← Back

Microenvironment

Topic spotlight
TopicWorld Wide

microenvironment

Discover seminars, jobs, and research tagged with microenvironment across World Wide.
10 curated items8 Seminars2 ePosters
Updated almost 2 years ago
10 items · microenvironment
10 results
SeminarNeuroscience

Astrocyte reprogramming / activation and brain homeostasis

Thomaidou Dimitra
Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
Dec 12, 2023

Astrocytes are multifunctional glial cells, implicated in neurogenesis and synaptogenesis, supporting and fine-tuning neuronal activity and maintaining brain homeostasis by controlling blood-brain barrier permeability. During the last years a number of studies have shown that astrocytes can also be converted into neurons if they force-express neurogenic transcription factors or miRNAs. Direct astrocytic reprogramming to induced-neurons (iNs) is a powerful approach for manipulating cell fate, as it takes advantage of the intrinsic neural stem cell (NSC) potential of brain resident reactive astrocytes. To this end, astrocytic cell fate conversion to iNs has been well-established in vitro and in vivo using combinations of transcription factors (TFs) or chemical cocktails. Challenging the expression of lineage-specific TFs is accompanied by changes in the expression of miRNAs, that post-transcriptionally modulate high numbers of neurogenesis-promoting factors and have therefore been introduced, supplementary or alternatively to TFs, to instruct direct neuronal reprogramming. The neurogenic miRNA miR-124 has been employed in direct reprogramming protocols supplementary to neurogenic TFs and other miRNAs to enhance direct neurogenic conversion by suppressing multiple non-neuronal targets. In our group we aimed to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced-neurons (iNs) on its own both in vitro and in vivo and elucidate its independent mechanism of reprogramming action. Our in vitro data indicate that miR-124 is a potent driver of the reprogramming switch of astrocytes towards an immature neuronal fate. Elucidation of the molecular pathways being triggered by miR-124 by RNA-seq analysis revealed that miR-124 is sufficient to instruct reprogramming of cortical astrocytes to immature induced-neurons (iNs) in vitro by down-regulating genes with important regulatory roles in astrocytic function. Among these, the RNA binding protein Zfp36l1, implicated in ARE-mediated mRNA decay, was found to be a direct target of miR-124, that be its turn targets neuronal-specific proteins participating in cortical development, which get de-repressed in miR-124-iNs. Furthermore, miR-124 is potent to guide direct neuronal reprogramming of reactive astrocytes to iNs of cortical identity following cortical trauma, a novel finding confirming its robust reprogramming action within the cortical microenvironment under neuroinflammatory conditions. In parallel to their reprogramming properties, astrocytes also participate in the maintenance of blood-brain barrier integrity, which ensures the physiological functioning of the central nervous system and gets affected contributing to the pathology of several neurodegenerative diseases. To study in real time the dynamic physical interactions of astrocytes with brain vasculature under homeostatic and pathological conditions, we performed 2-photon brain intravital imaging in a mouse model of systemic neuroinflammation, known to trigger astrogliosis and microgliosis and to evoke changes in astrocytic contact with brain vasculature. Our in vivo findings indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, however this event is at compensated by the cross-talk of astrocytes with activated microglia, safeguarding blood vessel coverage and maintenance of blood-brain integrity.

SeminarNeuroscience

Malignant synaptic plasticity in pediatric high-grade gliomas

Kathryn Taylor
Stanford
May 24, 2022

Pediatric high-grade gliomas (pHGG) are a devastating group of diseases that urgently require novel therapeutic options. We have previously demonstrated that pHGGs directly synapse onto neurons and the subsequent tumor cell depolarization, mediated by calcium-permeable AMPA channels, promotes their proliferation. The regulatory mechanisms governing these postsynaptic connections are unknown. Here, we investigated the role of BDNF-TrkB signaling in modulating the plasticity of the malignant synapse. BDNF ligand activation of its canonical receptor, TrkB (which is encoded for by the gene NTRK2), has been shown to be one important modulator of synaptic regulation in the normal setting. Electrophysiological recordings of glioma cell membrane properties, in response to acute neurotransmitter stimulation, demonstrate in an inward current resembling AMPA receptor (AMPAR) mediated excitatory neurotransmission. Extracellular BDNF increases the amplitude of this glutamate-induced tumor cell depolarization and this effect is abrogated in NTRK2 knockout glioma cells. Upon examining tumor cell excitability using in situ calcium imaging, we found that BDNF increases the intensity of glutamate-evoked calcium transients in GCaMP6s expressing glioma cells. Western blot analysis indicates the tumors AMPAR properties are altered downstream of BDNF induced TrkB activation in glioma. Cell membrane protein capture (via biotinylation) and live imaging of pH sensitive GFP-tagged AMPAR subunits demonstrate an increase of calcium permeable channels at the tumors postsynaptic membrane in response to BDNF. We find that BDNF-TrkB signaling promotes neuron-to-glioma synaptogenesis as measured by high-resolution confocal and electron microscopy in culture and tumor xenografts. Our analysis of published pHGG transcriptomic datasets, together with brain slice conditioned medium experiments in culture, indicates the tumor microenvironment as the chief source of BDNF ligand. Disruption of the BDNF-TrkB pathway in patient-derived orthotopic glioma xenograft models, both genetically and pharmacologically, results in an increased overall survival and reduced tumor proliferation rate. These findings suggest that gliomas leverage normal mechanisms of plasticity to modulate the excitatory channels involved in synaptic neurotransmission and they reveal the potential to target the regulatory components of glioma circuit dynamics as a therapeutic strategy for these lethal cancers.

