Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell ; 181(2): 219-222, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302564

ABSTRACT

Mounting evidence indicates that the nervous system plays a central role in cancer pathogenesis. In turn, cancers and cancer therapies can alter nervous system form and function. This Commentary seeks to describe the burgeoning field of "cancer neuroscience" and encourage multidisciplinary collaboration for the study of cancer-nervous system interactions.


Subject(s)
Neoplasms/metabolism , Nervous System/metabolism , Humans , Neurosciences
2.
Cell ; 162(5): 1127-39, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26279190

ABSTRACT

The peripheral nervous system has remarkable regenerative capacities in that it can repair a fully cut nerve. This requires Schwann cells to migrate collectively to guide regrowing axons across a 'bridge' of new tissue, which forms to reconnect a severed nerve. Here we show that blood vessels direct the migrating cords of Schwann cells. This multicellular process is initiated by hypoxia, selectively sensed by macrophages within the bridge, which via VEGF-A secretion induce a polarized vasculature that relieves the hypoxia. Schwann cells then use the blood vessels as "tracks" to cross the bridge taking regrowing axons with them. Importantly, disrupting the organization of the newly formed blood vessels in vivo, either by inhibiting the angiogenic signal or by re-orienting them, compromises Schwann cell directionality resulting in defective nerve repair. This study provides important insights into how the choreography of multiple cell-types is required for the regeneration of an adult tissue.


Subject(s)
Blood Vessels/metabolism , Macrophages/metabolism , Peripheral Nerves/physiology , Schwann Cells/metabolism , Animals , Axons/metabolism , Cell Hypoxia , Endothelial Cells/metabolism , Inflammation/metabolism , Male , Mice , Neovascularization, Physiologic , Rats , Rats, Sprague-Dawley , Regeneration , Vascular Endothelial Growth Factor A/genetics
3.
Cell ; 154(6): 1194-205, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034244

ABSTRACT

An adult animal consists of cells of vastly different size and activity, but the regulation of cell size remains poorly understood. Recent studies uncovering some of the signaling pathways important for size/growth control, together with the identification of diseases resulting from aberrations in these pathways, have renewed interest in this field. This Review will discuss our current understanding of how a cell sets its size, how it can adapt its size to a changing environment, and how these processes are relevant to human disease.


Subject(s)
Cell Size , Animals , Cell Cycle , Cell Proliferation , Homeostasis , Humans , Neoplasms/metabolism , Neoplasms/pathology , Organ Size , Signal Transduction
4.
Cell ; 143(1): 145-55, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20869108

ABSTRACT

The peripheral nervous system has astonishing regenerative capabilities in that cut nerves are able to reconnect and re-establish their function. Schwann cells are important players in this process, during which they dedifferentiate to a progenitor/stem cell and promote axonal regrowth. Here, we report that fibroblasts also play a key role. Upon nerve cut, ephrin-B/EphB2 signaling between fibroblasts and Schwann cells results in cell sorting, followed by directional collective cell migration of Schwann cells out of the nerve stumps to guide regrowing axons across the wound. Mechanistically, we find that cell-sorting downstream of EphB2 is mediated by the stemness factor Sox2 through N-cadherin relocalization to Schwann cell-cell contacts. In vivo, loss of EphB2 signaling impaired organized migration of Schwann cells, resulting in misdirected axonal regrowth. Our results identify a link between Ephs and Sox proteins, providing a mechanism by which progenitor cells can translate environmental cues to orchestrate the formation of new tissue.


Subject(s)
Nerve Regeneration , Peripheral Nerves/physiology , Receptor, EphB2/metabolism , SOXB1 Transcription Factors/metabolism , Schwann Cells/physiology , Animals , Axons/metabolism , Cadherins/metabolism , Cell Movement , Extracellular Matrix/metabolism , Fibroblasts/physiology , Rats , Schwann Cells/cytology , Signal Transduction
5.
Development ; 145(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30413560

ABSTRACT

Peripheral nerves are highly regenerative, in contrast to the poor regenerative capabilities of the central nervous system (CNS). Here, we show that adult peripheral nerve is a more quiescent tissue than the CNS, yet all cell types within a peripheral nerve proliferate efficiently following injury. Moreover, whereas oligodendrocytes are produced throughout life from a precursor pool, we find that the corresponding cell of the peripheral nervous system, the myelinating Schwann cell (mSC), does not turn over in the adult. However, following injury, all mSCs can dedifferentiate to the proliferating progenitor-like Schwann cells (SCs) that orchestrate the regenerative response. Lineage analysis shows that these newly migratory, progenitor-like cells redifferentiate to form new tissue at the injury site and maintain their lineage, but can switch to become a non-myelinating SC. In contrast, increased plasticity is observed during tumourigenesis. These findings show that peripheral nerves have a distinct mechanism for maintaining homeostasis and can regenerate without the need for an additional stem cell population.This article has an associated 'The people behind the papers' interview.


