TY - JOUR AB - Importance Climate change, pollution, urbanization, socioeconomic inequality, and psychosocial effects of the COVID-19 pandemic have caused massive changes in environmental conditions that affect brain health during the life span, both on a population level as well as on the level of the individual. How these environmental factors influence the brain, behavior, and mental illness is not well known. Observations A research strategy enabling population neuroscience to contribute to identify brain mechanisms underlying environment-related mental illness by leveraging innovative enrichment tools for data federation, geospatial observation, climate and pollution measures, digital health, and novel data integration techniques is described. This strategy can inform innovative treatments that target causal cognitive and molecular mechanisms of mental illness related to the environment. An example is presented of the environMENTAL Project that is leveraging federated cohort data of over 1.5 million European citizens and patients enriched with deep phenotyping data from large-scale behavioral neuroimaging cohorts to identify brain mechanisms related to environmental adversity underlying symptoms of depression, anxiety, stress, and substance misuse. Conclusions and Relevance This research will lead to the development of objective biomarkers and evidence-based interventions that will significantly improve outcomes of environment-related mental illness. AU - Schumann, Gunter AU - Andreassen, Ole A. AU - Banaschewski, Tobias AU - Calhoun, Vince D. AU - Clinton, Nicholas AU - Desrivieres, Sylvane AU - Brandlistuen, Ragnhild Eek AU - Feng, Jianfeng AU - Hese, Soeren AU - Hitchen, Esther AU - Hoffmann, Per AU - Jia, Tianye AU - Jirsa, Viktor AU - Marquand, Andre F. AU - Nees, Frauke AU - Nöthen, Markus M. AU - Novarino, Gaia AU - Polemiti, Elli AU - Ralser, Markus AU - Rapp, Michael AU - Schepanski, Kerstin AU - Schikowski, Tamara AU - Slater, Mel AU - Sommer, Peter AU - Stahl, Bernd Carsten AU - Thompson, Paul M. AU - Twardziok, Sven AU - Van Der Meer, Dennis AU - Walter, Henrik AU - Westlye, Lars ID - 14443 IS - 10 JF - JAMA Psychiatry TI - Addressing global environmental challenges to mental health using population neuroscience: A review VL - 80 ER - TY - JOUR AB - Urban-living individuals are exposed to many environmental factors that may combine and interact to influence mental health. While individual factors of an urban environment have been investigated in isolation, no attempt has been made to model how complex, real-life exposure to living in the city relates to brain and mental health, and how this is moderated by genetic factors. Using the data of 156,075 participants from the UK Biobank, we carried out sparse canonical correlation analyses to investigate the relationships between urban environments and psychiatric symptoms. We found an environmental profile of social deprivation, air pollution, street network and urban land-use density that was positively correlated with an affective symptom group (r = 0.22, Pperm < 0.001), mediated by brain volume differences consistent with reward processing, and moderated by genes enriched for stress response, including CRHR1, explaining 2.01% of the variance in brain volume differences. Protective factors such as greenness and generous destination accessibility were negatively correlated with an anxiety symptom group (r = 0.10, Pperm < 0.001), mediated by brain regions necessary for emotion regulation and moderated by EXD3, explaining 1.65% of the variance. The third urban environmental profile was correlated with an emotional instability symptom group (r = 0.03, Pperm < 0.001). Our findings suggest that different environmental profiles of urban living may influence specific psychiatric symptom groups through distinct neurobiological pathways. AU - Xu, Jiayuan AU - Liu, Nana AU - Polemiti, Elli AU - Garcia-Mondragon, Liliana AU - Tang, Jie AU - Liu, Xiaoxuan AU - Lett, Tristram AU - Yu, Le AU - Nöthen, Markus M. AU - Feng, Jianfeng AU - Yu, Chunshui AU - Marquand, Andre AU - Schumann, Gunter AU - Walter, Henrik AU - Heinz, Andreas AU - Ralser, Markus AU - Twardziok, Sven AU - Vaidya, Nilakshi AU - Serin, Emin AU - Jentsch, Marcel AU - Hitchen, Esther AU - Eils, Roland AU - Taron, Ulrike Helene AU - Schütz, Tatjana AU - Schepanski, Kerstin AU - Banks, Jamie AU - Banaschewski, Tobias AU - Jansone, Karina AU - Christmann, Nina AU - Meyer-Lindenberg, Andreas AU - Tost, Heike AU - Holz, Nathalie AU - Schwarz, Emanuel AU - Stringaris, Argyris AU - Neidhart, Maja AU - Nees, Frauke AU - Siehl, Sebastian