18 research outputs found

    Refining iPSC-based 3D neural cell models and characterization tools to address brain microenvironment-related diseases

    Get PDF
    Brain microenvironment plays important roles in neurodevelopment and pathology and can affect therapy efficacy. Neural cell culture typically relies on the use of heterologous matrices that poorly resemble brain extracellular matrix (ECM) or reflect its pathological features. We have shown that perfusion bioreactor-based 3D differentiation of iPSC-derived human neural stem cells (hiPSC-NSC) sustains the concomitant differentiation of the three neural lineages (neurospheroids). If this neurospheroid culture strategy also allows deposition of native neural ECM it would be possible to (i) mimic cellular and microenvironment remodeling during neural differentiation, without the confounding effects of exogenous matrices and (ii) recapitulate pathological phenotypic features of diseases in which homotypic/ heterotypic cell-cell interactions and ECM are relevant. To characterize the neural extracellular space we employed quantitative transcriptomic (NGS) and proteome (SWATH-MS) analysis. Neurogenic developmental pathways were recapitulated in neurospheroids, with significant changes in cell membrane and ECM composition along differentiation; a significant enrichment in structural proteoglycans, typical of brain ECM, a downregulation of basement membrane proteins constituents and a higher expression of synaptic and ion transport machinery were observed. Neurospheroids were generated using hiPSC-NSC derived from Mucopolysaccharidosis type VII (MPS VII) patients. MPS VII is a rare neuronopathic lysosomal storage disease caused by deficient β-glucuronidase (β-gluc) activity, leading to glycosaminoglycan (GAGs) accumulation in the brain. The main MPS VII molecular hallmarks were recapitulated, e.g. accumulation og GAGs. By combining the neurospheroid culture with a 3D neuronal connectivity assay based on calcium imaging analysis we refined a new analytical strategy to characterize neuronal connectivity defects in a more predictive setting. We showed that MPS VII neurospheroids presented reduced neuronal activity and disturbances in network functionality, with alterations in connectivity and synchronization. These data provide insights into the interplay between reduced β-gluc activity, GAGs accumulation, alterations in neuronal network and its impact on MPS VII-associated cognitive defects Applying the characterization tools refined in this work to cope with 3D neurospheroid cultures, namely the neuronal connectivity assay, we provide a new platform to unveil the cellular processes responsible for brain dysfunction in neurological disorders and to test and optimize new therapies. Acknowledgments: iNOVA4Health – UID/Multi/04462/2013, a program financially supported by Fundação para a Ciência e Tecnologia/Ministério da Educação e Ciência, through national funds and co-funded by FEDER under the PT2020 Partnership Agreement, is acknowledged. PD/BD/52473/2014, PD/BD/52481/2014, and PD/BD/128371/2017 PhD fellowships funded by FCT, Portugal. N.R. is supported by the European Research Council Starting Grant 337327. MS data were obtained by UniMS – Mass Spectrometry Unit, ITQB and iBET, Oeiras, Portugal

    iPSC-derived neurospheroids recapitulate development and pathological signatures of human brain microenvironment

