we estimate that phage induction and lysis occurs at a low average rate (~0.001-0.01 per bacterium per day), imposing only a modest fitness burden on their bacterial hosts. Consistent with these estimates, we find that the phage composition of a diverse synthetic community in gnotobiotic mice can be quantitatively predicted from bacterial abundances alone, while still exhibiting phage diversity comparable to native human microbiomes. These results provide a foundation for interpreting existing and future studies on links between the gut virome and human health.
Collapse
Affiliation(s)
- Jamie A. Lopez
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Saria McKeithen-Mead
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Handuo Shi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taylor H. Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Benjamin H. Good
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Department of Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Chun JH, Lim BS, Roy S, Walsh MJ, Abhiraman GC, Zhangxu K, Atajanova T, Revach OY, Clark EC, Li P, Palin CA, Khanna A, Tower S, Kureshi R, Hoffman MT, Sharova T, Lawless A, Cohen S, Boland GM, Nguyen T, Peprah F, Tello JG, Liu SY, Kim CJ, Shin H, Quijano-Rubio A, Jude KM, Gerben S, Murray A, Heine P, DeWitt M, Ulge UY, Carter L, King NP, Silva DA, Kueh HY, Kalia V, Sarkar S, Jenkins RW, Garcia KC, Leonard WJ, Dougan M, Dougan SK, Baker D. Potent antitumor activity of a designed interleukin-21 mimic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.626481. [PMID: 39677740 PMCID: PMC11643023 DOI: 10.1101/2024.12.06.626481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Long-standing goals of cancer immunotherapy are to activate cytotoxic antitumor T cells across a broad range of affinities while dampening suppressive regulatory T (Treg) cell responses, but current approaches achieve these goals with limited success. Here, we report a de novo IL-21 mimic, 21h10, designed to have augmented stability and high signaling potency in both humans and mice. In multiple animal models and in ex vivo human melanoma patient derived organotypic tumor spheroids (PDOTS), 21h10 showed robust antitumor activity. 21h10 generates significantly prolonged STAT signaling in vivo compared with native IL-21, and has considerably stronger anti-tumor activity. Toxicities associated with systemic administration of 21h10 could be mitigated by TNFα blockade without compromising antitumor efficacy. In the tumor microenvironment, 21h10 induced highly cytotoxic antitumor T cells from clonotypes with a range of affinities for endogenous tumor antigens, robustly expanding low-affinity cytotoxic T cells and driving high expression of interferon-𝛾 (IFN-𝛾) and granzyme B compared to native IL-21, while increasing the frequency of IFN-𝛾 + Th1 cells and reducing that of Foxp3 + Tregs. As 21h10 has full human/mouse cross-reactivity, high stability and potency, and potentiates low-affinity antitumor responses, it has considerable translational potential.
Collapse
|
27
|
Arutyunyan A, Seuma M, Faure AJ, Bolognesi B, Lehner B. Massively parallel genetic perturbation reveals the energetic architecture of an amyloid beta nucleation reaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604935. [PMID: 39091732 PMCID: PMC11291115 DOI: 10.1101/2024.07.24.604935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Amyloid protein aggregates are pathological hallmarks of more than fifty human diseases but how soluble proteins nucleate to form amyloids is poorly understood. Here we use combinatorial mutagenesis, a kinetic selection assay, and machine learning to massively perturb the energetics of the nucleation reaction of amyloid beta (Aβ42), the protein that aggregates in Alzheimer's disease. In total, we quantify the nucleation rates of >140,000 variants of Aβ42. This allows us to accurately quantify the changes in reaction activation energy for all possible amino acid substitutions in a protein for the first time and, in addition, to quantify >600 energetic interactions between mutations. The data reveal the simple and interpretable genetic architecture of an amyloid nucleation reaction. Strikingly, strong energetic couplings are rare and identify a subset of structural contacts in mature fibrils. Together with the activation energy changes, this strongly suggests that the Aβ42 nucleation reaction transition state is structured in a short C-terminal region, providing a structural model for the reaction that may initiate Alzheimer's disease. We believe this approach can be widely applied to probe the energetics and transition state structures of protein reactions.
Collapse
Affiliation(s)
| | - Mireia Seuma
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10-12, 08028, Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Andre J. Faure
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Current address: ALLOX, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Benedetta Bolognesi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10-12, 08028, Barcelona, Spain
| | - Ben Lehner
- Wellcome Sanger Institute, Cambridge, UK
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
28
|
Ochana BL, Nudelman D, Cohen D, Peretz A, Piyanzin S, Gal O, Horn A, Loyfer N, Varshavsky M, Raisch R, Shapiro I, Friedlander Y, Hochner H, Glaser B, Dor Y, Kaplan T, Shemer R. Time is encoded by methylation changes at clustered CpG sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626674. [PMID: 39677642 PMCID: PMC11642928 DOI: 10.1101/2024.12.03.626674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Age-dependent changes in DNA methylation allow chronological and biological age inference, but the underlying mechanisms remain unclear. Using ultra-deep sequencing of >300 blood samples from healthy individuals, we show that age-dependent DNA methylation changes are regional and occur at multiple adjacent CpG sites, either stochastically or in a coordinated block-like manner. Deep learning analysis of single-molecule patterns in two genomic loci achieved accurate age prediction with a median error of 1.46-1.7 years on held-out human blood samples, dramatically improving current epigenetic clocks. Factors such as gender, BMI, smoking and other measures of biological aging do not affect chronological age inference. Longitudinal 10-year samples revealed that early deviations from epigenetic age are maintained throughout life and subsequent changes faithfully record time. Lastly, the model inferred chronological age from as few as 50 DNA molecules, suggesting that age is encoded by individual cells. Overall, DNA methylation changes in clustered CpG sites illuminate the principles of time measurement by cells and tissues, and facilitate medical and forensic applications.
