Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrea Tedeschi is active.

Publication


Featured researches published by Andrea Tedeschi.


Nature Neuroscience | 2010

PTEN deletion enhances the regenerative ability of adult corticospinal neurons

Kai Liu; Yi Lu; Jae K. Lee; Ramsey F. Samara; Rafer Willenberg; Ilse Sears-Kraxberger; Andrea Tedeschi; Kevin Park; Duo Jin; Bin Cai; Bengang Xu; Lauren Connolly; Oswald Steward; Binhai Zheng; Zhigang He

Despite the essential role of the corticospinal tract (CST) in controlling voluntary movements, successful regeneration of large numbers of injured CST axons beyond a spinal cord lesion has never been achieved. We found that PTEN/mTOR are critical for controlling the regenerative capacity of mouse corticospinal neurons. After development, the regrowth potential of CST axons was lost and this was accompanied by a downregulation of mTOR activity in corticospinal neurons. Axonal injury further diminished neuronal mTOR activity in these neurons. Forced upregulation of mTOR activity in corticospinal neurons by conditional deletion of Pten, a negative regulator of mTOR, enhanced compensatory sprouting of uninjured CST axons and enabled successful regeneration of a cohort of injured CST axons past a spinal cord lesion. Furthermore, these regenerating CST axons possessed the ability to reform synapses in spinal segments distal to the injury. Thus, modulating neuronal intrinsic PTEN/mTOR activity represents a potential therapeutic strategy for promoting axon regeneration and functional repair after adult spinal cord injury.


Annual Review of Neuroscience | 2011

Neuronal Intrinsic Mechanisms of Axon Regeneration

Kai Liu; Andrea Tedeschi; Kevin Park; Zhigang He

Failure of axon regeneration after central nervous system (CNS) injuries results in permanent functional deficits. Numerous studies in the past suggested that blocking extracellular inhibitory influences alone is insufficient to allow the majority of injured axons to regenerate, pointing to the importance of revisiting the hypothesis that diminished intrinsic regenerative ability critically underlies regeneration failure. Recent studies in different species and using different injury models have started to reveal important cellular and molecular mechanisms within neurons that govern axon regeneration. This review summarizes these observations and discusses possible strategies for stimulating axon regeneration and perhaps functional recovery after CNS injury.


Science | 2015

Systemic administration of epothilone B promotes axon regeneration after spinal cord injury

Jörg Ruschel; Farida Hellal; Kevin C. Flynn; Sebastian Dupraz; David A. Elliott; Andrea Tedeschi; Margaret L. Bates; Gary Brook; Kristina Dobrindt; Michael Peitz; Oliver Brüstle; Michael D. Norenberg; Armin Blesch; Norbert Weidner; Mary Bartlett Bunge; John L. Bixby; Frank Bradke

Progress toward fixing a broken back? Axon regeneration after a spinal cord injury requires interference with neuronal mechanisms to promote axon extension and early suppression of scar formation. Microtubule stabilization could provide, in principle, a basis for such intervention. Ruschel et al. used animal models of spinal cord injury, time-lapse imaging in vivo, primary neuronal cultures, and behavioral studies to tackle this challenge (see the Perspective by Tran and Silver). They showed that epothilone B, a U.S. Food and Drug Administration–approved microtubule-stabilizing drug that can cross the blood-brain barrier, does promote functional axon regeneration, even after injury. Science, this issue p. 347; see also p. 285 Stabilizing microtubules after a spinal cord injury reduces the migratory activity of scar-forming meningeal fibroblasts. [Also see Perspective by Tran and Silver] After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier–permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.


Science | 2015

Axonal regeneration. Systemic administration of epothilone B promotes axon regeneration after spinal cord injury.