SeminarNeuroscienceRecording

Exploring mechanisms of human brain expansion in cerebral organoids

Madeline Lancaster
MRC Laboratory of Molecular Biology, Cambridge
May 16, 2022

The human brain sets us apart as a species, with its size being one of its most striking features. Brain size is largely determined during development as vast numbers of neurons and supportive glia are generated. In an effort to better understand the events that determine the human brain’s cellular makeup, and its size, we use a human model system in a dish, called cerebral organoids. These 3D tissues are generated from pluripotent stem cells through neural differentiation and a supportive 3D microenvironment to generate organoids with the same tissue architecture as the early human fetal brain. Such organoids are allowing us to tackle questions previously impossible with more traditional approaches. Indeed, our recent findings provide insight into regulation of brain size and neuron number across ape species, identifying key stages of early neural stem cell expansion that set up a larger starting cell number to enable the production of increased numbers of neurons. We are also investigating the role of extrinsic regulators in determining numbers and types of neurons produced in the human cerebral cortex. Overall, our findings are pointing to key, human-specific aspects of brain development and function, that have important implications for neurological disease.

SeminarNeuroscience

From function to cognition: New spectroscopic tools for studying brain neurochemistry in-vivo

Assaf Tal
Weizmann Institute
Apr 21, 2021

In this seminar, I will present new methods in magnetic resonance spectroscopy (MRS) we’ve been working on in the lab. The talk will be divided into two parts. In the first, I will talk about neurochemical changes we observe in glutamate and GABA during various paradigms, including simple motors tasks and reinforcement learning. In the second part, I’ll present a new approach to MRS that focuses on measuring the relaxation times (T1, T2) of metabolites, which reflect changes to specific cellular microenvironments. I will explain why these can be exciting markers for studying several in-vivo pathologies, and also present some preliminary data from a cohort of mild cognitive impairment (MCI) patients, showing changes that correlate to cognitive decline.

SeminarNeuroscienceRecording

Microenvironment role in axonal regeneration- looking beyond the neurons

Oshri Avraham
Wash U
Oct 27, 2020

After an injury in the adult mammalian central nervous system, lesioned axons fail to regenerate. This failure to regenerate contrasts with the remarkable potential of axons to grow during embryonic development and after an injury in the peripheral nervous system. Peripheral sensory neurons with cell soma in dorsal root ganglia (DRG) switch to a regenerative state after nerve injury to enable axon regeneration and functional recovery. Decades of research have focused on the signaling pathways elicited by injury in sensory neurons and in Schwann cells that insulate axons as central mechanisms regulating nerve repair. However, neuronal microenvironment is far more complex and is composed of multiple cell types including endothelial, immune and glial cells. Whether the microenvironment surrounding neuronal soma contribute to the poor regenerative outcomes following central injuries remains largely unexplored. To answer this question, we performed a single cell transcriptional profiling of the DRG neuronal microenvironment response to peripheral and central injuries. In dissecting the roles of the microenvironment contribution, we have focused on a poorly studied population of Satellite Glial Cells (SGC) surrounding the neuronal cell soma. This study has uncovered a previously unknown role for SGC in nerve regeneration and defined SGC as transcriptionally distinct from Schwann cells while sharing similarities with astrocytes. Upon a peripheral injury, SGC contribute to axon regeneration via Fatty acid synthase (Fasn)-PPARα signaling pathway. Through repurposing fenofibrate, an FDA- approved PPARα agonist used for dyslipidemia treatment, we were able to rescue the impaired regeneration in mice lacking Fasn in SGC. Our analysis reveals that in response to central injuries, SGC do not activate the PPAR signaling pathway. However, induction of this pathway with fenofibrate treatment, rescued axon regeneration following an injury to the central nerves. Collectively, our results uncovered a previously unappreciated role of the neuronal microenvironment differential response in central and peripheral injuries.