Collapse
Affiliation(s)
- Bracha-Lea Ochana
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Daniel Nudelman
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Daniel Cohen
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ayelet Peretz
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Sheina Piyanzin
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ofer Gal
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amit Horn
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Netanel Loyfer
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Miri Varshavsky
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Ron Raisch
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Ilona Shapiro
- Braun School of Public Health, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yechiel Friedlander
- Braun School of Public Health, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hagit Hochner
- Braun School of Public Health, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Glaser
- Dept. of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Yuval Dor
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tommy Kaplan
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Israel
| | - Ruth Shemer
- Dept. of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
29
|
Vogler NW, Chen R, Virkler A, Tu VY, Gottfried JA, Geffen MN. Direct Piriform-to-Auditory Cortical Projections Shape Auditory-Olfactory Integration. J Neurosci 2024; 44:e1140242024. [PMID: 39510831 PMCID: PMC11622214 DOI: 10.1523/jneurosci.1140-24.2024] [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: 06/14/2024] [Revised: 09/12/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
In a real-world environment, the brain must integrate information from multiple sensory modalities, including the auditory and olfactory systems. However, little is known about the neuronal circuits governing how odors influence and modulate sound processing. Here, we investigated the mechanisms underlying auditory-olfactory integration using anatomical, electrophysiological, and optogenetic approaches, focusing on the auditory cortex as a key locus for cross-modal integration. First, retrograde and anterograde viral tracing strategies revealed a direct projection from the piriform cortex to the auditory cortex. Next, using in vivo electrophysiological recordings of neuronal activity in the auditory cortex of awake male or female mice, we found that odors modulate auditory cortical responses to sound. Finally, we used in vivo optogenetic manipulations during electrophysiology to demonstrate that olfactory modulation in the auditory cortex, specifically, odor-driven enhancement of sound responses, depends on direct input from the piriform cortex. Together, our results identify a novel role of piriform-to-auditory cortical circuitry in shaping olfactory modulation in the auditory cortex, shedding new light on the neuronal mechanisms underlying auditory-olfactory integration.
Collapse
Affiliation(s)
- Nathan W Vogler
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Ruoyi Chen
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Alister Virkler
- Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Violet Y Tu
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Jay A Gottfried
- Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Maria N Geffen
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
- Neurology, Perelman School of Medicine, University of Pennsylvania
- Neuroscience, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
30
|
Kenny TC, Birsoy K. Mitochondria and Cancer. Cold Spring Harb Perspect Med 2024; 14:a041534. [PMID: 38692736 PMCID: PMC11610758 DOI: 10.1101/cshperspect.a041534] [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: 05/03/2024]
Abstract
Mitochondria are semiautonomous organelles with diverse metabolic and cellular functions including anabolism and energy production through oxidative phosphorylation. Following the pioneering observations of Otto Warburg nearly a century ago, an immense body of work has examined the role of mitochondria in cancer pathogenesis and progression. Here, we summarize the current state of the field, which has coalesced around the position that functional mitochondria are required for cancer cell proliferation. In this review, we discuss how mitochondria influence tumorigenesis by impacting anabolism, intracellular signaling, and the tumor microenvironment. Consistent with their critical functions in tumor formation, mitochondria have become an attractive target for cancer therapy. We provide a comprehensive update on the numerous therapeutic modalities targeting the mitochondria of cancer cells making their way through clinical trials.
Collapse
Affiliation(s)
- Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
31
|
Wongtrakun J, Zhou SH, Bellgrove MA, Chong TTJ, Coxon JP. The Effect of Congruent versus Incongruent Distractor Positioning on Electrophysiological Signals during Perceptual Decision-Making. J Neurosci 2024; 44:e2079232024. [PMID: 39299801 PMCID: PMC11551889 DOI: 10.1523/jneurosci.2079-23.2024] [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: 11/05/2023] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
Key event-related potentials (ERPs) of perceptual decision-making such as centroparietal positivity (CPP) elucidate how evidence is accumulated toward a given choice. Furthermore, this accumulation can be impacted by visual target selection signals such as the N2 contralateral (N2c). How these underlying neural mechanisms of perceptual decision-making are influenced by the spatial congruence of distractors relative to target stimuli remains unclear. Here, we used electroencephalography (EEG) in humans of both sexes to investigate the effect of distractor spatial congruency (same vs different hemifield relative to targets) on perceptual decision-making. We confirmed that responses for perceptual decisions were slower for spatially incongruent versus congruent distractors of high salience. Similarly, markers of target selection (N2c peak amplitude) and evidence accumulation (CPP slope) were found to be lower when distractors were spatially incongruent versus congruent. To evaluate the effects of congruency further, we applied drift diffusion modeling to participant responses, which showed that larger amplitudes of both ERPs were correlated with shorter nondecision times when considering the effect of congruency. The modeling also suggested that congruency's effect on behavior occurred prior to and during evidence accumulation when considering the effects of the N2c peak and CPP slope. These findings point to spatially incongruent distractors, relative to congruent distractors, influencing decisions as early as the initial sensory processing phase and then continuing to exert an effect as evidence is accumulated throughout the decision-making process. Overall, our findings highlight how key electrophysiological signals of perceptual decision-making are influenced by the spatial congruence of target and distractor.