AU - A. Andreassen, Ole AU - T. Westlye, Lars AU - Van Der Meer, Dennis AU - Fernandez, Sara AU - Kjelkenes, Rikka AU - Ask, Helga AU - Rapp, Michael AU - Tschorn, Mira AU - Böttger, Sarah Jane AU - Novarino, Gaia AU - Marr, Lena AU - Slater, Mel AU - Viapiana, Guillem Feixas AU - Orosa, Francisco Eiroa AU - Gallego, Jaime AU - Pastor, Alvaro AU - Forstner, Andreas AU - Hoffmann, Per AU - M. Nöthen, Markus AU - J. Forstner, Andreas AU - Claus, Isabelle AU - Miller, Abbi AU - Heilmann-Heimbach, Stefanie AU - Sommer, Peter AU - Boye, Mona AU - Wilbertz, Johannes AU - Schmitt, Karen AU - Jirsa, Viktor AU - Petkoski, Spase AU - Pitel, Séverine AU - Otten, Lisa AU - Athanasiadis, Anastasios Polykarpos AU - Pearmund, Charlie AU - Spanlang, Bernhard AU - Alvarez, Elena AU - Sanchez, Mavi AU - Giner, Arantxa AU - Hese, Sören AU - Renner, Paul AU - Jia, Tianye AU - Gong, Yanting AU - Xia, Yunman AU - Chang, Xiao AU - Calhoun, Vince AU - Liu, Jingyu AU - Thompson, Paul AU - Clinton, Nicholas AU - Desrivieres, Sylvane AU - H. Young, Allan AU - Stahl, Bernd AU - Ogoh, George ID - 13168 JF - Nature Medicine SN - 1078-8956 TI - Effects of urban living environments on mental health in adults VL - 29 ER - TY - JOUR AU - Narzisi, Antonio AU - Halladay, Alycia AU - Masi, Gabriele AU - Novarino, Gaia AU - Lord, Catherine ID - 14455 JF - Frontiers in Psychiatry TI - Tempering expectations: Considerations on the current state of stem cells therapy for autism treatment VL - 14 ER - TY - JOUR AB - Three-dimensional (3D) reconstruction of living brain tissue down to an individual synapse level would create opportunities for decoding the dynamics and structure–function relationships of the brain’s complex and dense information processing network; however, this has been hindered by insufficient 3D resolution, inadequate signal-to-noise ratio and prohibitive light burden in optical imaging, whereas electron microscopy is inherently static. Here we solved these challenges by developing an integrated optical/machine-learning technology, LIONESS (live information-optimized nanoscopy enabling saturated segmentation). This leverages optical modifications to stimulated emission depletion microscopy in comprehensively, extracellularly labeled tissue and previous information on sample structure via machine learning to simultaneously achieve isotropic super-resolution, high signal-to-noise ratio and compatibility with living tissue. This allows dense deep-learning-based instance segmentation and 3D reconstruction at a synapse level, incorporating molecular, activity and morphodynamic information. LIONESS opens up avenues for studying the dynamic functional (nano-)architecture of living brain tissue. AU - Velicky, Philipp AU - Miguel Villalba, Eder AU - Michalska, Julia M AU - Lyudchik, Julia AU - Wei, Donglai AU - Lin, Zudi AU - Watson, Jake AU - Troidl, Jakob AU - Beyer, Johanna AU - Ben Simon, Yoav AU - Sommer, Christoph M AU - Jahr, Wiebke AU - Cenameri, Alban AU - Broichhagen, Johannes AU - Grant, Seth G.N. AU - Jonas, Peter M AU - Novarino, Gaia AU - Pfister, Hanspeter AU - Bickel, Bernd AU - Danzl, Johann G ID - 13267 JF - Nature Methods SN - 1548-7091 TI - Dense 4D nanoscale reconstruction of living brain tissue VL - 20 ER - TY - JOUR AB - Little is known about the critical metabolic changes that neural cells have to undergo during development and how temporary shifts in this program can influence brain circuitries and behavior. Inspired by the discovery that mutations in SLC7A5, a transporter of metabolically essential large neutral amino acids (LNAAs), lead to autism, we employed metabolomic profiling to study the metabolic states of the cerebral cortex across different developmental stages. We found that the forebrain undergoes significant metabolic remodeling throughout development, with certain groups of metabolites showing stage-specific changes, but what are the consequences of perturbing this metabolic program? By manipulating Slc7a5 expression in neural cells, we found that the metabolism of LNAAs and lipids are interconnected in the cortex. Deletion of Slc7a5 in neurons affects the postnatal metabolic state, leading to a shift in lipid metabolism. Additionally, it causes stage- and cell-type-specific alterations in neuronal activity patterns, resulting in a long-term circuit dysfunction. AU - Knaus, Lisa AU - Basilico, Bernadette AU - Malzl, Daniel AU - Gerykova Bujalkova, Maria AU - Smogavec, Mateja AU - Schwarz, Lena A. AU - Gorkiewicz, Sarah AU - Amberg, Nicole AU - Pauler, Florian AU - Knittl-Frank, Christian AU - Tassinari, Marianna AU - Maulide, Nuno