Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 116(6): 2328-2337, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30659145

ABSTRACT

Mutations in the MFN2 gene encoding Mitofusin 2 lead to the development of Charcot-Marie-Tooth type 2A (CMT2A), a dominant axonal form of peripheral neuropathy. Mitofusin 2 is localized at both the outer membrane of mitochondria and the endoplasmic reticulum and is particularly enriched at specialized contact regions known as mitochondria-associated membranes (MAM). We observed that expression of MFN2R94Q induces distal axonal degeneration in the absence of overt neuronal death. The presence of mutant protein leads to reduction in endoplasmic reticulum and mitochondria contacts in CMT2A patient-derived fibroblasts, in primary neurons and in vivo, in motoneurons of a mouse model of CMT2A. These changes are concomitant with endoplasmic reticulum stress, calcium handling defects, and changes in the geometry and axonal transport of mitochondria. Importantly, pharmacological treatments reinforcing endoplasmic reticulum-mitochondria cross-talk, or reducing endoplasmic reticulum stress, restore the mitochondria morphology and prevent axonal degeneration. These results highlight defects in MAM as a cellular mechanism contributing to CMT2A pathology mediated by mutated MFN2.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Animals , Axons/metabolism , Biological Transport , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Disease Models, Animal , Endoplasmic Reticulum/ultrastructure , Female , Gait , Locomotion/genetics , Male , Mice , Mice, Transgenic , Mitochondria/ultrastructure , Motor Neurons/metabolism , Muscle Denervation , Muscle Fibers, Slow-Twitch , Signal Transduction
2.
Glia ; 69(1): 124-136, 2021 01.
Article in English | MEDLINE | ID: mdl-32686211

ABSTRACT

Recent studies in neuron-glial metabolic coupling have shown that, in the CNS, astrocytes and oligodendrocytes support neurons with energy-rich lactate/pyruvate via monocarboxylate transporters (MCTs). The presence of such transporters in the PNS, in both Schwann cells and neurons, has prompted us to question if a similar interaction may be present. Here we describe the generation and characterization of conditional knockout mouse models where MCT1 or MCT4 is specifically deleted in Schwann cells (named MCT1 and MCT4 cKO). We show that MCT1 cKO and MCT4 cKO mice develop normally and that myelin in the PNS is preserved. However, MCT1 expressed by Schwann cells is necessary for long-term maintenance of motor end-plate integrity as revealed by disrupted neuromuscular innervation in mutant mice, while MCT4 appears largely dispensable for the support of motor neurons. Concomitant to detected structural alterations, lumbar motor neurons from MCT1 cKO mice show transcriptional changes affecting cytoskeletal components, transcriptional regulators, and mitochondria related transcripts, among others. Together, our data indicate that MCT1 plays a role in Schwann cell-mediated maintenance of motor end-plate innervation thus providing further insight into the emerging picture of the biology of the axon-glia metabolic crosstalk.


Subject(s)
Schwann Cells , Animals , Mice , Monocarboxylic Acid Transporters/genetics , Motor Endplate , Muscle Proteins , Myelin Sheath , Symporters/genetics
3.
Brain ; 142(5): 1227-1241, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30907403