Collapse
Affiliation(s)
- Jaeger Wongtrakun
- School of Psychological Sciences, Monash University, Melbourne, 3800 Victoria, Australia
| | - Shou-Han Zhou
- School of Psychological Sciences, Monash University, Melbourne, 3800 Victoria, Australia
- School of Engineering, Cardiff University, Cardiff CF24 3AA, United Kingdom
| | - Mark A Bellgrove
- School of Psychological Sciences, Monash University, Melbourne, 3800 Victoria, Australia
| | - Trevor T-J Chong
- School of Psychological Sciences, Monash University, Melbourne, 3800 Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, 3004 Victoria, Australia
- Department of Clinical Neurosciences, St Vincent's Hospital, Melbourne, 3065 Victoria, Australia
| | - James P Coxon
- School of Psychological Sciences, Monash University, Melbourne, 3800 Victoria, Australia
| |
Collapse
|
32
|
Wang Q, Su T, Cheng F, Zhou S, Liu X, Wang M, Xu Y, Tang R, Liao S, Dailey J, Xiao G, Yang C, Wen H, Zheng W, Wen B, Tyc KM, Liu J, Sun D, Wang S, Zhu G. Proteolysis-targeting vaccines (PROTAVs) for robust combination immunotherapy of melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.01.616069. [PMID: 39574605 PMCID: PMC11580958 DOI: 10.1101/2024.10.01.616069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Protein/peptide subunit vaccines are promising to promote the tumor therapeutic efficacy of immune checkpoint blockade (ICB). However, current protein/peptide vaccines elicit limited antitumor T cell responses, leading to suboptimal therapeutic efficacy. Here, we present proteolysis-targeting vaccines (PROTAVs) that facilitate antigen proteolytic processing and cross-presentation to potentiate T cell responses for robust ICB combination immunotherapy of melanoma. PROTAVs are modular conjugates of protein/peptide antigens, E3 ligase-binding ligands, and linkers. In antigen-presenting cells (APCs), PROTAVs bind to E3 ligases to rapidly ubiquitinate PROTAV antigens, facilitating antigen proteolytic processing by proteasome, and thereby promoting antigen cross-presentation to T cells and potentiating CD8+ T cell responses. We developed a melanoma PROTAV using a tandem peptide of trivalent melanoma-associated antigens. Co-delivered by lipid nanoparticles (LNPs) with bivalent immunostimulant adjuvants, this PROTAV promotes the quantity and quality of melanoma-specific CD8+ T cells in mice. Further, combining PROTAV and ICB ameliorates the immunosuppressive melanoma microenvironment. As a result, PROTAV and ICB combination enhances melanoma complete regression rates and eradicated 100% large Braf V600E melanoma without recurrence in syngeneic mice. PROTAVs hold the potential for robust tumor combination immunotherapy.
Collapse
Affiliation(s)
- Qiyan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ting Su
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Furong Cheng
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shurong Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mi Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - You Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ri Tang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shimiao Liao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jordan Dailey
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guolan Xiao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunpeng Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hanning Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weijia Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarzyna M Tyc
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinze Liu
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Bioinnovations in Brain Cancer, Biointerfaces Institute; Center for RNA Biomedicine. University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
33
|
Herstel LJ, Wierenga CJ. Distinct Modulation of I h by Synaptic Potentiation in Excitatory and Inhibitory Neurons. eNeuro 2024; 11:ENEURO.0185-24.2024. [PMID: 39406481 PMCID: PMC11574699 DOI: 10.1523/eneuro.0185-24.2024] [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: 04/29/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
Selective modifications in the expression or function of dendritic ion channels regulate the propagation of synaptic inputs and determine the intrinsic excitability of a neuron. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open upon membrane hyperpolarization and conduct a depolarizing inward current (I h). HCN channels are enriched in the dendrites of hippocampal pyramidal neurons where they regulate the integration of synaptic inputs. Synaptic plasticity can bidirectionally modify dendritic HCN channels in excitatory neurons depending on the strength of synaptic potentiation. In inhibitory neurons, however, the dendritic expression and modulation of HCN channels are largely unknown. In this study, we systematically compared the modulation of I h by synaptic potentiation in hippocampal CA1 pyramidal neurons and stratum radiatum (sRad) interneurons in mouse organotypic cultures. I h properties were similar in inhibitory and excitatory neurons and contributed to resting membrane potential and action potential firing. We found that in sRad interneurons, HCN channels were downregulated after synaptic plasticity, irrespective of the strength of synaptic potentiation. This suggests differential regulation of I h in excitatory and inhibitory neurons, possibly signifying their distinct role in network activity.
Collapse
Affiliation(s)
- Lotte J Herstel
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| |
Collapse
|
34
|
Beckwith KS, Brunner A, Morero NR, Jungmann R, Ellenberg J. Nanoscale 3D DNA tracing reveals the mechanism of self-organization of mitotic chromosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620625. [PMID: 39554202 PMCID: PMC11565811 DOI: 10.1101/2024.10.28.620625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
How genomic DNA is folded during cell division to form the characteristic rod-shaped mitotic chromosomes essential for faithful genome inheritance is a long-standing open question in biology. Here, we use nanoscale DNA-tracing in single dividing cells to directly visualize how the 3D fold of genomic DNA changes during mitosis, at scales from single loops to entire chromosomes. Our structural analysis reveals a characteristic genome scaling minimum at 6-8 Mbp in mitosis. Combined with data-driven modeling and molecular perturbations, we can show that very large and strongly overlapping loops formed by Condensins are the fundamental structuring principle of mitotic chromosomes. These loops compact chromosomes locally and globally to the limit set by chromatin self-repulsion. The characteristic length, density and increasingly overlapping structure of mitotic loops we observe in 3D, fully explain how the rod-shaped mitotic chromosome structure emerges by self-organization during cell division.