AU - Rülicke, Thomas AU - Menche, Jörg AU - Hippenmeyer, Simon AU - Novarino, Gaia ID - 12802 IS - 9 JF - Cell KW - General Biochemistry KW - Genetics and Molecular Biology SN - 0092-8674 TI - Large neutral amino acid levels tune perinatal neuronal excitability and survival VL - 186 ER - TY - JOUR AB - Mapping the complex and dense arrangement of cells and their connectivity in brain tissue demands nanoscale spatial resolution imaging. Super-resolution optical microscopy excels at visualizing specific molecules and individual cells but fails to provide tissue context. Here we developed Comprehensive Analysis of Tissues across Scales (CATS), a technology to densely map brain tissue architecture from millimeter regional to nanometer synaptic scales in diverse chemically fixed brain preparations, including rodent and human. CATS uses fixation-compatible extracellular labeling and optical imaging, including stimulated emission depletion or expansion microscopy, to comprehensively delineate cellular structures. It enables three-dimensional reconstruction of single synapses and mapping of synaptic connectivity by identification and analysis of putative synaptic cleft regions. Applying CATS to the mouse hippocampal mossy fiber circuitry, we reconstructed and quantified the synaptic input and output structure of identified neurons. We furthermore demonstrate applicability to clinically derived human tissue samples, including formalin-fixed paraffin-embedded routine diagnostic specimens, for visualizing the cellular architecture of brain tissue in health and disease. AU - Michalska, Julia M AU - Lyudchik, Julia AU - Velicky, Philipp AU - Korinkova, Hana AU - Watson, Jake AU - Cenameri, Alban AU - Sommer, Christoph M AU - Amberg, Nicole AU - Venturino, Alessandro AU - Roessler, Karl AU - Czech, Thomas AU - Höftberger, Romana AU - Siegert, Sandra AU - Novarino, Gaia AU - Jonas, Peter M AU - Danzl, Johann G ID - 14257 JF - Nature Biotechnology SN - 1087-0156 TI - Imaging brain tissue architecture across millimeter to nanometer scales ER - TY - JOUR AB - Vacuolar-type H+-ATPase (V-ATPase) is a multimeric complex present in a variety of cellular membranes that acts as an ATP-dependent proton pump and plays a key role in pH homeostasis and intracellular signalling pathways. In humans, 22 autosomal genes encode for a redundant set of subunits allowing the composition of diverse V-ATPase complexes with specific properties and expression. Sixteen subunits have been linked to human disease. Here we describe 26 patients harbouring 20 distinct pathogenic de novo missense ATP6V1A variants, mainly clustering within the ATP synthase α/β family-nucleotide-binding domain. At a mean age of 7 years (extremes: 6 weeks, youngest deceased patient to 22 years, oldest patient) clinical pictures included early lethal encephalopathies with rapidly progressive massive brain atrophy, severe developmental epileptic encephalopathies and static intellectual disability with epilepsy. The first clinical manifestation was early hypotonia, in 70%; 81% developed epilepsy, manifested as developmental epileptic encephalopathies in 58% of the cohort and with infantile spasms in 62%; 63% of developmental epileptic encephalopathies failed to achieve any developmental, communicative or motor skills. Less severe outcomes were observed in 23% of patients who, at a mean age of 10 years and 6 months, exhibited moderate intellectual disability, with independent walking and variable epilepsy. None of the patients developed communicative language. Microcephaly (38%) and amelogenesis imperfecta/enamel dysplasia (42%) were additional clinical features. Brain MRI demonstrated hypomyelination and generalized atrophy in 68%. Atrophy was progressive in all eight individuals undergoing repeated MRIs. Fibroblasts of two patients with developmental epileptic encephalopathies showed decreased LAMP1 expression, Lysotracker staining and increased organelle pH, consistent with lysosomal impairment and loss of V-ATPase function. Fibroblasts of two patients with milder disease, exhibited a different phenotype with increased Lysotracker staining, decreased organelle pH and no significant modification in LAMP1 expression. Quantification of substrates for lysosomal enzymes in cellular extracts from four patients revealed discrete accumulation. Transmission electron microscopy of fibroblasts of four patients with variable severity and of induced pluripotent stem cell-derived neurons from two patients with developmental epileptic encephalopathies showed electron-dense inclusions, lipid droplets, osmiophilic material