    Get PDF
    Brain microenvironment plays an important role in neurodevelopment and pathology, where extracellular matrix (ECM) and soluble factors modulate multiple cellular processes. Neural cell culture typically relies on the use of heterologous matrices that poorly resemble the brain ECM or reflect its pathological features. We have previously demonstrated that perfusion stirred-tank bioreactor-based 3D differentiation of human neural stem cells (NSC) - pSTR-neurospheroids, sustains the concomitant differentiation of the three neural cell lineages (neurons, astrocytes and oligodendrocytes) and the establishment of physiologically relevant cell-cell interactions. Here, we hypothesized that if the pSTR-neurospheroid strategy would also allow the deposition of native neural ECM components and diffusion of secreted factors, it would be possible to: (i) mimic the cellular and microenvironment remodeling occurring during neural differentiation without the confounding effects of exogenous matrices; (ii) recapitulate the pathological phenotypes of diseases in which alteration of homotypic and heterotypic cell-cell interactions and ECM components are relevant. To demonstrate the first point, we analyzed pSTR-neurospheroid differentiation by quantitative transcriptome (NGS) and proteome (SWATH-MS). Data showed that neurogenic developmental pathways were recapitulated, with significant changes at cell membrane and ECM composition, diverging from the 2D differentiation profile. A significant enrichment in structural proteoglycans typical of brain ECM, along with downregulation of basement membrane constituents was observed. Moreover, higher expression of synaptic and ion transport machinery in pSTR-neurospheroids suggest higher neuronal maturation than in 2D. Having shown recapitulation of neural microenvironmental dynamics in pSTR-neurospheroids, we used Mucopolysaccharidosis VII (MPSVII) as a disease case study. MPS VII is a lysosomal storage disease caused by deficient β-glucuronidase (β-gluc) activity, which leads to accumulation of glycosaminoglycans (GAGs) in many tissues, including the brain. In pSTR-neurospheroids generated from hiPSC of a MPS VII patient, the main molecular disease hallmarks were recapitulated, namely accumulation of GAGs. Notably, MPS VII neurospheroids showed reduced neuronal activity and a disturbance in network functionality, with alterations both in connectivity and synchronization, not observed in 2D cultures. These data provide insight into the interplay between reduced β-gluc activity, GAG accumulation, alterations in the neural network, and its impact on MPS VII-associated cognitive defects. Overall we demonstrate that neural cellular and extracellular developmental and pathological features are recapitulated in healthy and diseased pSTR-neurospheroids, respectively. These can be valuable in vitro models to address molecular defects associated with neurological disorders that affect neural microenvironment homeostasis. Moreover, the 3D neuronal connectivity assay developed is a new tool with potential to assess other lysosomal storage diseases and neurodegenerative diseases that have variable phenotypes. Acknowledgements: SFRH/BD/78308/2011, SFRH/BD/52202/2013 and SFRH/BD/52473/2014 PhD fellowships from FCT, Portugal and iNOVA4Health-UID/Multi/04462/2013, supported by FCT/ MEC, through national funds and co-funded by FEDER under the PT2020 Partnership Agreement

    Lysosomal and network alterations in human mucopolysaccharidosis type VII iPSC-derived neural cells

    Get PDF
    Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by deficient β-glucuronidase (β-gluc) activity. Significantly reduced β-gluc activity leads to accumulation of glycosaminoglycans (GAGs) in many tissues, including the brain. Numerous combinations of mutations in GUSB (the gene that codes for β-gluc) cause a range of neurological features that make disease prognosis and treatment challenging. Currently, there is little understanding of the molecular basis for MPS VII brain anomalies. To identify a neuronal phenotype that could be used to complement genetic analyses, we generated two iPSC clones derived from skin fibroblasts of an MPS VII patient. We found that MPS VII neurons exhibited reduced β-gluc activity and showed previously established disease-associated phenotypes, including GAGs accumulation, expanded endocytic compartments, accumulation of lipofuscin granules, more autophagosomes, and altered lysosome function. Addition of recombinant β-gluc to MPS VII neurons, which mimics enzyme replacement therapy, restored disease-associated phenotypes to levels similar to the healthy control. MPS VII neural cells cultured as 3D neurospheroids showed upregulated GFAP gene expression, which was associated with astrocyte reactivity, and downregulation of GABAergic neuron markers. Spontaneous calcium imaging analysis of MPS VII neurospheroids showed reduced neuronal activity and altered network connectivity in patient-derived neurospheroids compared to a healthy control. These results demonstrate the interplay between reduced β-gluc activity, GAG accumulation and alterations in neuronal activity, and provide a human experimental model for elucidating the bases of MPS VII-associated cognitive defects