Collapse
Affiliation(s)
- Kai Sandvold Beckwith
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
- Dept. Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andreas Brunner
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
- Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | | | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
35
|
Tu Li AZ, LiWang A, Subramaniam AB. Insights into a clock's fidelity through vesicular encapsulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.617916. [PMID: 39463922 PMCID: PMC11507718 DOI: 10.1101/2024.10.13.617916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The single-celled cyanobacterium, Synechococcus elongatus , generates circadian rhythms with exceptional fidelity and synchrony despite their femtoliter volumes. Here, we explore the mechanistic aspects of this fidelity, by reconstituting the KaiABC post-translational oscillator (PTO) in cell-mimetic giant vesicles (GUVs) under well-defined conditions in vitro . PTO proteins were encapsulated with a coefficient of variation that closely matched protein variations observed in live cells. Using fluorescently labeled KaiB and confocal microscopy, we were able to measure circadian rhythms generated by thousands of encapsulated PTOs at the single-vesicle level for several days as a function of protein concentration and GUV size. We find that PTO fidelity decreased with decreasing levels of encapsulated PTO proteins and in smaller GUVs. We also observed that in encapsulated PTOs, a significant fraction of KaiB localized to GUV membranes like it does in cyanobacteria. A mathematical model that uses empirical bulk concentration and stoichiometry limitations suggests that cyanobacteria overcome challenges to fidelity by expressing high levels of PTO proteins along with the CikA and SasA proteins, which buffer stochastic variations in the levels of KaiA and KaiB, respectively. Further, the model suggests that the transcription-translation feedback loop (TTFL) contributes at most a small percentage to the overall fidelity of the cyanobacterial circadian clock under constant conditions but is essential for maintaining phase synchrony. Our results are the first experimental demonstration of populations of synthetic cells that can autonomously keep circadian time. Additionally, the approach of using bulk relationships to understand complex phenomena in cell-like systems could be useful for understanding other collective behavior important in biology, such as liquid-liquid phase separation.
Collapse
|
36
|
Li ZP, Moreau H, Petit JD, Souza-Moraes T, Smokvarska M, Perez-Sancho J, Petrel M, Decoeur F, Brocard L, Chambaud C, Grison M, Paterlini A, Glavier M, Hoornaert L, Joshi AS, Gontier E, Prinz WA, Jaillais Y, Taly A, Campelo F, Caillaud MC, Bayer EM. Plant plasmodesmata bridges form through ER-dependent incomplete cytokinesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.12.571296. [PMID: 39464151 PMCID: PMC11507753 DOI: 10.1101/2023.12.12.571296] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Diverging from conventional cell division models, plant cells undergo incomplete division to generate plasmodesmata communication bridges between daughter cells. While fundamental for plant multicellularity, the molecular events leading to bridge stabilization, as opposed to severing, remain unknown. Using electron tomography, we mapped the transition from cell plate fenestrae to plasmodesmata. We show that the ER connects daughter cells across fenestrae, and as the cell plate matures, fenestrae contract, causing the PM to mold around constricted ER tubes. The ER's presence prevents fenestrae fusion, forming plasmodesmata, while its absence results in closure. The ER-PM tethers MCTP3, 4, and 6 further stabilize nascent plasmodesmata during fenestrae contraction. Genetic deletion in Arabidopsis reduces plasmodesmata formation. Our findings reveal how plants undergo incomplete division to promote intercellular communication. One-Sentence Summary The ER is important for stabilizing nascent plasmodesmata, a process integral to incomplete cytokinesis in plants.
Collapse
|
37
|
Khazali MF, Brandt A, Reinacher PC, Kahana M, Jacobs J, Schulze-Bonhage A, Kunz L. A preserved neural code for temporal order between memory formation and recall in the human medial temporal lobe. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.618011. [PMID: 39416044 PMCID: PMC11482969 DOI: 10.1101/2024.10.12.618011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Temporal memory enables us to remember the temporal order of events happening in our life. The human medial temporal lobe (MTL) appears to contain neural representations supporting temporal memory formation, but the cellular mechanisms that preserve temporal order information for recall are largely unknown. Here, we examined whether human MTL neuronal activity represents the temporal position of events during memory formation and recall, using invasive single and multi-unit recordings in human epilepsy patients (n = 19). Participants freely navigated a virtual environment in order to explore and remember locations and temporal positions of objects. During each exploration period, they sequentially encountered two or three different objects, placed in different locations. This allowed us to examine single- and multi-unit neuronal firing rates (FR) as a function of the temporal position the objects were presented in. We found that a significant number of multi-units and single-units in various MTL regions including the hippocampus showed selectivity to the temporal position of objects during the exploration period. During recall, patients were asked to indicate which one of two objects from the same trial was found latter. Neural firing rates during recall showed a selectivity supporting recall of temporal positions. Interestingly, most of the selective single-units that stayed selective during encoding and recall preserved their temporal position preference. Our results thus suggest that neuronal activity in the human MTL contains a preserved neural code for temporal order in memory formation and recall.
Collapse
Affiliation(s)
- Mohammad Farhan Khazali
- Epilepsy Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Armin Brandt
- Epilepsy Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter C. Reinacher
- Department of Stereotactic and Functional Neurosurgery, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Fraunhofer Institute for Laser Technology, Aachen, Germany
| | - Michael Kahana
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua Jacobs
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Andreas Schulze-Bonhage
- Epilepsy Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Kunz
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
38
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585262. [PMID: 38559190 PMCID: PMC10980014 DOI: 10.1101/2024.03.17.585262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, the most common neurodegenerative disorder, remain incompletely understood. To define relevant pathways, we integrated the results of an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila with human and Drosophila Alzheimer's disease-associated multi-omics. We measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. We used a network optimization approach to integrate these data with previously published Alzheimer's disease multi-omic data. We computationally predicted and experimentally demonstrated how HNRNPA2B1 and MEPCE enhance tau-mediated neurotoxicity. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: Stephen and Denise Adams Center of Yale School of Medicine, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
39
|
Hsieh KL, Chu Y, Li X, Pilié PG, Dai Y. scEMB: Learning context representation of genes based on large-scale single-cell transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614685. [PMID: 39386549 PMCID: PMC11463607 DOI: 10.1101/2024.09.24.614685] [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
Background The rapid advancement of single-cell transcriptomic technologies has led to the curation of millions of cellular profiles, providing unprecedented insights into cellular heterogeneity across various tissues and developmental stages. This growing wealth of data presents an opportunity to uncover complex gene-gene relationships, yet also poses significant computational challenges. Results We present scEMB, a transformer-based deep learning model developed to capture context-aware gene embeddings from large-scale single-cell transcriptomics data. Trained on over 30 million single-cell transcriptomes, scEMB utilizes an innovative binning strategy that integrates data across multiple platforms, effectively preserving both gene expression hierarchies and cell-type specificity. In downstream tasks such as batch integration, clustering, and cell type annotation, scEMB demonstrates superior performance compared to existing models like scGPT and Geneformer. Notably, scEMB excels in silico correlation analysis, accurately predicting gene perturbation effects in CRISPR-edited datasets and microglia state transition, identifying a few known Alzheimer's disease (AD) risks genes in top gene list. Additionally, scEMB offers robust fine-tuning capabilities for domain-specific applications, making it a versatile tool for tackling diverse biological problems such as therapeutic target discovery and disease modeling. Conclusions scEMB represents a powerful tool for extracting biologically meaningful insights from complex gene expression data. Its ability to model in silico perturbation effects and conduct correlation analyses in the embedding space highlights its potential to accelerate discoveries in precision medicine and therapeutic development.