ABSTRACT

Charcot-Marie-Tooth disease type 4C is the most common recessively inherited demyelinating neuropathy that results from loss of function mutations in the SH3TC2 gene. Sh3tc2-/- mice represent a well characterized disease model developing early onset progressive peripheral neuropathy with hypo- and demyelination, slowing of nerve conduction velocities and disturbed nodal architecture. The aim of this project was to develop a gene replacement therapy for treating Charcot-Marie-Tooth disease type 4C to rescue the phenotype of the Sh3tc2-/- mouse model. We generated a lentiviral vector LV-Mpz.SH3TC2.myc to drive expression of the human SH3TC2 cDNA under the control of the Mpz promoter specifically in myelinating Schwann cells. The vector was delivered into 3-week-old Sh3tc2-/- mice by lumbar intrathecal injection and gene expression was assessed 4-8 weeks after injection. Immunofluorescence analysis showed presence of myc-tagged human SH3TC2 in sciatic nerves and lumbar roots in the perinuclear cytoplasm of a subset of Schwann cells, in a dotted pattern co-localizing with physiologically interacting protein Rab11. Quantitative PCR analysis confirmed SH3TC2 mRNA expression in different peripheral nervous system tissues. A treatment trial was initiated in 3 weeks old randomized Sh3tc2-/- littermate mice which received either the full or mock (LV-Mpz.Egfp) vector. Behavioural analysis 8 weeks after injection showed improved motor performance in rotarod and foot grip tests in treated Sh3tc2-/- mice compared to mock vector-treated animals. Moreover, motor nerve conduction velocities were increased in treated Sh3tc2-/- mice. On a structural level, morphological analysis revealed significant improvement in g-ratios, myelin thickness, and ratios of demyelinated fibres in lumbar roots and sciatic nerves of treated Sh3tc2-/- mice. Finally, treated mice also showed improved nodal molecular architecture and reduction of blood neurofilament light levels, a clinically relevant biomarker for axonal injury/degeneration. This study provides a proof of principle for viral gene replacement therapy targeted to Schwann cells to treat Charcot-Marie-Tooth disease type 4C and potentially other similar demyelinating inherited neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Disease Models, Animal , Genetic Therapy/methods , Intracellular Signaling Peptides and Proteins/genetics , Animals , Charcot-Marie-Tooth Disease/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Knockout , Mice, Transgenic
4.
Int J Mol Sci ; 19(12)2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30562927

ABSTRACT

The neuromuscular junction (NMJ) appears to be a site of pathology in a number of peripheral nerve diseases. Charcot-Marie-Tooth (CMT) 4C is an autosomal recessive, early onset, demyelinating neuropathy. Numerous mutations in the SH3TC2 gene have been shown to underlie the condition often associated with scoliosis, foot deformities, and reduced nerve conduction velocities. Mice with exon 1 of the Sh3tc2 gene knocked out demonstrate many of the features seen in patients. To determine if NMJ pathology is contributory to the pathomechanisms of CMT4C we examined NMJs in the gastrocnemius muscle of SH3TC2-deficient mice. In addition, we performed proteomic assessment of the sciatic nerve to identify protein factors contributing to the NMJ alterations and the survival of demyelinated axons. Morphological and gene expression analysis of NMJs revealed a lack of continuity between the pre- and post-synaptic apparatus, increases in post-synaptic fragmentation and dispersal, and an increase in expression of the gamma subunit of the acetylcholine receptor. There were no changes in axonal width or the number of axonal inputs to the NMJ. Proteome investigations of the sciatic nerve revealed altered expression of extracellular matrix proteins important for NMJ integrity. Together these observations suggest that CMT4C pathology includes a compromised NMJ even in the absence of changes to the innervating axon.


Subject(s)
Carrier Proteins , Charcot-Marie-Tooth Disease , Muscle, Skeletal , Mutation , Neuromuscular Junction , Sciatic Nerve , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Exons , Gene Knockdown Techniques , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
5.
J Neurosci ; 35(10): 4151-6, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25762662

ABSTRACT

Lactate, a product of glycolysis, has been shown to play a key role in the metabolic support of neurons/axons in the CNS by both astrocytes and oligodendrocytes through monocarboxylate transporters (MCTs). Despite such importance in the CNS, little is known about MCT expression and lactate function in the PNS. Here we show that mouse MCT1, MCT2, and MCT4 are expressed in the PNS. While DRG neurons express MCT1, myelinating Schwann cells (SCs) coexpress MCT1 and MCT4 in a domain-specific fashion, mainly in regions of noncompact myelin. Interestingly, SC-specific downregulation of MCT1 expression in rat neuron/SC cocultures led to increased myelination, while its downregulation in neurons resulted in a decreased amount of neurofilament. Finally, pure rat SCs grown in the presence of lactate exhibited an increase in the level of expression of the main myelin regulator gene Krox20/Egr2 and the myelin gene P0. These data indicate that lactate homeostasis participates in the regulation of the SC myelination program and reveal that similar to CNS, PNS axon-glial metabolic interactions are most likely mediated by MCTs.