and lamellated membrane structures resembling phospholipids. Quantitative assessment in induced pluripotent stem cell-derived neurons identified significantly smaller lysosomes. ATP6V1A-related encephalopathy represents a new paradigm among lysosomal disorders. It results from a dysfunctional endo-lysosomal membrane protein causing altered pH homeostasis. Its pathophysiology implies intracellular accumulation of substrates whose composition remains unclear, and a combination of developmental brain abnormalities and neurodegenerative changes established during prenatal and early postanal development, whose severity is variably determined by specific pathogenic variants. AU - Guerrini, Renzo AU - Mei, Davide AU - Szigeti, Margit Katalin AU - Pepe, Sara AU - Koenig, Mary Kay AU - Von Allmen, Gretchen AU - Cho, Megan T AU - McDonald, Kimberly AU - Baker, Janice AU - Bhambhani, Vikas AU - Powis, Zöe AU - Rodan, Lance AU - Nabbout, Rima AU - Barcia, Giulia AU - Rosenfeld, Jill A AU - Bacino, Carlos A AU - Mignot, Cyril AU - Power, Lillian H AU - Harris, Catharine J AU - Marjanovic, Dragan AU - Møller, Rikke S AU - Hammer, Trine B AU - Keski Filppula, Riikka AU - Vieira, Päivi AU - Hildebrandt, Clara AU - Sacharow, Stephanie AU - Maragliano, Luca AU - Benfenati, Fabio AU - Lachlan, Katherine AU - Benneche, Andreas AU - Petit, Florence AU - de Sainte Agathe, Jean Madeleine AU - Hallinan, Barbara AU - Si, Yue AU - Wentzensen, Ingrid M AU - Zou, Fanggeng AU - Narayanan, Vinodh AU - Matsumoto, Naomichi AU - Boncristiano, Alessandra AU - la Marca, Giancarlo AU - Kato, Mitsuhiro AU - Anderson, Kristin AU - Barba, Carmen AU - Sturiale, Luisa AU - Garozzo, Domenico AU - Bei, Roberto AU - Masuelli, Laura AU - Conti, Valerio AU - Novarino, Gaia AU - Fassio, Anna ID - 12174 IS - 8 JF - Brain KW - Neurology (clinical) SN - 0006-8950 TI - Phenotypic and genetic spectrum of ATP6V1A encephalopathy: A disorder of lysosomal homeostasis VL - 145 ER - TY - GEN AB - Complex wiring between neurons underlies the information-processing network enabling all brain functions, including cognition and memory. For understanding how the network is structured, processes information, and changes over time, comprehensive visualization of the architecture of living brain tissue with its cellular and molecular components would open up major opportunities. However, electron microscopy (EM) provides nanometre-scale resolution required for full in-silico reconstruction1–5, yet is limited to fixed specimens and static representations. Light microscopy allows live observation, with super-resolution approaches6–12 facilitating nanoscale visualization, but comprehensive 3D-reconstruction of living brain tissue has been hindered by tissue photo-burden, photobleaching, insufficient 3D-resolution, and inadequate signal-to-noise ratio (SNR). Here we demonstrate saturated reconstruction of living brain tissue. We developed an integrated imaging and analysis technology, adapting stimulated emission depletion (STED) microscopy6,13 in extracellularly labelled tissue14 for high SNR and near-isotropic resolution. Centrally, a two-stage deep-learning approach leveraged previously obtained information on sample structure to drastically reduce photo-burden and enable automated volumetric reconstruction down to single synapse level. Live reconstruction provides unbiased analysis of tissue architecture across time in relation to functional activity and targeted activation, and contextual understanding of molecular labelling. This adoptable technology will facilitate novel insights into the dynamic functional architecture of living brain tissue. AU - Velicky, Philipp AU - Miguel Villalba, Eder AU - Michalska, Julia M AU - Wei, Donglai AU - Lin, Zudi AU - Watson, Jake AU - Troidl, Jakob AU - Beyer, Johanna AU - Ben Simon, Yoav AU - Sommer, Christoph M AU - Jahr, Wiebke AU - Cenameri, Alban AU - Broichhagen, Johannes AU - Grant, Seth G. N. AU - Jonas, Peter M AU - Novarino, Gaia AU - Pfister, Hanspeter AU - Bickel, Bernd AU - Danzl, Johann G ID - 11943 T2 - bioRxiv TI - Saturated reconstruction of living brain tissue ER - TY - GEN AB - Mapping the complex and dense arrangement of cells and their connectivity in brain tissue demands nanoscale spatial resolution imaging. Super-resolution optical microscopy excels at visualizing specific molecules and individual cells but fails to provide tissue context. Here we developed Comprehensive Analysis of Tissues across Scales (CATS), a technology to densely map brain tissue architecture from millimeter regional to nanoscopic synaptic scales in diverse chemically fixed