Collapse
Affiliation(s)
- Kang-Lin Hsieh
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Chu
- Department of Radiation Physics, Division of Radiation Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoyang Li
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Patrick G. Pilié
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
40
|
Martello SE, Xia J, Kusunose J, Hacker BC, Mayeaux MA, Lin EJ, Hawkes A, Singh A, Caskey CF, Rafat M. Ultrafast Power Doppler Ultrasound Enables Longitudinal Tracking of Vascular Changes that Correlate with Immune Response After Radiotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.04.552076. [PMID: 37577718 PMCID: PMC10418282 DOI: 10.1101/2023.08.04.552076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Background While immunotherapy shows great promise in patients with triple negative breast cancer, many will not respond to treatment. Radiotherapy has the potential to prime the tumor-immune microenvironment for immunotherapy. However, predicting response is difficult due to tumor heterogeneity across patients, which necessitates personalized medicine strategies that incorporate tumor tracking into the therapeutic approach. Here, we investigated the use of ultrasound (US) imaging of the tumor vasculature to monitor the tumor response to treatment. Methods We utilized ultrafast power doppler US to track the vascular response to radiotherapy over time. We used 4T1 (metastatic) and 67NR (non-metastatic) breast cancer models to determine if US measurements corroborate conventional immunostaining analysis of the tumor vasculature. To evaluate the effects of radiation, tumor volume and vascular index were calculated using US, and the correlation between vascular changes and immune cell infiltration was determined. Results US tumor measurements and the quantified vascular response to radiation were confirmed with caliper measurements and immunostaining, respectively, demonstrating a proof-of-principle method for non-invasive vascular monitoring. Additionally, we found significant infiltration of CD8 + T cells into irradiated tumors 10 days after radiation, which followed a sustained decline in vascular index and an increase in splenic CD8 + T cells that was first observed 1 day post-radiation. Conclusions Our findings reveal that ultrafast power doppler US can evaluate changes in tumor vasculature that are indicative of shifts in the tumor-immune microenvironment. This work may lead to improved patient outcomes through observing and predicting response to therapy.
Collapse
|
41
|
Vogler NW, Chen R, Virkler A, Tu VY, Gottfried JA, Geffen MN. Direct piriform-to-auditory cortical projections shape auditory-olfactory integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602976. [PMID: 39071445 PMCID: PMC11275881 DOI: 10.1101/2024.07.11.602976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In a real-world environment, the brain must integrate information from multiple sensory modalities, including the auditory and olfactory systems. However, little is known about the neuronal circuits governing how odors influence and modulate sound processing. Here, we investigated the mechanisms underlying auditory-olfactory integration using anatomical, electrophysiological, and optogenetic approaches, focusing on the auditory cortex as a key locus for cross-modal integration. First, retrograde and anterograde viral tracing strategies revealed a direct projection from the piriform cortex to the auditory cortex. Next, using in vivo electrophysiological recordings of neuronal activity in the auditory cortex of awake male or female mice, we found that odors modulate auditory cortical responses to sound. Finally, we used in vivo optogenetic manipulations during electrophysiology to demonstrate that olfactory modulation in auditory cortex, specifically, odor-driven enhancement of sound responses, depends on direct input from the piriform cortex. Together, our results identify a novel role of piriform-to-auditory cortical circuitry in shaping olfactory modulation in the auditory cortex, shedding new light on the neuronal mechanisms underlying auditory-olfactory integration.
Collapse
Affiliation(s)
- Nathan W. Vogler
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Ruoyi Chen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Alister Virkler
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Violet Y. Tu
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Jay A. Gottfried
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Maria N. Geffen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
42
|
Salamzade R, Tran PQ, Martin C, Manson AL, Gilmore MS, Earl AM, Anantharaman K, Kalan LR. zol & fai: large-scale targeted detection and evolutionary investigation of gene clusters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.07.544063. [PMID: 37333121 PMCID: PMC10274777 DOI: 10.1101/2023.06.07.544063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Many universally and conditionally important genes are genomically aggregated within clusters. Here, we introduce fai and zol, which together enable large-scale comparative analysis of different types of gene clusters and mobile-genetic elements (MGEs), such as biosynthetic gene clusters (BGCs) or viruses. Fundamentally, they overcome a current bottleneck to reliably perform comprehensive orthology inference at large scale across broad taxonomic contexts and thousands of genomes. First, fai allows the identification of orthologous instances of a query gene cluster of interest amongst a database of target genomes. Subsequently, zol enables reliable, context-specific inference of ortholog groups for individual protein-encoding genes across gene cluster instances. In addition, zol performs functional annotation and computes a variety of evolutionary statistics for each inferred ortholog group. Importantly, in comparison to tools for visual exploration of homologous relationships between gene clusters, zol can scale to thousands of gene cluster instances and produce detailed reports that are easy to digest. To showcase fai and zol, we apply them for: (i) longitudinal tracking of a virus in metagenomes, (ii) discovering novel population-level genetic insights of two common BGCs in the fungal species Aspergillus flavus, and (iii) uncovering large-scale evolutionary trends of a virulence-associated gene cluster across thousands of genomes from a diverse bacterial genus.