    Early-Stage Breast Cancer Detection in Breast Milk

    Get PDF
    Breast cancer; Breast milkCáncer de mama; Leche maternaCàncer de mama; Llet maternaBreast cancer occurring during pregnancy (PrBC) and postpartum (PPBC) is usually diagnosed at more advanced stages compared with other breast cancer, worsening its prognosis. PPBC is particularly aggressive, with increased metastatic risk and mortality. Thus, effective screening methods to detect early PrBC and PPBC are needed. We report for the first time that cell-free tumor DNA (ctDNA) is present in breast milk (BM) collected from patients with breast cancer. Analysis of ctDNA from BM detects tumor variants in 87% of the cases by droplet digital PCR, while variants remain undetected in 92% of matched plasma samples. Retrospective next-generation sequencing analysis in BM ctDNA recapitulates tumor variants, with an overall clinical sensitivity of 71.4% and specificity of 100%. In two cases, ctDNA was detectable in BM collected 18 and 6 months prior to standard diagnosis. Our results open up the potential use of BM as a new source for liquid biopsy for PPBC detection. Significance: For the first time, we show that BM obtained from patients with breast cancer carries ctDNA, surpassing plasma-based liquid biopsy for detection and molecular profiling of early-stage breast cancer, even prior to diagnosis by image.We thank the patients who participated in the study and donated samples for analysis for their generous contribution, with particular thanks to the first patient, Maite, and her daughter Àneu, who inspired us to initiate this study (oral consent to name the patient and her daughter was provided by the patient, and her legal partner provided written consent after patient's exitus). We are grateful to Javier Carmona for his valuable contributions and support in the manuscript's conceptualization, preparation, and revision. VHIO would like to acknowledge the Cellex Foundation for providing research facilities and equipment and the CERCA Programme from the Generalitat de Catalunya for their support of this research. The authors from VHIO acknowledge the State Agency for Research (Agencia Estatal de Investigación) for the financial support as a Center of Excellence Severo Ochoa (CEX2020-001024-S/AEI/10.13039/501100011033). This research is financially supported by the “El paseíco de la mama” Foundation. C. Saura was the recipient of a II FERO-GHD grant from the FERO Foundation (FERO/5086), a Junior Clinical award from the Spanish Association Against Cancer Foundation (FAECC; CLJUN212026ORTI), and a SEOM-Daiichi Sankyo grant for its support on the Breast Cancer Research Projects 2021 (SEOM/FECMA2022) and received funding from the Department of Health (Generalitat de Catalunya SLT008/18/00198) and from the Instituto de Salud Carlos III (ISCIII) and Fondo Europeo de Desarrollo Regional (FEDER), cofunded by the European Union (PI21/01020). C. Ortiz was the recipient of a Junior Clinician award from the FAECC (CLJUN212026ORTI) and a SEOM-Daiichi Sankyo grant for its support on the Breast Cancer Research Projects 2021 (SEOM/FECMA2022), and received funding from the Department of Health (Generalitat de Catalunya SLT008/18/00198). N. Bayó-Puxan received funding from the Department of Health (Generalitat de Catalunya SLT008/18/00205), MCIN/AEI/10.13039/501100011033 (GPE2022-001029) and MCIN/AEI/10.130.39/501100011033, and the European Union “Next GenerationEU/PRTR” (ECT2020-000827). J.M. Miquel received funding from the Department of Health (Generalitat de Catalunya SLT008/18/00205), MCIN/AEI/10.130.39/501100011033, and the European Union “Next GenerationEU/PRTR” (ECT2020-000827). J. Arribas is funded by the Breast Cancer Research Foundation (BCRF-23-008), Instituto de Salud Carlos III (project reference numbers AC15/00062, CB16/12/00449, and PI22/00001), and the European Commission under the framework of the ERA-NET TRANSCAN-2 initiative cofinanced by FEDER and Asociación Española Contra el Cáncer. A. Vivancos was the recipient of a project award from the FAECC (AVP/18/AECC/3219) and received funding from the Advanced Molecular Diagnostic (DIAMAV) program from the FERO Foundation (8361) and from ISDIN for supporting the development of liquid biopsy applications at the Cancer Genomics Lab (1848). M. Sansó was the recipient of a II FERO-GHD grant from the FERO Foundation (FERO/5086) and an investigator award from the FAECC (INVES19056SANS), and received funding from the Health Research Institute of the Balearic Islands (IdISBa), the RADIX-Janssen program (RADIX/JANSSEN21/01), and the Miguel Servet Program funded by the ISCIII (CP22/00131)