Subject(s)
Central Nervous System/physiology , Homeostasis , Nerve Regeneration/physiology , Neural Stem Cells/cytology , Peripheral Nerves/physiology , Animals , Axons/metabolism , Carcinogenesis/pathology , Cell Proliferation , Extracellular Matrix Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Neural Stem Cells/metabolism , Neuronal Plasticity , Peripheral Nerves/cytology , Peripheral Nerves/ultrastructure , Schwann Cells/metabolism
6.
Development ; 144(17): 3114-3125, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28743796

ABSTRACT

Correct myelination is crucial for the function of the peripheral nervous system. Both positive and negative regulators within the axon and Schwann cell function to ensure the correct onset and progression of myelination during both development and following peripheral nerve injury and repair. The Sox2 transcription factor is well known for its roles in the development and maintenance of progenitor and stem cell populations, but has also been proposed in vitro as a negative regulator of myelination in Schwann cells. We wished to test fully whether Sox2 regulates myelination in vivo and show here that, in mice, sustained Sox2 expression in vivo blocks myelination in the peripheral nerves and maintains Schwann cells in a proliferative non-differentiated state, which is also associated with increased inflammation within the nerve. The plasticity of Schwann cells allows them to re-myelinate regenerated axons following injury and we show that re-myelination is also blocked by Sox2 expression in Schwann cells. These findings identify Sox2 as a physiological regulator of Schwann cell myelination in vivo and its potential to play a role in disorders of myelination in the peripheral nervous system.


Subject(s)
Macrophages/metabolism , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , SOXB1 Transcription Factors/metabolism , Schwann Cells/metabolism , Animals , Biomarkers/metabolism , Cadherins/metabolism , Cell Proliferation , Early Growth Response Protein 2/metabolism , Green Fluorescent Proteins/metabolism , Mice, Transgenic , Motor Activity , Neural Conduction , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nerves/pathology , Peripheral Nerves/ultrastructure , Proto-Oncogene Proteins c-jun/metabolism , Rats , Recovery of Function , Schwann Cells/pathology , Transgenes , beta Catenin/metabolism
7.
Glia ; 67(11): 2203-2215, 2019 11.
Article in English | MEDLINE | ID: mdl-31215712

ABSTRACT

How tissues are maintained over a lifetime and repaired following injury are fundamental questions in biology with a disruption to these processes underlying pathologies such as cancer and degenerative disorders. It is becoming increasingly clear that each tissue has a distinct mechanism to maintain homeostasis and respond to injury utilizing different types of stem/progenitor cell populations depending on the insult and/or with a contribution from more differentiated cells that are able to dedifferentiate to aid tissue regeneration. Peripheral nerves are highly quiescent yet show remarkable regenerative capabilities. Remarkably, there is no evidence for a classical stem cell population, rather all cell-types within the nerve are able to proliferate to produce new nerve tissue. Co-ordinating the regeneration of this tissue are Schwann cells (SCs), the main glial cells of the peripheral nervous system. SCs exist in architecturally stable structures that can persist for the lifetime of an animal, however, they are not postmitotic, in that following injury they are reprogrammed at high efficiency to a progenitor-like state, with these cells acting to orchestrate the nerve regeneration process. During nerve regeneration, SCs show little plasticity, maintaining their identity in the repaired tissue. However, once free of the nerve environment they appear to exhibit increased plasticity with reported roles in the repair of other tissues. In this review, we will discuss the mechanisms underlying the homeostasis and regeneration of peripheral nerves and how reprogrammed progenitor-like SCs have broader roles in the repair of other tissues with implications for pathologies such as cancer.