Collapse
Affiliation(s)
- Rauf Salamzade
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Patricia Q. Tran
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Freshwater and Marine Science Doctoral Program, University of Wisconsin-Madison, WI, USA
| | - Cody Martin
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Abigail L. Manson
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Michael S. Gilmore
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
| | - Ashlee M. Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Lindsay R. Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
43
|
Aldana A, Sebek M, Ispirova G, Dorantes-Gilardi R, Barabási AL, Loscalzo J, Menichetti G. NetMedPy: A Python package for Large-Scale Network Medicine Screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611537. [PMID: 39569139 PMCID: PMC11577252 DOI: 10.1101/2024.09.05.611537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Network medicine leverages the quantification of information flow within sub-cellular networks to elucidate disease etiology and comorbidity, as well as to predict drug efficacy and identify potential therapeutic targets. However, current Network Medicine toolsets often lack computationally efficient data processing pipelines that support diverse scoring functions, network distance metrics, and null models. These limitations hamper their application in large-scale molecular screening, hypothesis testing, and ensemble modeling. To address these challenges, we introduce NetMedPy, a highly efficient and versatile computational package designed for comprehensive Network Medicine analyses.
Collapse
Affiliation(s)
- Andrés Aldana
- Network Science Institute, Northeastern University, 360 Huntington Ave, 02115, MA, USA
| | - Michael Sebek
- Network Science Institute, Northeastern University, 360 Huntington Ave, 02115, MA, USA
| | - Gordana Ispirova
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, 02115, MA, USA
| | | | - Albert-László Barabási
- Network Science Institute, Northeastern University, 360 Huntington Ave, 02115, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, 02115, MA, USA
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, 02115, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, 02115, MA, USA
| | - Giulia Menichetti
- Network Science Institute, Northeastern University, 360 Huntington Ave, 02115, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, 02115, MA, USA
- Harvard Data Science Initiative, Harvard University, 114 Western Avenue, 02134, MA, USA
| |
Collapse
|
44
|
Bruce R, Weber MA, Bova A, Volkman R, Jacobs C, Sivakumar K, Kim Y, Curtu R, Narayanan N. Complementary cognitive roles for D2-MSNs and D1-MSNs during interval timing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.25.550569. [PMID: 37546735 PMCID: PMC10402049 DOI: 10.1101/2023.07.25.550569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The role of striatal pathways in cognitive processing is unclear. We studied dorsomedial striatal cognitive processing during interval timing, an elementary cognitive task that requires mice to estimate intervals of several seconds and involves working memory for temporal rules as well as attention to the passage of time. We harnessed optogenetic tagging to record from striatal D2-dopamine receptor-expressing medium spiny neurons (D2-MSNs) in the indirect pathway and from D1-dopamine receptor-expressing MSNs (D1-MSNs) in the direct pathway. We found that D2-MSNs and D1-MSNs exhibited distinct dynamics over temporal intervals as quantified by principal component analyses and trial-by-trial generalized linear models. MSN recordings helped construct and constrain a four-parameter drift-diffusion computational model. This model predicted that disrupting either D2-MSNs or D1-MSNs would increase interval timing response times and alter MSN firing. In line with this prediction, we found that optogenetic inhibition or pharmacological disruption of either D2-MSNs or D1-MSNs increased interval timing response times. Pharmacologically disrupting D2-MSNs or D1-MSNs also changed MSN dynamics and degraded trial-by-trial temporal decoding. Together, our findings demonstrate that D2-MSNs and D1-MSNs make complementary contributions to interval timing despite opposing dynamics, implying that striatal direct and indirect pathways work together to shape temporal control of action. These data provide novel insight into basal ganglia cognitive operations beyond movement and have implications for human striatal diseases and therapies targeting striatal pathways.
Collapse
|
45
|
Qi Z, Xue S, Chen J, Zhao W, Johnson K, Wen X, Richard JLC, Zhong S. Genome-Wide Mapping of RNA-Protein Associations via Sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611288. [PMID: 39282297 PMCID: PMC11398515 DOI: 10.1101/2024.09.04.611288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
RNA-protein interactions are crucial for regulating gene expression and cellular functions, with their dysregulation potentially impacting disease progression. Systematically mapping these interactions is resource-intensive due to the vast number of potential RNA and protein interactions. Here, we introduce PRIM-seq (Protein-RNA Interaction Mapping by sequencing), a method for the concurrent de novo identification of RNA-binding proteins (RBPs) and the elucidation of their associated RNAs. PRIM-seq works by converting each RNA-protein pair into a unique chimeric DNA sequence, which is then decoded through DNA sequencing. Applied to two human cell types, PRIM-seq generated a comprehensive human RNA-protein association network (HuRPA), consisting of more than 350,000 RNA-proteins pairs involving approximately 7,000 RNAs and 11,000 proteins. The data revealed an enrichment of previously reported RBPs and RNA-protein interactions within HuRPA. We also identified LINC00339 as a protein-associating non-coding RNA and PHGDH as an RNA-associating protein. Notably, PHGDH interacts with BECN1 and ATF4 mRNAs, suppressing their protein expression and consequently inhibiting autophagy, apoptosis, and neurite outgrowth while promoting cell proliferation. PRIM-seq offers a powerful tool for discovering RBPs and RNA-protein associations, contributing to more comprehensive functional genome annotations.