Subject(s)
Gene Expression Regulation/physiology , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , Sensory Receptor Cells/metabolism , Actins/metabolism , Age Factors , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Coculture Techniques , Early Growth Response Protein 2/genetics , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Lactic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Monocarboxylic Acid Transporters/classification , Monocarboxylic Acid Transporters/genetics , Myelin Basic Protein/metabolism , Myelin P0 Protein/genetics , Neurofilament Proteins/metabolism , Peripheral Nerves/cytology , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Schwann Cells/metabolism , Sensory Receptor Cells/drug effects
6.
Brain ; 138(Pt 4): 875-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25678561

ABSTRACT

Mutations in Sigma 1 receptor (SIGMAR1) have been previously identified in patients with amyotrophic lateral sclerosis and disruption of Sigmar1 in mouse leads to locomotor deficits. However, cellular mechanisms underlying motor phenotypes in human and mouse with disturbed SIGMAR1 function have not been described so far. Here we used a combination of in vivo and in vitro approaches to investigate the role of SIGMAR1 in motor neuron biology. Characterization of Sigmar1(-/-) mice revealed that affected animals display locomotor deficits associated with muscle weakness, axonal degeneration and motor neuron loss. Using primary motor neuron cultures, we observed that pharmacological or genetic inactivation of SIGMAR1 led to motor neuron axonal degeneration followed by cell death. Disruption of SIGMAR1 function in motor neurons disturbed endoplasmic reticulum-mitochondria contacts, affected intracellular calcium signalling and was accompanied by activation of endoplasmic reticulum stress and defects in mitochondrial dynamics and transport. These defects were not observed in cultured sensory neurons, highlighting the exacerbated sensitivity of motor neurons to SIGMAR1 function. Interestingly, the inhibition of mitochondrial fission was sufficient to induce mitochondria axonal transport defects as well as axonal degeneration similar to the changes observed after SIGMAR1 inactivation or loss. Intracellular calcium scavenging and endoplasmic reticulum stress inhibition were able to restore mitochondrial function and consequently prevent motor neuron degeneration. These results uncover the cellular mechanisms underlying motor neuron degeneration mediated by loss of SIGMAR1 function and provide therapeutically relevant insight into motor neuronal diseases.


Subject(s)
Endoplasmic Reticulum/genetics , Mitochondria/genetics , Motor Neuron Disease/genetics , Receptors, sigma/genetics , Animals , Cells, Cultured , Endoplasmic Reticulum/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Motor Neuron Disease/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Rats , Receptor Cross-Talk , Sigma-1 Receptor
7.
Hum Mol Genet ; 22(20): 4224-32, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23777631

ABSTRACT

Charcot-Marie-Tooth disease (CMT) comprises a clinically and genetically heterogeneous group of peripheral neuropathies characterized by progressive distal muscle weakness and atrophy, foot deformities and distal sensory loss. Following the analysis of two consanguineous families affected by a medium to late-onset recessive form of intermediate CMT, we identified overlapping regions of homozygosity on chromosome 1p36 with a combined maximum LOD score of 5.4. Molecular investigation of the genes from this region allowed identification of two homozygous mutations in PLEKHG5 that produce premature stop codons and are predicted to result in functional null alleles. Analysis of Plekhg5 in the mouse revealed that this gene is expressed in neurons and glial cells of the peripheral nervous system, and that knockout mice display reduced nerve conduction velocities that are comparable with those of affected individuals from both families. Interestingly, a homozygous PLEKHG5 missense mutation was previously reported in a recessive form of severe childhood onset lower motor neuron disease (LMND) leading to loss of the ability to walk and need for respiratory assistance. Together, these observations indicate that different mutations in PLEKHG5 lead to clinically diverse outcomes (intermediate CMT or LMND) affecting the function of neurons and glial cells.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Genes, Recessive , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Adult , Age of Onset , Animals , Child , Chromosomes, Human, Pair 1/genetics , Codon, Nonsense , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Motor Neuron Disease/genetics , Mutation, Missense , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Young Adult
8.
Eur J Neurosci ; 42(2): 1788-96, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25899854