Jörg Ruschel; Farida Hellal; Kevin C. Flynn; Sebastian Dupraz; David A. Elliott; Andrea Tedeschi; Margaret L. Bates; Gary Brook; Kristina Dobrindt; Michael Peitz; Oliver Brüstle; Norenberg; Armin Blesch; Norbert Weidner; Mary Bartlett Bunge; John L. Bixby; Frank Bradke

Progress toward fixing a broken back? Axon regeneration after a spinal cord injury requires interference with neuronal mechanisms to promote axon extension and early suppression of scar formation. Microtubule stabilization could provide, in principle, a basis for such intervention. Ruschel et al. used animal models of spinal cord injury, time-lapse imaging in vivo, primary neuronal cultures, and behavioral studies to tackle this challenge (see the Perspective by Tran and Silver). They showed that epothilone B, a U.S. Food and Drug Administration–approved microtubule-stabilizing drug that can cross the blood-brain barrier, does promote functional axon regeneration, even after injury. Science, this issue p. 347; see also p. 285 Stabilizing microtubules after a spinal cord injury reduces the migratory activity of scar-forming meningeal fibroblasts. [Also see Perspective by Tran and Silver] After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier–permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.


EMBO Reports | 2013

The DLK signalling pathway—a double-edged sword in neural development and regeneration

Andrea Tedeschi; Frank Bradke

Dual leucine zipper kinase (DLK), a mitogen‐activated protein kinase kinase kinase, controls axon growth, apoptosis and neuron degeneration during neural development, as well as neurodegeneration after various insults to the adult nervous system. Interestingly, recent studies have also highlighted a role of DLK in promoting axon regeneration in diverse model systems. Invertebrates and vertebrates, cold‐ and warm‐blooded animals, as well as central and peripheral mammalian nervous systems all differ in their ability to regenerate injured axons. Here, we discuss how DLK‐dependent signalling regulates apparently contradictory functions during neural development and regeneration in different species. In addition, we outline strategies to fine‐tune DLK function, either alone or together with other approaches, to promote axon regeneration in the adult mammalian central nervous system.


Frontiers in Molecular Neuroscience | 2012

Tuning the orchestra: transcriptional pathways controlling axon regeneration.

Andrea Tedeschi

Trauma in the adult mammalian central nervous system leads to irreversible structural and functional impairment due to failed regeneration attempts. In contrast, neurons in the peripheral nervous system exhibit a greater regenerative ability. It has been proposed that an orchestrated sequence of transcriptional events controlling the expression of specific sets of genes may be the underlying basis of an early cell-autonomous regenerative response. Understanding whether transcriptional fine tuning, in parallel with strategies aimed at counteracting extrinsic impediments promotes axon re-growth following central nervous system injuries represents an exciting challenge for future studies. Transcriptional pathways controlling axon regeneration are presented and discussed in this review.


Current Opinion in Neurobiology | 2017

Spatial and temporal arrangement of neuronal intrinsic and extrinsic mechanisms controlling axon regeneration

Andrea Tedeschi; Frank Bradke

Axon regeneration and neuronal tissue repair varies across animal lineages as well as in the mammalian central and peripheral nervous systems. While the peripheral nervous system retains the ability to self-repair, the majority of axons in the adult mammalian central nervous system (CNS) fail to reactivate intrinsic growth programs after injury. Recent findings, however, suggest that long-distance axon regeneration, neuronal circuit assembly and recovery of functions in the adult mammalian CNS are possible. Here, we discuss our current knowledge of the cell signaling pathways and networks controlling axon regeneration. In addition, we outline a number of combinatorial strategies that include among others microtubule-based treatments to foster regeneration and functional connectivity after CNS trauma.


The Journal of Neuroscience | 2014

Characterization of Long Descending Premotor Propriospinal Neurons in the Spinal Cord

Yingchun Ni; Homaira Nawabi; Xuefeng Liu; Liu Yang; Kazunari Miyamichi; Andrea Tedeschi; Bengang Xu; Nicholas R. Wall; Edward M. Callaway; Zhigang He