Collapse
Affiliation(s)
- Zhijie Qi
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Shuanghong Xue
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Junchen Chen
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Wenxin Zhao
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Kara Johnson
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Xingzhao Wen
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
| | | | - Sheng Zhong
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
46
|
Lendemeijer B, Unkel M, Smeenk H, Mossink B, Hijazi S, Gordillo-Sampedro S, Shpak G, Slump DE, van den Hout MCGN, van IJcken WFJ, Bindels EMJ, Hoogendijk WJG, Nadif Kasri N, de Vrij FMS, Kushner SA. Human Pluripotent Stem Cell-Derived Astrocyte Functionality Compares Favorably with Primary Rat Astrocytes. eNeuro 2024; 11:ENEURO.0148-24.2024. [PMID: 39227152 PMCID: PMC11404293 DOI: 10.1523/eneuro.0148-24.2024] [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: 04/03/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024] Open
Abstract
Astrocytes are essential for the formation and maintenance of neural networks. However, a major technical challenge for investigating astrocyte function and disease-related pathophysiology has been the limited ability to obtain functional human astrocytes. Despite recent advances in human pluripotent stem cell (hPSC) techniques, primary rodent astrocytes remain the gold standard in coculture with human neurons. We demonstrate that a combination of leukemia inhibitory factor (LIF) and bone morphogenetic protein-4 (BMP4) directs hPSC-derived neural precursor cells to a highly pure population of astroglia in 28 d. Using single-cell RNA sequencing, we confirm the astroglial identity of these cells and highlight profound transcriptional adaptations in cocultured hPSC-derived astrocytes and neurons, consistent with their further maturation. In coculture with human neurons, multielectrode array recordings revealed robust network activity of human neurons in a coculture with hPSC-derived or rat astrocytes [3.63 ± 0.44 min-1 (hPSC-derived), 2.86 ± 0.64 min-1 (rat); p = 0.19]. In comparison, we found increased spike frequency within network bursts of human neurons cocultured with hPSC-derived astrocytes [56.31 ± 8.56 Hz (hPSC-derived), 24.77 ± 4.04 Hz (rat); p < 0.01], and whole-cell patch-clamp recordings revealed an increase of postsynaptic currents [2.76 ± 0.39 Hz (hPSC-derived), 1.07 ± 0.14 Hz (rat); p < 0.001], consistent with a corresponding increase in synapse density [14.90 ± 1.27/100 μm2 (hPSC-derived), 8.39 ± 0.63/100 μm2 (rat); p < 0.001]. Taken together, we show that hPSC-derived astrocytes compare favorably with rat astrocytes in supporting human neural network activity and maturation, providing a fully human platform for investigating astrocyte function and neuronal-glial interactions.
Collapse
Affiliation(s)
- Bas Lendemeijer
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
- Department of Psychiatry, Columbia University, New York, New York 10027
- Stavros Niarchos Foundation (SNF) Center for Precision Psychiatry & Mental Health, Columbia University, New York, New York 10027
| | - Maurits Unkel
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Hilde Smeenk
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Britt Mossink
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Sara Hijazi
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Sara Gordillo-Sampedro
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Guy Shpak
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Denise E Slump
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Mirjam C G N van den Hout
- Department of Cell Biology, Center for Biomics, Erasmus University Medical Center, Rotterdam 3015AA, The Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Center for Biomics, Erasmus University Medical Center, Rotterdam 3015AA, The Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center, Rotterdam 3015AA, The Netherlands
| | - Witte J G Hoogendijk
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525GA, The Netherlands
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam 3015AA, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam 3015 AA, The Netherlands
- Department of Psychiatry, Columbia University, New York, New York 10027
- Stavros Niarchos Foundation (SNF) Center for Precision Psychiatry & Mental Health, Columbia University, New York, New York 10027
| |
Collapse
|
47
|
Norris MR, Kuo CC, Dunn SS, Kim JR, Becker LJ, Borges G, Thang LV, Parker KE, McCall JG. Mu opioid receptors gate the locus coeruleus pain generator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.20.562785. [PMID: 37961541 PMCID: PMC10634678 DOI: 10.1101/2023.10.20.562785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The locus coeruleus (LC) plays a paradoxical role in chronic pain. Although largely known as a potent source of endogenous analgesia, increasing evidence suggests injury can transform the LC into a chronic pain generator. We sought to clarify the role of this system in pain. Here, we show optogenetic inhibition of LC activity is acutely antinociceptive. Following long-term spared nerve injury, the same LC inhibition is analgesic - further supporting its pain generator function. To identify inhibitory substrates that may naturally serve this function, we turned to endogenous LC mu opioid receptors (LC-MOR). These receptors provide powerful LC inhibition and exogenous activation of LC-MOR is antinociceptive. We therefore hypothesized that endogenous LC-MOR-mediated inhibition is critical to how the LC modulates pain. Using cell type-selective conditional knockout and rescue of LC-MOR receptor signaling, we show these receptors bidirectionally regulate thermal and mechanical hyperalgesia - providing a functional gate on the LC pain generator.
Collapse
Affiliation(s)
- Makenzie R. Norris
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Samantha S. Dunn
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Léa J. Becker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Gustavo Borges
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Loc V. Thang
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Kyle E. Parker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
48
|
Colins Rodriguez A, Perich MG, Miller LE, Humphries MD. Motor Cortex Latent Dynamics Encode Spatial and Temporal Arm Movement Parameters Independently. J Neurosci 2024; 44:e1777232024. [PMID: 39060178 PMCID: PMC11358606 DOI: 10.1523/jneurosci.1777-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The fluid movement of an arm requires multiple spatiotemporal parameters to be set independently. Recent studies have argued that arm movements are generated by the collective dynamics of neurons in motor cortex. An untested prediction of this hypothesis is that independent parameters of movement must map to independent components of the neural dynamics. Using a task where three male monkeys made a sequence of reaching movements to randomly placed targets, we show that the spatial and temporal parameters of arm movements are independently encoded in the low-dimensional trajectories of population activity in motor cortex: each movement's direction corresponds to a fixed neural trajectory through neural state space and its speed to how quickly that trajectory is traversed. Recurrent neural network models show that this coding allows independent control over the spatial and temporal parameters of movement by separate network parameters. Our results support a key prediction of the dynamical systems view of motor cortex, and also argue that not all parameters of movement are defined by different trajectories of population activity.