brain preparations, including rodent and human. CATS leverages fixation-compatible extracellular labeling and advanced optical readout, in particular stimulated-emission depletion and expansion microscopy, to comprehensively delineate cellular structures. It enables 3D-reconstructing single synapses and mapping synaptic connectivity by identification and tailored analysis of putative synaptic cleft regions. Applying CATS to the hippocampal mossy fiber circuitry, we demonstrate its power to reveal the system’s molecularly informed ultrastructure across spatial scales and assess local connectivity by reconstructing and quantifying the synaptic input and output structure of identified neurons. AU - Michalska, Julia M AU - Lyudchik, Julia AU - Velicky, Philipp AU - Korinkova, Hana AU - Watson, Jake AU - Cenameri, Alban AU - Sommer, Christoph M AU - Venturino, Alessandro AU - Roessler, Karl AU - Czech, Thomas AU - Siegert, Sandra AU - Novarino, Gaia AU - Jonas, Peter M AU - Danzl, Johann G ID - 11950 T2 - bioRxiv TI - Uncovering brain tissue architecture across scales with super-resolution light microscopy ER - TY - JOUR AB - Mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are a frequent cause of autism spectrum disorder (ASD). While its phenotypic spectrum often encompasses macrocephaly, implicating cortical abnormalities, how CHD8 haploinsufficiency affects neurodevelopmental is unclear. Here, employing human cerebral organoids, we find that CHD8 haploinsufficiency disrupted neurodevelopmental trajectories with an accelerated and delayed generation of, respectively, inhibitory and excitatory neurons that yields, at days 60 and 120, symmetrically opposite expansions in their proportions. This imbalance is consistent with an enlargement of cerebral organoids as an in vitro correlate of patients’ macrocephaly. Through an isogenic design of patient-specific mutations and mosaic organoids, we define genotype-phenotype relationships and uncover their cell-autonomous nature. Our results define cell-type-specific CHD8-dependent molecular defects related to an abnormal program of proliferation and alternative splicing. By identifying cell-type-specific effects of CHD8 mutations, our study uncovers reproducible developmental alterations that may be employed for neurodevelopmental disease modeling. AU - Villa, Carlo Emanuele AU - Cheroni, Cristina AU - Dotter, Christoph AU - López-Tóbon, Alejandro AU - Oliveira, Bárbara AU - Sacco, Roberto AU - Yahya, Aysan Çerağ AU - Morandell, Jasmin AU - Gabriele, Michele AU - Tavakoli, Mojtaba AU - Lyudchik, Julia AU - Sommer, Christoph M AU - Gabitto, Mariano AU - Danzl, Johann G AU - Testa, Giuseppe AU - Novarino, Gaia ID - 11160 IS - 1 JF - Cell Reports KW - General Biochemistry KW - Genetics and Molecular Biology SN - 2211-1247 TI - CHD8 haploinsufficiency links autism to transient alterations in excitatory and inhibitory trajectories VL - 39 ER - TY - JOUR AB - Mutations affecting mTOR or RAS signaling underlie defined syndromes (the so-called mTORopathies and RASopathies) with high risk for Autism Spectrum Disorder (ASD). These syndromes show a broad variety of somatic phenotypes including cancers, skin abnormalities, heart disease and facial dysmorphisms. Less well studied are the neuropsychiatric symptoms such as ASD. Here, we assess the relevance of these signalopathies in ASD reviewing genetic, human cell model, rodent studies and clinical trials. We conclude that signalopathies have an increased liability for ASD and that, in particular, ASD individuals with dysmorphic features and intellectual disability (ID) have a higher chance for disruptive mutations in RAS- and mTOR-related genes. Studies on rodent and human cell models confirm aberrant neuronal development as the underlying pathology. Human studies further suggest that multiple hits are necessary to induce the respective phenotypes. Recent clinical trials do only report improvements for comorbid conditions such as epilepsy or cancer but not for behavioral aspects. Animal models show that treatment during early development can rescue behavioral phenotypes. Taken together, we suggest investigating the differential roles of mTOR and RAS signaling in both human and rodent models, and to test drug treatment both during and after neuronal development in the available model systems AU - Vasic, Verica AU - Jones, Mattson S.O. AU - Haslinger, Denise AU - Knaus, Lisa AU - Schmeisser, Michael J. AU - Novarino, Gaia AU - Chiocchetti, Andreas G. ID - 10281 IS - 11 JF - Genes TI - Translating the role of mtor-and ras-associated signalopathies in autism spectrum