Subject(s)
Cell Plasticity/physiology , Homeostasis/physiology , Nerve Regeneration/physiology , Peripheral Nerves/physiology , Animals , Cell Differentiation/physiology , Humans , Neuronal Plasticity , Peripheral Nerves/pathology , Schwann Cells/metabolism
8.
Am J Med Genet A ; 173(6): 1714-1721, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28436162

ABSTRACT

The Annual Children's Tumor Foundation International Neurofibromatosis Meeting is the premier venue for connecting discovery, translational and clinical scientists who are focused on neurofibromatosis types 1 and 2 (NF1 and NF2) and schwannomatosis (SWN). The meeting also features rare tumors such as glioma, meningioma, sarcoma, and neuroblastoma that occur both within these syndromes and spontaneously; associated with somatic mutations in NF1, NF2, and SWN. The meeting addresses both state of the field for current clinical care as well as emerging preclinical models fueling discovery of new therapeutic targets and discovery science initiatives investigating mechanisms of tumorigenesis. Importantly, this conference is a forum for presenting work in progress and bringing together all stakeholders in the scientific community. A highlight of the conference was the involvement of scientists from the pharmaceutical industry who presented growing efforts for rare disease therapeutic development in general and specifically, in pediatric patients with rare tumor syndromes. Another highlight was the focus on new investigators who presented new data about biomarker discovery, tumor pathogenesis, and diagnostic tools for NF1, NF2, and SWN. This report summarizes the themes of the meeting and a synthesis of the scientific discoveries presented at the conference in order to make the larger research community aware of progress in the neurofibromatoses.


Subject(s)
Neurilemmoma/therapy , Neurofibromatoses/therapy , Neurofibromatosis 1/therapy , Neurofibromatosis 2/therapy , Skin Neoplasms/therapy , Child , Humans , Neurilemmoma/genetics , Neurofibromatoses/genetics , Neurofibromatosis 1/genetics , Neurofibromatosis 2/genetics , Pediatrics/trends , Skin Neoplasms/genetics
9.
Genome Res ; 21(4): 515-24, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21324880

ABSTRACT

Aberrant DNA methylation (DNAm) was first linked to cancer over 25 yr ago. Since then, many studies have associated hypermethylation of tumor suppressor genes and hypomethylation of oncogenes to the tumorigenic process. However, most of these studies have been limited to the analysis of promoters and CpG islands (CGIs). Recently, new technologies for whole-genome DNAm (methylome) analysis have been developed, enabling unbiased analysis of cancer methylomes. By using MeDIP-seq, we report a sequencing-based comparative methylome analysis of malignant peripheral nerve sheath tumors (MPNSTs), benign neurofibromas, and normal Schwann cells. Analysis of these methylomes revealed a complex landscape of DNAm alterations. In contrast to what has been reported for other tumor types, no significant global hypomethylation was observed in MPNSTs using methylome analysis by MeDIP-seq. However, a highly significant (P < 10(-100)) directional difference in DNAm was found in satellite repeats, suggesting these repeats to be the main target for hypomethylation in MPNSTs. Comparative analysis of the MPNST and Schwann cell methylomes identified 101,466 cancer-associated differentially methylated regions (cDMRs). Analysis showed these cDMRs to be significantly enriched for two satellite repeat types (SATR1 and ARLα) and suggests an association between aberrant DNAm of these sequences and transition from healthy cells to malignant disease. Significant enrichment of hypermethylated cDMRs in CGI shores (P < 10(-60)), non-CGI-associated promoters (P < 10(-4)) and hypomethylated cDMRs in SINE repeats (P < 10(-100)) was also identified. Integration of DNAm and gene expression data showed that the expression pattern of genes associated with CGI shore cDMRs was able to discriminate between disease phenotypes. This study establishes MeDIP-seq as an effective method to analyze cancer methylomes.


Subject(s)
DNA Methylation/genetics , Epigenomics , Nerve Sheath Neoplasms/genetics , Cluster Analysis , CpG Islands/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Minisatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis/methods
10.
Article in English | MEDLINE | ID: mdl-38199866

ABSTRACT

Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.


Subject(s)
Nerve Regeneration , Schwann Cells , Schwann Cells/physiology , Nerve Regeneration/physiology , Humans , Animals , Peripheral Nerves/physiology , Axons/physiology
11.
Elife ; 132024 Apr 09.
Article in English | MEDLINE | ID: mdl-38591541

ABSTRACT

Collective cell migration is fundamental for the development of organisms and in the adult for tissue regeneration and in pathological conditions such as cancer. Migration as a coherent group requires the maintenance of cell-cell interactions, while contact inhibition of locomotion (CIL), a local repulsive force, can propel the group forward. Here we show that the cell-cell interaction molecule, N-cadherin, regulates both adhesion and repulsion processes during Schwann cell (SC) collective migration, which is required for peripheral nerve regeneration. However, distinct from its role in cell-cell adhesion, the repulsion process is independent of N-cadherin trans-homodimerisation and the associated adherens junction complex. Rather, the extracellular domain of N-cadherin is required to present the repulsive Slit2/Slit3 signal at the cell surface. Inhibiting Slit2/Slit3 signalling inhibits CIL and subsequently collective SC migration, resulting in adherent, nonmigratory cell clusters. Moreover, analysis of ex vivo explants from mice following sciatic nerve injury showed that inhibition of Slit2 decreased SC collective migration and increased clustering of SCs within the nerve bridge. These findings provide insight into how opposing signals can mediate collective cell migration and how CIL pathways are promising targets for inhibiting pathological cell migration.