Collapse
Affiliation(s)
| | - Matt G Perich
- Département de neurosciences, Faculté de médecine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- Québec Artificial Intelligence Institute (Mila), Montreal, Quebec H2S 3H1, Canada
| | - Lee E Miller
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois 60208
| | - Mark D Humphries
- School of Psychology, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
49
|
Rohwer RR, Kirkpatrick M, Garcia SL, Kellom M, McMahon KD, Baker BJ. Bacterial ecology and evolution converge on seasonal and decadal scales. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579087. [PMID: 38370794 PMCID: PMC10871203 DOI: 10.1101/2024.02.06.579087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Ecology and evolution are often viewed as distinct processes, which interact on contemporary time scales in microbiomes. To observe these processes in a natural system, we collected a two-decade, 471-sample freshwater lake time series, creating the longest metagenome dataset to date. Among 2,855 species-representative genomes, diverse species and strains followed cyclical seasonal patterns, and one in five species experienced decadal shifts in strain composition. The most globally abundant freshwater bacterium had constant species-level abundance, but environmental extremes appeared to trigger a shift in strain composition and positive selection of amino acid and nucleic acid metabolism genes. These genes identify organic nitrogen compounds as potential drivers of freshwater responses to global change. Seasonal and long-term strain dynamics could be regarded as ecological processes or equivalently as evolutionary change. Rather than as distinct processes that interact, we propose a conceptualization where ecology and evolution converge along a continuum to better describe change in diverse microbial communities.
Collapse
Affiliation(s)
- Robin R. Rohwer
- The University of Texas at Austin, Department of Integrative Biology, 2415 Speedway #C0930, Austin, TX 78712, USA
| | - Mark Kirkpatrick
- The University of Texas at Austin, Department of Integrative Biology, 2415 Speedway #C0930, Austin, TX 78712, USA
| | - Sarahi L. Garcia
- Carl von Ossietzky Universität Oldenburg, Institute for Chemistry and Biology of the Marine Environment (ICBM), 26129 Oldenburg, Germany
- Stockholm University, Department of Ecology, Environment, and Plant Sciences, Science for Life Laboratory, 106 91 Stockholm, Sweden
| | - Matthew Kellom
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Katherine D. McMahon
- University of Wisconsin-Madison, Department of Bacteriology, 1550 Linden Drive, Madison, WI, 53706, USA
- University of Wisconsin-Madison, Department of Civil and Environmental Engineering, 1550 Linden Drive, Madison, WI, 53706, USA
| | - Brett J. Baker
- The University of Texas at Austin, Department of Integrative Biology, 2415 Speedway #C0930, Austin, TX 78712, USA
- The University of Texas at Austin, Department of Marine Science, Port Aransas, Austin, TX 78373, USA
| |
Collapse
|
50
|
Seo S, Bharmauria V, Schütz A, Yan X, Wang H, Crawford JD. Multiunit Frontal Eye Field Activity Codes the Visuomotor Transformation, But Not Gaze Prediction or Retrospective Target Memory, in a Delayed Saccade Task. eNeuro 2024; 11:ENEURO.0413-23.2024. [PMID: 39054056 PMCID: PMC11373882 DOI: 10.1523/eneuro.0413-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Single-unit (SU) activity-action potentials isolated from one neuron-has traditionally been employed to relate neuronal activity to behavior. However, recent investigations have shown that multiunit (MU) activity-ensemble neural activity recorded within the vicinity of one microelectrode-may also contain accurate estimations of task-related neural population dynamics. Here, using an established model-fitting approach, we compared the spatial codes of SU response fields with corresponding MU response fields recorded from the frontal eye fields (FEFs) in head-unrestrained monkeys (Macaca mulatta) during a memory-guided saccade task. Overall, both SU and MU populations showed a simple visuomotor transformation: the visual response coded target-in-eye coordinates, transitioning progressively during the delay toward a future gaze-in-eye code in the saccade motor response. However, the SU population showed additional secondary codes, including a predictive gaze code in the visual response and retention of a target code in the motor response. Further, when SUs were separated into regular/fast spiking neurons, these cell types showed different spatial code progressions during the late delay period, only converging toward gaze coding during the final saccade motor response. Finally, reconstructing MU populations (by summing SU data within the same sites) failed to replicate either the SU or MU pattern. These results confirm the theoretical and practical potential of MU activity recordings as a biomarker for fundamental sensorimotor transformations (e.g., target-to-gaze coding in the oculomotor system), while also highlighting the importance of SU activity for coding more subtle (e.g., predictive/memory) aspects of sensorimotor behavior.
Collapse
Affiliation(s)
- Serah Seo
- Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, Toronto, Ontario M3J 1P3, Canada
| | - Vishal Bharmauria
- Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, Toronto, Ontario M3J 1P3, Canada
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida 33606
| | - Adrian Schütz
- Department of Neurophysics, Philipps-Universität Marburg, 35032 Marburg, Germany
- Center for Mind, Brain, and Behavior - CMBB, Philipps-Universität Marburg, 35032 Marburg, and Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Xiaogang Yan
- Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, Toronto, Ontario M3J 1P3, Canada
| | - Hongying Wang
- Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, Toronto, Ontario M3J 1P3, Canada
| | - J Douglas Crawford
- Centre for Vision Research and Centre for Integrative and Applied Neuroscience, York University, Toronto, Ontario M3J 1P3, Canada
- Departments of Psychology, Biology, Kinesiology & Health Sciences, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|