disorder: Models, mechanisms and treatment VL - 12 ER - TY - JOUR AB - P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) restrict at the blood–brain barrier (BBB) the brain distribution of the majority of currently known molecularly targeted anticancer drugs. To improve brain delivery of dual ABCB1/ABCG2 substrates, both ABCB1 and ABCG2 need to be inhibited simultaneously at the BBB. We examined the feasibility of simultaneous ABCB1/ABCG2 inhibition with i.v. co-infusion of erlotinib and tariquidar by studying brain distribution of the model ABCB1/ABCG2 substrate [11C]erlotinib in mice and rhesus macaques with PET. Tolerability of the erlotinib/tariquidar combination was assessed in human embryonic stem cell-derived cerebral organoids. In mice and macaques, baseline brain distribution of [11C]erlotinib was low (brain distribution volume, VT,brain < 0.3 mL/cm3). Co-infusion of erlotinib and tariquidar increased VT,brain in mice by 3.0-fold and in macaques by 3.4- to 5.0-fold, while infusion of erlotinib alone or tariquidar alone led to less pronounced VT,brain increases in both species. Treatment of cerebral organoids with erlotinib/tariquidar led to an induction of Caspase-3-dependent apoptosis. Co-infusion of erlotinib/tariquidar may potentially allow for complete ABCB1/ABCG2 inhibition at the BBB, while simultaneously achieving brain-targeted EGFR inhibition. Our protocol may be applicable to enhance brain delivery of molecularly targeted anticancer drugs for a more effective treatment of brain tumors. AU - Tournier, N AU - Goutal, S AU - Mairinger, S AU - Lozano, IH AU - Filip, T AU - Sauberer, M AU - Caillé, F AU - Breuil, L AU - Stanek, J AU - Freeman, AF AU - Novarino, Gaia AU - Truillet, C AU - Wanek, T AU - Langer, O ID - 8730 IS - 7 JF - Journal of Cerebral Blood Flow and Metabolism SN - 0271-678x TI - Complete inhibition of ABCB1 and ABCG2 at the blood-brain barrier by co-infusion of erlotinib and tariquidar to improve brain delivery of the model ABCB1/ABCG2 substrate [11C]erlotinib VL - 41 ER - TY - JOUR AB - De novo loss of function mutations in the ubiquitin ligase-encoding gene Cullin3 lead to autism spectrum disorder (ASD). In mouse, constitutive haploinsufficiency leads to motor coordination deficits as well as ASD-relevant social and cognitive impairments. However, induction of Cul3 haploinsufficiency later in life does not lead to ASD-relevant behaviors, pointing to an important role of Cul3 during a critical developmental window. Here we show that Cul3 is essential to regulate neuronal migration and, therefore, constitutive Cul3 heterozygous mutant mice display cortical lamination abnormalities. At the molecular level, we found that Cul3 controls neuronal migration by tightly regulating the amount of Plastin3 (Pls3), a previously unrecognized player of neural migration. Furthermore, we found that Pls3 cell-autonomously regulates cell migration by regulating actin cytoskeleton organization, and its levels are inversely proportional to neural migration speed. Finally, we provide evidence that cellular phenotypes associated with autism-linked gene haploinsufficiency can be rescued by transcriptional activation of the intact allele in vitro, offering a proof of concept for a potential therapeutic approach for ASDs. AU - Morandell, Jasmin AU - Schwarz, Lena A AU - Basilico, Bernadette AU - Tasciyan, Saren AU - Dimchev, Georgi A AU - Nicolas, Armel AU - Sommer, Christoph M AU - Kreuzinger, Caroline AU - Dotter, Christoph AU - Knaus, Lisa AU - Dobler, Zoe AU - Cacci, Emanuele AU - Schur, Florian KM AU - Danzl, Johann G AU - Novarino, Gaia ID - 9429 IS - 1 JF - Nature Communications KW - General Biochemistry KW - Genetics and Molecular Biology TI - Cul3 regulates cytoskeleton protein homeostasis and cell migration during a critical window of brain development VL - 12 ER - TY - JOUR AB - CLC chloride/proton exchangers may support acidification of endolysosomes and raise their luminal Cl− concentration. Disruption of endosomal ClC‐3 causes severe neurodegeneration. To assess the importance of ClC‐3 Cl−/H+ exchange, we now generate Clcn3unc/unc mice in which ClC‐3 is converted into a Cl− channel. Unlike Clcn3−/− mice, Clcn3unc/unc mice appear normal owing to compensation by ClC‐4 with which ClC‐3 forms heteromers. ClC‐4 protein levels are strongly reduced in Clcn3−/−, but not in Clcn3unc/unc mice because ClC‐3unc binds and stabilizes ClC‐4 like wild‐type ClC‐3. Although mice lacking ClC‐4 appear healthy, its absence in Clcn3unc/unc/Clcn4−/− mice entails even stronger neurodegeneration than observed in Clcn3−/− mice. A fraction