    CAV2 vector gene transfer into central nervious System

    No full text
    Estudi realitzat a partir d’una estada al Institut de Génétique Moléculaire de Montpellier, França, entre 2010 i 2012. En aquest projecte s’ha avaluat les avantatges dels vectors adenovirals canins tipus 2 (CAV2) com a vectors de transferència gènica al sistema nerviós central (SNC) en un model primat no-humà i en un model caní del síndrome de Sly (mucopolisacaridosis tipus 7, MPS VII), malaltia monogènica que cursa amb neurodegeneració. En una primera part del projecte s’ha avaluat la biodistribució, l’eficàcia i la durada de l’expressió del transgen en un model primat no humà, (Microcebus murinus). Com ha vector s’ha utilitzat un CAV2 de primera generació que expressa la proteïna verda fluorescent (CAVGFP). Els resultats aportats en aquesta memòria demostren que en primats no humans, com en d’altres espècies testades anteriorment per l’equip de l’EJ Kremer, la injecció intracerebral de CAV2 resulta en una extensa transducció del SNC, siguent les neurones i els precursors neuronals les cèl•lules preferencialment transduïdes. Els vectors canins, servint-se de vesícules intracel•lulars són transportats, majoritàriament, des de les sinapsis cap al soma neuronal, aquest transport intracel•lular permet una extensa transducció del SNC a partir d’una única injecció intracerebral dels vectors virals. En una segona part d’aquest projecte s’ha avaluat l’ús terapèutic dels CAV2. S’ha injectat un vector helper-dependent que expressa el gen la b-glucuronidasa i el gen de la proteïna verda fluorescent (HD-RIGIE), en el SNC del model caní del síndrome de Sly (MPS VII). La biodistribució i la eficàcia terapèutica han estat avaluades. Els nivells d’activitat enzimàtica en animals malalts injectats amb el vector terapèutic va arribar a valors similars als dels animals no afectes. A més a més s’ha observat una reducció en la quantitat dels GAGs acumulats en les cèl•lules dels animals malalts tractats amb el vector terapèutic, demostrant la potencialitat terapèutica dels CAV2 per a malalties que afecten al SNC. Els resultats aportats en aquest treball ens permeten dir que els CAV2 són unes bones eines terapèutiques per al tractament de malalties que afecten al SNC.Report for the scientific sojourn carried out at the al Institut de Génétique Moléculaire de Montpellier, France, from 2010 to 2012. Advantages of canine adenoviral vector type 2 (CAV2) as a CNS gene transfer vector have been assessed in a non-human primate model and in a canine model for Sly Syndrome; monogenic neurodegenerative disease. Biodistribution, efficacy and transgene expression has been assessed in a non-human primate model. CAV2 first generation vector expressing GFP has been used. Results show a widespread CNS transduction of CAV2 in non-human primates, as in other many species previously tested. As in other species, CAV2 preferentially transduces neurons and neural precursor cells. CAV2 are naturally retrogradelly transported allowing widespread distribution along the brain. Therapeutical use of CAV2 vectors has been also assessed. A helper-dependent vector expressing b-glucuronidase and GFP (HD-RIGIE) has been intracerebraly injected into Sly dogs. Biodistribution and therapeutical benefits have been analyzed. Enzymatic activity of b-glucuronidase has been restored on Sly-dogs treated with HD-RIGIE reaching to normal enzymatic levels. Moreover a reduction on GAG accumulation has also been observed on HD-RIGIE-treated dogs. CAV2-based vectors are promising therapeutical tools to treat diseases affecting CNS

    Lysosomal and network alterations in human mucopolysaccharidosis type VII iPSC-derived neurons

    Get PDF
    Abstract Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by deficient β-glucuronidase (β-gluc) activity. Significantly reduced β-gluc activity leads to accumulation of glycosaminoglycans (GAGs) in many tissues, including the brain. Numerous combinations of mutations in GUSB (the gene that codes for β-gluc) cause a range of neurological features that make disease prognosis and treatment challenging. Currently, there is little understanding of the molecular basis for MPS VII brain anomalies. To identify a neuronal phenotype that could be used to complement genetic analyses, we generated two iPSC clones derived from skin fibroblasts of an MPS VII patient. We found that MPS VII neurons exhibited reduced β-gluc activity and showed previously established disease-associated phenotypes, including GAGs accumulation, expanded endocytic compartments, accumulation of lipofuscin granules, more autophagosomes, and altered lysosome function. Addition of recombinant β-gluc to MPS VII neurons, which mimics enzyme replacement therapy, restored disease-associated phenotypes to levels similar to the healthy control. MPS VII neural cells cultured as 3D neurospheroids showed upregulated GFAP gene expression, which was associated with astrocyte reactivity, and downregulation of GABAergic neuron markers. Spontaneous calcium imaging analysis of MPS VII neurospheroids showed reduced neuronal activity and altered network connectivity in patient-derived neurospheroids compared to a healthy control. These results demonstrate the interplay between reduced β-gluc activity, GAG accumulation and alterations in neuronal activity, and provide a human experimental model for elucidating the bases of MPS VII-associated cognitive defects
    corecore