SeminarPhysics of LifeRecording

Building a synthetic cell: Understanding the clock design and function

Qiong Yang
U Michigan - Ann Arbor
Oct 19, 2020

Clock networks containing the same central architectures may vary drastically in their potential to oscillate, raising the question of what controls robustness, one of the essential functions of an oscillator. We computationally generate an atlas of oscillators and found that, while core topologies are critical for oscillations, local structures substantially modulate the degree of robustness. Strikingly, two local structures, incoherent and coherent inputs, can modify a core topology to promote and attenuate its robustness, additively. The findings underscore the importance of local modifications to the performance of the whole network. It may explain why auxiliary structures not required for oscillations are evolutionary conserved. We also extend this computational framework to search hidden network motifs for other clock functions, such as tunability that relates to the capabilities of a clock to adjust timing to external cues. Experimentally, we developed an artificial cell system in water-in-oil microemulsions, within which we reconstitute mitotic cell cycles that can perform self-sustained oscillations for 30 to 40 cycles over multiple days. The oscillation profiles, such as period, amplitude, and shape, can be quantitatively varied with the concentrations of clock regulators, energy levels, droplet sizes, and circuit design. Such innate flexibility makes it crucial to studying clock functions of tunability and stochasticity at the single-cell level. Combined with a pressure-driven multi-channel tuning setup and long-term time-lapse fluorescence microscopy, this system enables a high-throughput exploration in multi-dimension continuous parameter space and single-cell analysis of the clock dynamics and functions. We integrate this experimental platform with mathematical modeling to elucidate the topology-function relation of biological clocks. With FRET and optogenetics, we also investigate spatiotemporal cell-cycle dynamics in both homogeneous and heterogeneous microenvironments by reconstructing subcellular compartments.

SeminarNeuroscienceRecording

Sparks, flames, and inferno: epileptogenesis in the glioblastoma microenvironment

Jeff Noebels
Baylor College of Medicine
Oct 6, 2020

Glioblastoma cells trigger pharmacoresistant seizures that may promote tumor growth and diminish the quality of remaining life. To define the relationship between growth of glial tumors and their neuronal microenvironment, and to identify genomic biomarkers and mechanisms that may point to better prognosis and treatment of drug resistant epilepsy in brain cancer, we are analyzing a new generation of genetically defined CRISPR/in utero electroporation inborn glioblastoma (GBM) tumor models engineered in mice. The molecular pathophysiology of glioblastoma cells and surrounding neurons and untransformed astrocytes are compared at serial stages of tumor development. Initial studies reveal that epileptiform EEG spiking is a very early and reliable preclinical signature of GBM expansion in these mice, followed by rapidly progressive seizures and death within weeks. FACS-sorted transcriptomic analysis of cortical astrocytes reveals the expansion of a subgroup enriched in pro-synaptogenic genes that may drive hyperexcitability, a novel mechanism of epileptogenesis. Using a prototypical GBM IUE model, we systematically define and correlate the earliest appearance of cortical hyperexcitability with progressive cortical tumor cell invasion, including spontaneous episodes of spreading cortical depolarization, innate inflammation, and xCT upregulation in the peritumoral microenvironment. Blocking this glutamate exporter reduces seizure load. We show that the host genome contributes to seizure risk by generating tumors in a monogenic deletion strain (MapT/tau -/-) that raises cortical seizure threshold. We also show that the tumor variant profile determines epilepsy risk. Our genetic dissection approach sets the stage to broadly explore the developmental biology of personalized tumor/host interactions in mice engineered with novel human tumor mutations in specified glial cell lineages.

ePoster

Assessment of metabolome in the tumor microenvironment by cerebral open flow microperfusion (cOFM) in a human glioblastoma xenograft animal model

Thomas Altendorfer-Kroath, Denise Schimek, Fernanda Monedeiro, Elmar Zügner, Eva-Maria Prugger, Christoph Magnes, Thomas Birngruber

FENS Forum 2024

ePoster

Prodromal gut dysbiosis, anhedonia and depression-like behavior in the A53T mouse model of Parkinson’s disease – the impact of social microenvironment

Olga Dubljević, Dušanka Popović, Željko Pavković, Milica Potrebić, Srbovan Maja, Emilija Brdarić, Vesna Pešić

FENS Forum 2024