of ClC‐3 is found on synaptic vesicles, but miniature postsynaptic currents and synaptic vesicle acidification are not affected in Clcn3unc/unc or Clcn3−/− mice before neurodegeneration sets in. Both, Cl−/H+‐exchange activity and the stabilizing effect on ClC‐4, are central to the biological function of ClC‐3. AU - Weinert, Stefanie AU - Gimber, Niclas AU - Deuschel, Dorothea AU - Stuhlmann, Till AU - Puchkov, Dmytro AU - Farsi, Zohreh AU - Ludwig, Carmen F. AU - Novarino, Gaia AU - López-Cayuqueo, Karen I. AU - Planells-Cases, Rosa AU - Jentsch, Thomas J. ID - 7586 JF - EMBO Journal SN - 02614189 TI - Uncoupling endosomal CLC chloride/proton exchange causes severe neurodegeneration VL - 39 ER - TY - JOUR AB - Neurodevelopmental disorders (NDDs) are a class of disorders affecting brain development and function and are characterized by wide genetic and clinical variability. In this review, we discuss the multiple factors that influence the clinical presentation of NDDs, with particular attention to gene vulnerability, mutational load, and the two-hit model. Despite the complex architecture of mutational events associated with NDDs, the various proteins involved appear to converge on common pathways, such as synaptic plasticity/function, chromatin remodelers and the mammalian target of rapamycin (mTOR) pathway. A thorough understanding of the mechanisms behind these pathways will hopefully lead to the identification of candidates that could be targeted for treatment approaches. AU - Parenti, Ilaria AU - Garcia Rabaneda, Luis E AU - Schön, Hanna AU - Novarino, Gaia ID - 7957 IS - 8 JF - Trends in Neurosciences SN - 01662236 TI - Neurodevelopmental disorders: From genetics to functional pathways VL - 43 ER - TY - GEN AB - De novo loss of function mutations in the ubiquitin ligase-encoding gene Cullin3 (CUL3) lead to autism spectrum disorder (ASD). Here, we used Cul3 mouse models to evaluate the consequences of Cul3 mutations in vivo. Our results show that Cul3 haploinsufficient mice exhibit deficits in motor coordination as well as ASD-relevant social and cognitive impairments. Cul3 mutant brain displays cortical lamination abnormalities due to defective neuronal migration and reduced numbers of excitatory and inhibitory neurons. In line with the observed abnormal columnar organization, Cul3 haploinsufficiency is associated with decreased spontaneous excitatory and inhibitory activity in the cortex. At the molecular level, employing a quantitative proteomic approach, we show that Cul3 regulates cytoskeletal and adhesion protein abundance in mouse embryos. Abnormal regulation of cytoskeletal proteins in Cul3 mutant neuronal cells results in atypical organization of the actin mesh at the cell leading edge, likely causing the observed migration deficits. In contrast to these important functions early in development, Cul3 deficiency appears less relevant at adult stages. In fact, induction of Cul3 haploinsufficiency in adult mice does not result in the behavioral defects observed in constitutive Cul3 haploinsufficient animals. Taken together, our data indicate that Cul3 has a critical role in the regulation of cytoskeletal proteins and neuronal migration and that ASD-associated defects and behavioral abnormalities are primarily due to Cul3 functions at early developmental stages. AU - Morandell, Jasmin AU - Schwarz, Lena A AU - Basilico, Bernadette AU - Tasciyan, Saren AU - Nicolas, Armel AU - Sommer, Christoph M AU - Kreuzinger, Caroline AU - Knaus, Lisa AU - Dobler, Zoe AU - Cacci, Emanuele AU - Danzl, Johann G AU - Novarino, Gaia ID - 7800 T2 - bioRxiv TI - Cul3 regulates cytoskeleton protein homeostasis and cell migration during a critical window of brain development ER - TY - JOUR AB - The possibility to generate construct valid animal models enabled the development and testing of therapeutic strategies targeting the core features of autism spectrum disorders (ASDs). At the same time, these studies highlighted the necessity of identifying sensitive developmental time windows for successful therapeutic interventions. Animal and human studies also uncovered the possibility to stratify the variety of ASDs in molecularly distinct subgroups, potentially facilitating effective treatment design. Here, we focus on the molecular pathways emerging as commonly affected by mutations in diverse ASD-risk genes, on their role during critical windows of brain development and the potential treatments targeting these biological processes. AU - Basilico, Bernadette AU - Morandell, Jasmin AU - Novarino, Gaia ID - 8131 IS - 12 JF - Current Opinion in Genetics and Development SN - 0959437X TI - Molecular mechanisms for targeted ASD treatments VL - 65 ER - TY - JOUR AB - Clinical Utility Gene Card. 1. Name of Disease (Synonyms): Pontocerebellar hypoplasia type 9 (PCH9) and spastic paraplegia-63 (SPG63). 