Product Citations: 9

Granulin Loss of Function in Human Mature Brain Organoids Implicates Astrocytes in TDP-43 Pathology

Preprint on BioRxiv : the Preprint Server for Biology on 25 October 2022 by de Majo, M., Koontz, M., et al.

Summary Loss of function (LoF) of Tar-binding protein 43 (TDP-43) and mislocalization, together with TDP-43-positive and hyperphosphorylated inclusions, are found in postmortem tissue of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients, including those carrying LoF variants in the progranulin gene ( GRN ). Modelling TDP-43 pathology has been challenging in vivo and in vitro . We present a 3D-induced pluripotent stem cell (iPSC)-derived paradigm - mature brain organoids (mbOrg) - composed of cortical-like-astrocytes (iA) and neurons (iN). When devoid of GRN , mbOrgs spontaneously recapitulate TDP-43 mislocalization, hyperphosphorylation and LoF phenotypes. Mixing-and-matching genotypes in mbOrgs showed that GRN −/− iA are drivers for TDP-43 pathology. Finally, we rescued TDP-43 LoF by adding exogenous progranulin, demonstrating a link between TDP-43 LoF and progranulin expression. In conclusion, we present an iPSC-derived platform that shows striking features of human TDP-43 proteinopathy and provides a tool for mechanistic modelling of TDP-43 pathology and patient-tailored therapeutic screening for FTD and ALS. Highlights GRN −/− iPSC-derived 3D paradigm (mbOrg) composed of mature cortical-like astrocytes and neurons recapitulates features of TDP-43 proteinopathy GRN −/− cortical-like astrocytes are necessary and sufficient for the development of the TDP-43 loss of function phenotype in mbOrg. A TDP-43 phenotype can be rescued in neurons by treating neuron and astrocyte co-cultures with progranulin full length protein. eTOC blurb In this article, de Majo and colleagues present a novel 3D iPSC-derived model to study neurodegenerative disorders such as ALS and FTD. When devoid of GRN expression, these cultures present features of ALS and FTD associated pathology hardly ever observed in vitro . These phenotypes are shown to be primarily driven by diseased astrocytes and can be rescued by progranulin supplementation.

  • Homo sapiens (Human)
  • Neuroscience
  • Pathology

CD36 Identifies a Subpopulation of Adipose-Derived Mesenchymal Stromal Cells with Enhanced Osteogenic Ability.

Preprint on Research Square on 25 October 2021 by Camilleri, E. T., Riester, S. M., et al.

h4>Background: /h4> Adipose-derived mesenchymal stromal cells (AMSCs) represent attractive cellular therapeutics for treatment of various diseases including osteoarthritis and bone degeneration. Preparation and characterization of AMSCs show donor-to-donor variability that indicating that AMSCs are heterogeneous populations of cells. Cell surface markers to identify MSCs, such as CD44, CD90, and CD105, are relatively invariable among AMSCs and do not reflect the heterogeneity of AMSC populations. CD36, a multifunctional surface receptor associated with transport of oxidized low-density lipoproteins and fatty acids, is also expressed by AMSCs but this marker is considerable more variable. Therefore, we assessed whether CD36 can be used to identify subpopulations in AMSCs. Methods Clinical-grade AMSCs were isolated from patient fat biopsies, cultured under xenobiotic-free conditions using human platelet lysate and sorted by flow cytometry or magnetic cell sorting to characterize CD36 expression. CD36 enriched, CD36 depleted, and unsorted AMSCs were characterized using cellular and molecular techniques including proliferation, multilineage potential, RNA-sequencing and bioinformatics, metabolomics, and sensitivity to the CD36 inhibitor sulfosuccinimidyl oleate (SSO). Results CD36 exhibited biphasic expression on AMSCs grown in human platelet lysate. Enrichment of CD36 + AMSCs showed that CD36 + expression identifies a stable subpopulation. Transcriptomic analysis revealed enhancement of calcium transporter genes. Osteogenic differentiation of CD36 + enriched, CD36 + depleted, and unsorted AMSCs showed strikingly enhanced osteogenic calcium deposition and enhanced expression of alkaline phosphatase (ALPL) and osteoprotegrin (TNFRSF11B) in the CD36 + population. While lipid droplets were not altered, adipogenic differentiation showed upregulated gene expression of key adipogenic markers, including fatty acid-binding protein 4 (FABP4) and Adiponectin (ADIPOQ), with CD36 + enrichment. Treatment of CD36 + enriched AMSCs with the SSO showed reduced calcium deposition whereas CD36 + depleted AMSCs were unaffected. Conclusions CD36 exhibits variable expression amongst AMSCs. CD36 + enriched AMSCs are a stable subpopulation with enhanced osteogenic potential that linked to CD36 receptor function. These results may further refine production and clinical application of AMSC cellular therapeutics. Furthermore, the enhanced osteogenic potential of CD36 + AMSCs may be considered for therapeutic regeneration of bone.