ABSTRACT

In order to identify new regulators of Schwann cell myelination potentially playing a role in peripheral nervous system (PNS) pathologies, we analysed gene expression profiling data from three mouse models of demyelinating neuropathies and from the developing PNS. This analysis revealed that Sox4, which encodes a member of the Sry-related high-mobility group box protein family, was consistently upregulated in all three analysed models of neuropathy. Moreover, Sox4 showed a peak in its expression during development that corresponded with the onset of myelination. To gain further insights into the role of Sox4 in PNS development, we generated a transgenic mouse that specifically overexpresses Sox4 in Schwann cells. Sox4 overexpression led to a temporary delay in PNS myelination without affecting axonal sorting. Importantly, we observed that, whereas Sox4 mRNA could be efficiently overexpressed, Sox4 protein expression in Schwann cells was strictly regulated. Finally, our data showed that enforced expression of Sox4 in the mouse model for Charcot-Marie-Tooth 4C aggravated its neuropathic phenotype. Together, these observations reveal that Sox4 contributes to the regulation of Schwann cell myelination, and also indicates its involvement in the pathophysiology of peripheral neuropathies.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Myelin Sheath/metabolism , Peripheral Nervous System/metabolism , SOXC Transcription Factors/metabolism , Schwann Cells/metabolism , Age Factors , Animals , Animals, Newborn , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Disease Models, Animal , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , RNA, Messenger/genetics , SOXC Transcription Factors/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
9.
Glia ; 62(9): 1502-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24849898

ABSTRACT

Peripheral myelin protein 2 (Pmp2, P2 or Fabp8), a member of the fatty acid binding protein family, was originally described together with myelin basic protein (Mbp or P1) and myelin protein zero (Mpz or P0) as one of the most abundant myelin proteins in the peripheral nervous system (PNS). Although Pmp2 is predominantly expressed in myelinated Schwann cells, its role in glia is currently unknown. To study its function in PNS biology, we have generated a complete Pmp2 knockout mouse (Pmp2(-/-) ). Comprehensive characterization of Pmp2(-/-) mice revealed a temporary reduction in their motor nerve conduction velocity (MNCV). While this change was not accompanied by any defects in general myelin structure, we detected transitory alterations in the myelin lipid profile of Pmp2(-/-) mice. It was previously proposed that Pmp2 and Mbp have comparable functions in the PNS suggesting that the presence of Mbp can partially mask the Pmp2(-/-) phenotype. Indeed, we found that Mbp lacking Shi(-/-) mice, similar to Pmp2(-/-) animals, have preserved myelin structure and reduced MNCV, but this phenotype was not aggravated in Pmp2(-/-) /Shi(-/-) mutants indicating that Pmp2 and Mbp do not substitute each other's functions in the PNS. These data, together with our observation that Pmp2 binds and transports fatty acids to membranes, uncover a role for Pmp2 in lipid homeostasis of myelinating Schwann cells.


Subject(s)
Myelin P2 Protein/metabolism , Schwann Cells/metabolism , Animals , Cell Membrane/metabolism , Fatty Acids/metabolism , Homeostasis/physiology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin P2 Protein/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neural Conduction , Phenotype , RNA, Messenger/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology
10.
Redox Biol ; 73: 103204, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38810421

ABSTRACT

The ELN gene encodes tropoelastin which is used to generate elastic fibers that insure proper tissue elasticity. Decreased amounts of elastic fibers and/or accumulation of bioactive products of their cleavage, named elastokines, are thought to contribute to aging. Cellular senescence, characterized by a stable proliferation arrest and by the senescence-associated secretory phenotype (SASP), increases with aging, fostering the onset and progression of age-related diseases and overall aging, and has so far never been linked with elastin. Here, we identified that decrease in ELN either by siRNA in normal human fibroblasts or by knockout in mouse embryonic fibroblasts results in premature senescence. Surprisingly this effect is independent of elastic fiber degradation or elastokines production, but it relies on the rapid increase in HMOX1 after ELN downregulation. Moreover, the induction of HMOX1 depends on p53 and NRF2 transcription factors, and leads to an increase in iron, further mediating ELN downregulation-induced senescence. Screening of iron-dependent DNA and histones demethylases revealed a role for histone PHF8 demethylase in mediating ELN downregulation-induced senescence. Collectively, these results unveil a role for ELN in protecting cells from cellular senescence through a non-canonical mechanism involving a ROS/HMOX1/iron accumulation/PHF8 histone demethylase pathway reprogramming gene expression towards a senescence program.