The motor function of the spinal cord requires the computation of the local neuronal circuits within the same segments as well as the long-range coordination of different spinal levels. Implicated players in this process are the propriospinal neurons (PPNs) that project their axons across different levels of the spinal cord. However, their cellular, molecular, and functional properties remain unknown. Here we use a recombinant rabies virus-based method to label a specific type of long-projecting premotor PPNs in the mouse upper spinal cord that are monosynaptically connected to the motor neurons in the lumbar spinal cord. With a whole spinal cord imaging method, we find that these neurons are distributed along the entire length of the upper spinal cord with more in the lower thoracic levels. Among them, a subset of thoracic PPNs receive substantial numbers of sensory inputs, suggesting a function in coordinating the activity of trunk and hindlimb muscles. Although many PPNs in the cervical and thoracic spinal cord receive the synaptic inputs from corticospinal tract or serotonergic axons, limited bouton numbers suggested that these supraspinal inputs might not be major regulators of the PPNs in intact animals. Molecularly, these PPNs appear to be distinct from other known premotor interneurons, but some are derived from Chx10+ lineages. This study provides an anatomical basis for further exploring different functions of PPNs.


Journal of Lipid Research | 2017

Localization of 1-deoxysphingolipids to mitochondria induces mitochondrial dysfunction

Irina Alecu; Andrea Tedeschi; Natascha Behler; Klaus Wunderling; Christian Lamberz; Mario A.R. Lauterbach; Anne Gaebler; Daniela Ernst; Paul P. Van Veldhoven; Ashraf Al-Amoudi; Eicke Latz; Alaa Othman; Lars Kuerschner; Thorsten Hornemann; Frank Bradke; Christoph Thiele; Anke Penno

1-Deoxysphingolipids (deoxySLs) are atypical sphingolipids that are elevated in the plasma of patients with type 2 diabetes and hereditary sensory and autonomic neuropathy type 1 (HSAN1). Clinically, diabetic neuropathy and HSAN1 are very similar, suggesting the involvement of deoxySLs in the pathology of both diseases. However, very little is known about the biology of these lipids and the underlying pathomechanism. We synthesized an alkyne analog of 1-deoxysphinganine (doxSA), the metabolic precursor of all deoxySLs, to trace the metabolism and localization of deoxySLs. Our results indicate that the metabolism of these lipids is restricted to only some lipid species and that they are not converted to canonical sphingolipids or fatty acids. Furthermore, exogenously added alkyne-doxSA [(2S,3R)-2-aminooctadec-17-yn-3-ol] localized to mitochondria, causing mitochondrial fragmentation and dysfunction. The induced mitochondrial toxicity was also shown for natural doxSA, but not for sphinganine, and was rescued by inhibition of ceramide synthase activity. Our findings therefore indicate that mitochondrial enrichment of an N-acylated doxSA metabolite may contribute to the neurotoxicity seen in diabetic neuropathy and HSAN1. Hence, we provide a potential explanation for the characteristic vulnerability of peripheral nerves to elevated levels of deoxySLs.


Experimental Neurology | 2017

CNS repair and axon regeneration: Using genetic variation to determine mechanisms

Andrea Tedeschi; Takao Omura; Michael Costigan

&NA; The importance of genetic diversity in biological investigation has been recognized since the pioneering studies of Gregor Johann Mendel and Charles Darwin. Research in this area has been greatly informed recently by the publication of genomes from multiple species. Genes regulate and create every part and process in a living organism, react with the environment to create each living form and morph and mutate to determine the history and future of each species. The regenerative capacity of neurons differs profoundly between animal lineages and within the mammalian central and peripheral nervous systems. Here, we discuss research that suggests that genetic background contributes to the ability of injured axons to regenerate in the mammalian central nervous system (CNS), by controlling the regulation of specific signaling cascades. We detail the methods used to identify these pathways, which include among others Activin signaling and other TGF‐&bgr; superfamily members. We discuss the potential of altering these pathways in patients with CNS damage and outline strategies to promote regeneration and repair by combinatorial manipulation of neuron‐intrinsic and extrinsic determinants.

Collaboration


Dive into the Andrea Tedeschi's collaboration.

Top Co-Authors

Avatar

Frank Bradke

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sebastian Dupraz

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Michael Costigan

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Elliott

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Farida Hellal

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Gary Brook

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin C. Flynn

German Center for Neurodegenerative Diseases

View shared research outputs
Researchain Logo
Decentralizing Knowledge