Subject(s)
Cadherins , Cell Movement , Contact Inhibition , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Nerve Regeneration , Nerve Tissue Proteins , Schwann Cells , Schwann Cells/metabolism , Schwann Cells/physiology , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice , Cadherins/metabolism , Cadherins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Nerve Regeneration/physiology , Locomotion/physiology , Cell Adhesion , Signal Transduction
12.
Dev Cell ; 58(3): 174-191.e8, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36706755

ABSTRACT

The blood barriers of the nervous system protect neural environments but can hinder therapeutic accessibility. The blood-brain barrier (BBB) is well characterized, consisting of endothelial cells with specialized tight junctions and low levels of transcytosis, properties conferred by contacting pericytes and astrocytes. In contrast, the blood-nerve barrier (BNB) of the peripheral nervous system is poorly defined. Here, we characterize the structure of the mammalian BNB, identify the processes that confer barrier function, and demonstrate how the barrier can be opened in response to injury. The homeostatic BNB is leakier than the BBB, which we show is due to higher levels of transcytosis. However, the barrier is reinforced by macrophages that specifically engulf leaked materials, identifying a role for resident macrophages as an important component of the BNB. Finally, we demonstrate the exploitation of these processes to effectively deliver RNA-targeting therapeutics to peripheral nerves, indicating new treatment approaches for nervous system pathologies.


Subject(s)
Blood-Nerve Barrier , Endothelial Cells , Animals , Blood-Nerve Barrier/physiology , Endothelial Cells/physiology , Blood-Brain Barrier/physiology , Macrophages , Pericytes/physiology , Mammals
13.
Acta Neuropathol ; 123(3): 369-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22083253

ABSTRACT

The 2011 annual meeting of the Children's Tumor Foundation, the annual gathering of the neurofibromatosis (NF) research and clinical communities, was attended by 330 participants who discussed integration of new signaling pathways into NF research, the appreciation for NF mutations in sporadic cancers, and an expanding pre-clinical and clinical agenda. NF1, NF2, and schwannomatosis collectively affect approximately 100,000 persons in US, and result from mutations in different genes. Benign tumors of NF1 (neurofibroma and optic pathway glioma) and NF2 (schwannoma, ependymoma, and meningioma) and schwannomatosis (schwannoma) can cause significant morbidity, and there are no proven drug treatments for any form of NF. Each disorder is associated with additional manifestations causing morbidity. The research presentations described in this review covered basic science, preclinical testing, and results from clinical trials, and demonstrate the remarkable strides being taken toward understanding of and progress toward treatments for these disorders based on the close interaction among scientists and clinicians.


Subject(s)
Neurofibromatosis 1/pathology , Neurofibromatosis 2/pathology , Child , Genes, Neurofibromatosis 1 , Genes, Neurofibromatosis 2 , Humans , Meningioma/genetics , Meningioma/pathology , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 2/genetics
14.
J Cell Sci ; 122(Pt 18): 3272-81, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19690052

ABSTRACT

Mammalian cells generally require both mitogens and anchorage signals in order to proliferate. An important characteristic of many tumour cells is that they have lost this anchorage-dependent cell-cycle checkpoint, allowing them to proliferate without signals provided by their normal microenvironment. In the absence of anchorage signals from the extracellular matrix, many cell types arrest cell-cycle progression in G1 phase as a result of Rb-dependent checkpoints. However, despite inactivation of p53 and Rb proteins, SV40LT-expressing cells retain anchorage dependency, suggesting the presence of an uncharacterised cell-cycle checkpoint, which can be overridden by coexpression of oncogenic Ras. We report here that, although cyclin-CDK complexes persisted in suspension, proliferation was inhibited in LT-expressing cells by the CDK inhibitor p27(Kip1) (p27). Interestingly, this did not induce a stable arrest, but aberrant cell-cycle progression associated with stalled DNA replication, rereplication and chromosomal instability, which was sufficient to increase the frequency of oncogenic transformation. These results firstly indicate loss of anchorage in Rb- and p53-deficient cells as a novel mechanism for promotion of genomic instability; secondly suggest that anchorage checkpoints that protect normal cells from inappropriate proliferation act deleteriously in Rb- and p53-deficient cells to promote tumourigenesis; and thirdly indicate caution in the use of CDK inhibitors for cancer treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Genomic Instability/genetics , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Rats , S Phase
15.
J Cell Sci ; 122(Pt 24): 4516-25, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19920079