Subject(s)
Cellular Senescence , Fibroblasts , Gene Expression Regulation , Heme Oxygenase-1 , Iron , Tropoelastin , Animals , Humans , Mice , Fibroblasts/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , Iron/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Tropoelastin/metabolism , Tropoelastin/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
11.
J Neurosci ; 32(22): 7493-8, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649228

ABSTRACT

Peripheral nerve hyperexcitability (PNH) is one of the distal peripheral neuropathy phenotypes often present in patients affected by type 2 diabetes mellitus (T2DM). Through in vivo and ex vivo electrophysiological recordings in db/db mice, a model of T2DM, we observed that, in addition to reduced nerve conduction velocity, db/db mice also develop PNH. By using pharmacological inhibitors, we demonstrated that the PNH is mediated by the decreased activity of K(v)1-channels. In agreement with these data, we observed that the diabetic condition led to a reduced presence of the K(v)1.2-subunits in juxtaparanodal regions of peripheral nerves in db/db mice and in nerve biopsies from T2DM patients. Together, these observations indicate that the T2DM condition leads to potassium channel-mediated PNH, thus identifying them as a potential drug target to treat some of the DPN related symptoms.


Subject(s)
Kv1.2 Potassium Channel/metabolism , Peripheral Nerves/metabolism , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/pathology , Ranvier's Nodes/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Electric Stimulation , Humans , Male , Mice , Mice, Mutant Strains , Mutation/genetics , Neural Conduction/physiology , Peripheral Nervous System Diseases/etiology , Potassium Channel Blockers/pharmacology , Protein Subunits/metabolism , Receptors, Leptin/genetics , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Tetrodotoxin/pharmacology
12.
Glia ; 61(7): 1041-51, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23553667

ABSTRACT

Mutations in SH3TC2 trigger autosomal recessive demyelinating Charcot-Marie-Tooth type 4C (CMT4C) neuropathy. Sh3tc2 is specifically expressed in Schwann cells and is necessary for proper myelination of peripheral axons. In line with the early onset of neuropathy observed in patients with CMT4C, our analyses of the murine model of CMT4C revealed that the myelinating properties of Sh3tc2-deficient Schwann cells are affected at an early stage. This early phenotype is associated with changes in the canonical Nrg1/ErbB pathway involved in control of myelination. We demonstrated that Sh3tc2 interacts with ErbB2 and plays a role in the regulation of ErbB2 intracellular trafficking from the plasma membrane upon Nrg1 activation. Interestingly, both the loss of Sh3tc2 function in mice and the pathological mutations present in CMT4C patients affect ErbB2 internalization, potentially altering its downstream intracellular signaling pathways. Altogether, our results indicate that the molecular mechanism for the axonal size sensing is disturbed in Sh3tc2-deficient myelinating Schwann cells, thus providing a novel insight into the pathophysiology of CMT4C neuropathy.


Subject(s)
Carrier Proteins/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-2/metabolism , Animals , Animals, Newborn , Carrier Proteins/genetics , Cells, Cultured , Gene Expression Regulation/genetics , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Neuregulin-1/genetics , Receptor, ErbB-2/genetics , Schwann Cells/metabolism , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Subcellular Fractions/metabolism
13.
Mol Biomed ; 4(1): 4, 2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36739330

ABSTRACT

Pancreatic cancer is one of the deadliest cancers owing to its late diagnosis and of the strong resistance to available treatments. Despite a better understanding of the disease in the last two decades, no significant improvement in patient care has been made. Senescent cells are characterized by a stable proliferation arrest and some resistance to cell death. Increasing evidence suggests that multiple lines of antitumor therapy can induce a senescent-like phenotype in cancer cells, which may participate in treatment resistance. In this study, we describe that gemcitabine, a clinically-used drug against pancreatic cancer, induces a senescent-like phenotype in highly chemoresistant pancreatic cancer cells in vitro and in xenografted tumors in vivo. The use of ABT-263, a well-described senolytic compound targeting Bcl2 anti-apoptotic proteins, killed pancreatic gemcitabine-treated senescent-like cancer cells in vitro. In vivo, the combination of gemcitabine and ABT-263 decreased tumor growth, whereas their individual administration had no effect. Together these data highlight the possibility of improving the efficacy of conventional chemotherapies against pancreatic cancer by eliminating senescent-like cancer cells through senolytic intervention. Further studies testing different senolytics or their combination with available treatments will be necessary to optimize preclinical data in mouse models before transferring these findings to clinical trials.