2. OMIM# of the Disease: 615809 and 615686. 3. Name of the Analysed Genes or DNA/Chromosome Segments: AMPD2 at 1p13.3. 4. OMIM# of the Gene(s): 102771. AU - Marsh, Ashley AU - Novarino, Gaia AU - Lockhart, Paul AU - Leventer, Richard ID - 105 JF - European Journal of Human Genetics TI - CUGC for pontocerebellar hypoplasia type 9 and spastic paraplegia-63 VL - 27 ER - TY - JOUR AB - P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters at the blood–brain barrier (BBB), which effectively restrict brain distribution of diverse drugs, such as tyrosine kinase inhibitors. There is a crucial need for pharmacological ABCB1 and ABCG2 inhibition protocols for a more effective treatment of brain diseases. In the present study, seven marketed drugs (osimertinib, erlotinib, nilotinib, imatinib, lapatinib, pazopanib, and cyclosporine A) and one nonmarketed drug (tariquidar), with known in vitro ABCB1/ABCG2 inhibitory properties, were screened for their inhibitory potency at the BBB in vivo. Positron emission tomography (PET) using the model ABCB1/ABCG2 substrate [11C]erlotinib was performed in mice. Tested inhibitors were administered as i.v. bolus injections at 30 min before the start of the PET scan, followed by a continuous i.v. infusion for the duration of the PET scan. Five of the tested drugs increased total distribution volume of [11C]erlotinib in the brain (VT,brain) compared to vehicle-treated animals (tariquidar, + 69%; erlotinib, + 19% and +23% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 22%; lapatinib, + 25%; and cyclosporine A, + 49%). For all drugs, increases in [11C]erlotinib brain distribution were lower than in Abcb1a/b(−/−)Abcg2(−/−) mice (+149%), which suggested that only partial ABCB1/ABCG2 inhibition was reached at the mouse BBB. The plasma concentrations of the tested drugs at the time of the PET scan were higher than clinically achievable plasma concentrations. Some of the tested drugs led to significant increases in blood radioactivity concentrations measured at the end of the PET scan (erlotinib, + 103% and +113% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 125%; and cyclosporine A, + 101%), which was most likely caused by decreased hepatobiliary excretion of radioactivity. Taken together, our data suggest that some marketed tyrosine kinase inhibitors may be repurposed to inhibit ABCB1 and ABCG2 at the BBB. From a clinical perspective, moderate increases in brain delivery despite the administration of high i.v. doses as well as peripheral drug–drug interactions due to transporter inhibition in clearance organs question the translatability of this concept. AU - Traxl, Alexander AU - Mairinger, Severin AU - Filip, Thomas AU - Sauberer, Michael AU - Stanek, Johann AU - Poschner, Stefan AU - Jäger, Walter AU - Zoufal, Viktoria AU - Novarino, Gaia AU - Tournier, Nicolas AU - Bauer, Martin AU - Wanek, Thomas AU - Langer, Oliver ID - 6088 IS - 3 JF - Molecular Pharmaceutics TI - Inhibition of ABCB1 and ABCG2 at the mouse blood-brain barrier with marketed drugs to improve brain delivery of the model ABCB1/ABCG2 substrate [11C]erlotinib VL - 16 ER - TY - JOUR AB - Until recently, a great amount of brain studies have been conducted in human post mortem tissues, cell lines and model organisms. These researches provided useful insights regarding cell-cell interactions occurring in the brain. However, such approaches suffer from technical limitations and inaccurate modeling of the tissue 3D cytoarchitecture. Importantly, they might lack a human genetic background essential for disease modeling. With the development of protocols to generate human cerebral organoids, we are now closer to reproducing the early stages of human brain development in vitro. As a result, more relevant cell-cell interaction studies can be conducted. In this review, we discuss the advantages of 3D cultures over 2D in modulating brain cell-cell interactions during physiological and pathological development, as well as the progress made in developing organoids in which neurons, macroglia, microglia and vascularization are present. Finally, we debate the limitations of those models and possible future directions. AU - Oliveira, Bárbara AU - Yahya, Aysan Çerağ AU - Novarino, Gaia ID - 6896 JF - Brain Research SN - 00068993 TI - Modeling cell-cell interactions in the brain using cerebral organoids VL - 1724 ER -