  • Homo sapiens (Human)

Fibroblastic reticular cells (FRCs) are stromal cells in secondary lymphoid organs, the major sites for HIV-1 infection of CD4+ T cells. Although FRCs regulate T cell survival, proliferation, and migration, whether they play any role in HIV-1 spread has not been studied. Here, we show that FRCs enhance HIV-1 spread via trans-infection in which FRCs capture HIV-1 and facilitate infection of T cells that come into contact with FRCs. FRCs mediate trans-infection in both two- and three-dimensional culture systems and in a manner dependent on the virus producer cells. This producer cell dependence, which was also observed for virus spread in secondary lymphoid tissues ex vivo, is accounted for by CD44 incorporated into virus particles and hyaluronan bound to such CD44 molecules. This virus-associated hyaluronan interacts with CD44 expressed on FRCs, thereby promoting virus capture by FRCs. Overall, our results reveal a novel role for FRCs in promoting HIV-1 spread.

  • FC/FACS
  • Homo sapiens (Human)
  • Immunology and Microbiology

Dysregulation of astrocyte extracellular signaling in Costello syndrome.

In Science Translational Medicine on 6 May 2015 by Krencik, R., Hokanson, K. C., et al.

Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity.
Copyright © 2015, American Association for the Advancement of Science.

  • Neuroscience

Aldosterone-producing adenoma formation in the adrenal cortex involves expression of stem/progenitor cell markers.

In Endocrinology on 1 December 2011 by Boulkroun, S., Samson-Couterie, B., et al.

Aldosterone producing adenoma (APA) is the most common form of surgically curable hypertension. To further understand mechanisms involved in APA formation, we investigated the expression of molecules linked to adrenal stem/precursor cells [β-catenin, Sonic hedgehog (Shh), CD56], and nuclear receptors that play key roles in adrenocortical development and function steroidogenic factor 1, dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1) in six control adrenal glands and 14 adrenals with APA and compared their expression with that of specific markers of zona glomerulosa (ZG) [CYP11B2, Disabled 2 (Dab2)]. Both Dab2 and CD56 were expressed in ZG. Although Dab2 associates uniquely with differentiated ZG cells and its expression is lost when cells transdifferentiate to zona fasciculata (ZF) cells, CD56 was also expressed in ZF and in aldosterone-producing cell clusters, confirming that these structures possess an intermediate phenotype between ZG and ZF cells. Shh was barely detectable in cells located to the outer part of the ZG in the control adrenal; in contrast, its expression was detected in the entire APA and was dramatically increased in the hyperplastic peritumoral ZG. Transcriptome profiling revealed differential expression of components of Shh signaling pathway in a subgroup of APA. Similarly, Wnt/β-catenin signaling was activated in the majority of APA as well as in the entire peritumoral adrenal cortex; however, no mutation was identified in the CTNNB1 gene that could account for β-catenin activation. Our data suggest that both APA and adjacent ZG present characteristics of stem/precursor cells; the reexpression of genes involved in fetal adrenal development could underlie excessive ZG cell proliferation and APA formation.

  • Endocrinology and Physiology
View this product on CiteAb