14.
Aging (Albany NY) ; 15(23): 13581-13592, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38095616

ABSTRACT

Smoking is the main risk factor for many lung diseases including chronic obstructive pulmonary disease. Cigarette smoke (CS) contains carcinogenic and reactive oxygen species that favor DNA mutations and perturb the homeostasis and environment of cells. CS induces lung cell senescence resulting in a stable proliferation arrest and a senescence-associated secretory phenotype. It was recently reported that senescent cell accumulation promotes several lung diseases. In this study, we performed a chemical screen, using an FDA-approved drug library, to identify compounds selectively promoting the death of CS-induced senescent lung cells. Aside from the well-known senolytic, ABT-263, we identified other potentially new senescence-eliminating compounds, including a new class of molecules, the dihydropyridine family of calcium voltage-gated channel (CaV) blockers. Among these blockers, Benidipine, decreased senescent lung cells and ameliorates lung emphysema in a mouse model. The dihydropyridine family of CaV blockers thus constitutes a new class of senolytics that could improve lung diseases. Hence, our work paves the way for further studies on the senolytic activity of CaV blockers in different senescence contexts and age-related diseases.


Subject(s)
Cigarette Smoking , Dihydropyridines , Emphysema , Pulmonary Emphysema , Mice , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Cigarette Smoking/adverse effects , Pulmonary Emphysema/genetics , Lung/metabolism , Dihydropyridines/pharmacology , Dihydropyridines/therapeutic use , Dihydropyridines/metabolism , Emphysema/metabolism , Cellular Senescence
15.
J Biol Chem ; 286(30): 26781-93, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21715287

ABSTRACT

The Lpin1 gene encodes the phosphatidate phosphatase (PAP1) enzyme Lipin 1, which plays a critical role in lipid metabolism. In this study we describe the identification and characterization of a rat model with a mutated Lpin1 gene (Lpin1(1Hubr)), generated by N-ethyl-N-nitrosourea mutagenesis. Lpin1(1Hubr) rats are characterized by hindlimb paralysis and mild lipodystrophy that are detectable from the second postnatal week. Sequencing of Lpin1 identified a point mutation in the 5'-end splice site of intron 18 resulting in mis-splicing, a reading frameshift, and a premature stop codon. As this mutation does not induce nonsense-mediated decay, it allows the production of a truncated Lipin 1 protein lacking PAP1 activity. Lpin1(1Hubr) rats developed hypomyelination and mild lipodystrophy rather than the pronounced demyelination and adipocyte defects characteristic of Lpin1(fld/fld) mice, which carry a null allele for Lpin1. Furthermore, biochemical, histological, and molecular analyses revealed that these lesions improve in older Lpin1(1Hubr) rats as compared with young Lpin1(1Hubr) rats and Lpin1(fld/fld) mice. We observed activation of compensatory biochemical pathways substituting for missing PAP1 activity that, in combination with a possible non-enzymatic Lipin 1 function residing outside of its PAP1 domain, may contribute to the less severe phenotypes observed in Lpin1(1Hubr) rats as compared with Lpin1(fld/fld) mice. Although we are cautious in making a direct parallel between the presented rodent model and human disease, our data may provide new insight into the pathogenicity of recently identified human LPIN1 mutations.


Subject(s)
Demyelinating Diseases/enzymology , Introns , Lipodystrophy/enzymology , Mutation , Phosphatidate Phosphatase/metabolism , Alkylating Agents/pharmacology , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Ethylnitrosourea/pharmacology , HEK293 Cells , Humans , Lipodystrophy/genetics , Lipodystrophy/pathology , Mice , Mutagenesis , Pancreatitis-Associated Proteins , Phosphatidate Phosphatase/genetics , Protein Structure, Tertiary , RNA Splice Sites , Rats , Rats, Mutant Strains
16.
Glia ; 60(5): 751-60, 2012 May.
Article in English | MEDLINE | ID: mdl-22337502

ABSTRACT

Both the central and the peripheral nervous systems are prone to multiple age-dependent neurological deficits, often attributed to still unknown alterations in the function of myelinating glia. To uncover the biological processes affected in glial cells by aging, we analyzed gene expression of the Schwann cell-rich mouse sciatic nerve at 17 time points throughout life, from day of birth until senescence. By combining these data with the gene expression data of myelin mouse mutants carrying deletions of either Pmp22, SCAP, or Lpin1, we found that the majority of age-related transcripts were also affected in myelin mutants (54.4%) and were regulated during PNS development (59.5%), indicating a high level of overlap in implicated molecular pathways. The expression profiles in aging copied the direction of transcriptional changes observed in neuropathy models; however, they had the opposite direction when compared with PNS development. The most significantly altered biological processes in aging involved the inflammatory/immune response and lipid metabolism. Interestingly, both these pathways were comparably changed in the aging optic nerve, suggesting that similar biological processes are affected in aging of glia-rich parts of the central and peripheral nervous systems. Our comprehensive comparison of gene expression in three distinct biological conditions including development, aging, and myelin disease thus revealed a previously unanticipated relationship among themselves and identified lipid metabolism and inflammatory/immune response pathways as potential therapeutical targets to prevent or delay so far incurable age-related and inherited forms of neuropathies.


Subject(s)
Cellular Senescence/immunology , Lipid Metabolism/immunology , Nerve Fibers, Myelinated/immunology , Nerve Fibers, Myelinated/metabolism , Neuroglia/immunology , Neuroglia/metabolism , Signal Transduction/immunology , Animals , Animals, Newborn , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/immunology , Neural Pathways/metabolism , Neuroglia/cytology , Sciatic Nerve/immunology , Sciatic Nerve/metabolism
17.
J Neurochem ; 123(5): 662-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22849425

ABSTRACT

Previous clinical observations and data from mouse models with defects in lipid metabolism suggested that epineurial adipocytes may play a role in peripheral nervous system myelination. We have used adipocyte-specific Lpin1 knockout mice to characterize the consequences of the presence of impaired epineurial adipocytes on the myelinating peripheral nerve. Our data revealed that the capacity of Schwann cells to establish myelin, and the functional properties of peripheral nerves, were not affected by compromised epineurial adipocytes in adipocyte-specific Lpin1 knockout mice. To evaluate the possibility that Lpin1-negative adipocytes are still able to support endoneurial Schwann cells, we also characterized sciatic nerves from mice carrying epiblast-specific deletion of peroxisome proliferator-activated receptor gamma, which develop general lipoatrophy. Interestingly, even the complete loss of adipocytes in the epineurium of peroxisome proliferator-activated receptor gamma knockout mice did not lead to detectable defects in Schwann cell myelination. However, probably as a consequence of their hyperglycemia, these mice have reduced nerve conduction velocity, thus mimicking the phenotype observed under diabetic condition. Together, our data indicate that while adipocytes, as regulators of lipid and glucose homeostasis, play a role in nerve function, their presence in epineurium is not essential for establishment or maintenance of proper myelin.


Subject(s)
Adipocytes/metabolism , Myelin Sheath/metabolism , Peripheral Nerves/cytology , Schwann Cells/metabolism , Adipocytes/cytology , Animals , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphatidate Phosphatase/deficiency , Phosphatidate Phosphatase/genetics , Real-Time Polymerase Chain Reaction , Schwann Cells/cytology , Sciatic Nerve/cytology , Sciatic Nerve/metabolism
18.
Proc Natl Acad Sci U S A ; 106(41): 17528-33, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19805030

ABSTRACT

Charcot-Marie-Tooth disease type 4C (CMT4C) is an early-onset, autosomal recessive form of demyelinating neuropathy. The clinical manifestations include progressive scoliosis, delayed age of walking, muscular atrophy, distal weakness, and reduced nerve conduction velocity. The gene mutated in CMT4C disease, SH3TC2/KIAA1985, was recently identified; however, the function of the protein it encodes remains unknown. We have generated knockout mice where the first exon of the Sh3tc2 gene is replaced with an enhanced GFP cassette. The Sh3tc2(DeltaEx1/DeltaEx1) knockout animals develop progressive peripheral neuropathy manifested by decreased motor and sensory nerve conduction velocity and hypomyelination. We show that Sh3tc2 is specifically expressed in Schwann cells and localizes to the plasma membrane and to the perinuclear endocytic recycling compartment, concordant with its possible function in myelination and/or in regions of axoglial interactions. Concomitantly, transcriptional profiling performed on the endoneurial compartment of peripheral nerves isolated from control and Sh3tc2(DeltaEx1/DeltaEx1) animals uncovered changes in transcripts encoding genes involved in myelination and cell adhesion. Finally, detailed analyses of the structures composed of compact and noncompact myelin in the peripheral nerve of Sh3tc2(DeltaEx1/DeltaEx1) animals revealed abnormal organization of the node of Ranvier, a phenotype that we confirmed in CMT4C patient nerve biopsies. The generated Sh3tc2 knockout mice thus present a reliable model of CMT4C neuropathy that was instrumental in establishing a role for Sh3tc2 in myelination and in the integrity of the node of Ranvier, a morphological phenotype that can be used as an additional CMT4C diagnostic marker.


Subject(s)
Proteins/genetics , Animals , Biopsy , Cell Membrane/pathology , Charcot-Marie-Tooth Disease/epidemiology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Exons , Genotype , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mutation , Myelin Sheath/pathology , Prevalence , Promoter Regions, Genetic , Schwann Cells/pathology , Sural Nerve/pathology , src Homology Domains/genetics
19.
Proc Natl Acad Sci U S A ; 106(50): 21383-8, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19948958

ABSTRACT

Myelination requires a massive increase in glial cell membrane synthesis. Here, we demonstrate that the acute phase of myelin lipid synthesis is regulated by sterol regulatory element-binding protein (SREBP) cleavage activation protein (SCAP), an activator of SREBPs. Deletion of SCAP in Schwann cells led to a loss of SREBP-mediated gene expression involving cholesterol and fatty acid synthesis. Schwann cell SCAP mutant mice show congenital hypomyelination and abnormal gait. Interestingly, aging SCAP mutant mice showed partial regain of function; they exhibited improved gait and produced small amounts of myelin indicating a slow SCAP-independent uptake of external lipids. Accordingly, extracellular lipoproteins partially rescued myelination by SCAP mutant Schwann cells. However, SCAP mutant myelin never reached normal thickness and had biophysical abnormalities concordant with abnormal lipid composition. These data demonstrate that SCAP-mediated regulation of glial lipogenesis is key to the proper synthesis of myelin membrane, and provide insight into abnormal Schwann cell function under conditions affecting lipid metabolism.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Lipids/biosynthesis , Membrane Proteins/physiology , Myelin Sheath/metabolism , Sterol Regulatory Element Binding Proteins/physiology , Aging , Animals , Ganglia, Spinal/cytology , Lipid Metabolism , Lipogenesis , Mice , Mice, Mutant Strains , Mutation , Myelin Sheath/chemistry , Neuroglia/metabolism , Recovery of Function , Schwann Cells/metabolism , Schwann Cells/ultrastructure
20.
Cancer Lett ; 546: 215850, 2022 10 10.
Article in English | MEDLINE | ID: mdl-35926818

ABSTRACT

Oncogenic stress-induced senescence initially inhibits tumor initiation by blocking proliferation and by attracting immune cells to clear potentially harmful cells. If these cells are not eliminated they may resume proliferation upon loss-of-tumor suppressors, and be at risk of transformation. During tumor formation, depending on the sequence of events of gain-of-oncogenes and/or loss-of-tumor suppressors, cancer cells may emerge from senescent cells. Here, we show that these transformed cells after senescence (TS) display more aggressive tumorigenic features, with a greater capacity to migrate and a higher resistance to anti-tumoral drugs than cells having undergone transformation without senescence. Bulk transcriptomic analysis and single cell RNA sequencing revealed a signature unique to TS cells. A score of this signature was then generated and a high score was correlated with decreased survival of patients with lung adenocarcinoma, head-neck squamous cell carcinoma, adrenocortical carcinoma, liver hepatocellular carcinoma, skin cutaneous melanoma and low-grade glioma. Together, these findings strongly support that cancer cells arising from senescent cells are more dangerous, and that a molecular signature of these cells may be of prognostic value for some human cancers. It also raises questions about modeling human tumors, using cells or mice, without regards to the sequence of events leading to transformation.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Melanoma , Skin Neoplasms , Animals , Cellular Senescence , Humans , Mice , Phenotype , Tumor Suppressor Protein p53 , Melanoma, Cutaneous Malignant
SELECTION OF CITATIONS
SEARCH DETAIL