ABSTRACT

Cells generate new organelles when stimulated by extracellular factors to grow and divide; however, little is known about how growth and mitogenic signalling pathways regulate organelle biogenesis. Using mitochondria as a model organelle, we have investigated this problem in primary Schwann cells, for which distinct factors act solely as mitogens (neuregulin) or as promoters of cell growth (insulin-like growth factor 1; IGF1). We find that neuregulin and IGF1 act synergistically to increase mitochondrial biogenesis and mitochondrial DNA replication, resulting in increased mitochondrial density in these cells. Moreover, constitutive oncogenic Ras signalling results in a further increase in mitochondrial density. This synergistic effect is seen at the global transcriptional level, requires both the ERK and phosphoinositide 3-kinase (PI3K) signalling pathways and is mediated by the transcription factor ERRalpha. Interestingly, the effect is independent of Akt-TOR signalling, a major regulator of cell growth in these cells. This separation of the pathways that drive mitochondrial biogenesis and cell growth provides a mechanism for the modulation of mitochondrial density according to the metabolic requirements of the cell.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Mitogens/metabolism , Neuregulins/metabolism , Schwann Cells/metabolism , Animals , Cell Line , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitochondria/genetics , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction
16.
Nat Cell Biol ; 4(2): E25-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11835050

ABSTRACT

It has long-been accepted that normal somatic cells have intrinsic mechanisms that limit their proliferative lifespan. Recent work has now challenged this view by demonstrating that extrinsic factors might be determining proliferative potential.


Subject(s)
Cell Culture Techniques/methods , Cell Division/physiology , Cellular Senescence/physiology , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Phenotype , Signal Transduction/physiology , Telomerase/metabolism
18.
Cell Rep ; 26(6): 1458-1472.e4, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726731

ABSTRACT

Slit-Robo signaling has been characterized as a repulsive signal for precise axon pathfinding and cell migration during embryonic development. Here, we describe a role for Sox2 in the regulation of Robo1 in Schwann cells and for Slit3-Robo1 signaling in controlling axon guidance within the newly formed nerve bridge following peripheral nerve transection injury. In particular, we show that macrophages form the outermost layer of the nerve bridge and secrete high levels of Slit3, while migratory Schwann cells and fibroblasts inside the nerve bridge express the Robo1 receptor. In line with this pattern of Slit3 and Robo1 expression, we observed multiple axon regeneration and cell migration defects in the nerve bridge of Sox2-, Slit3-, and Robo1-mutant mice. Our findings have revealed important functions for macrophages in the peripheral nervous system, utilizing Slit3-Robo1 signaling to control correct peripheral nerve bridge formation and precise axon targeting to the distal nerve stump following injury.


Subject(s)
Axon Guidance , Macrophages/metabolism , Membrane Proteins/metabolism , Nerve Regeneration , Peripheral Nerves/metabolism , Animals , Cell Movement , Cells, Cultured , Female , Fibroblasts/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peripheral Nerves/physiology , Rats , Rats, Wistar , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Schwann Cells/metabolism , Signal Transduction , Roundabout Proteins
19.
Trends Pharmacol Sci ; 28(3): 103-5, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17276519

ABSTRACT

The leprosy pathogen Mycobacterium leprae attacks Schwann cells in the peripheral nervous system, causing them to demyelinate. Recent work by Tapinos et al. shows that a direct mechanism of demyelination induced by M. leprae depends on the binding of the bacterium to the receptor tyrosine kinase ErbB2 on Schwann cells and the resulting activation of the Ras-Raf-MEK-ERK pathway. These findings have relevance for the potential treatment of leprosy and they highlight parallels between the dedifferentiation signal in leprosy and that in nerve injury and cancer.


Subject(s)
Leprostatic Agents/therapeutic use , Leprosy/drug therapy , Mycobacterium leprae/metabolism , Receptor, ErbB-2/metabolism , Humans , Leprostatic Agents/economics , Leprosy/metabolism , Leprosy/microbiology , Models, Biological , Mycobacterium leprae/genetics , Protein Binding/drug effects , Schwann Cells/drug effects , Schwann Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL