Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 39(13): 111001, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35767949

ABSTRACT

TDP-43 mediates proper Stathmin-2 (STMN2) mRNA splicing, and STMN2 protein is reduced in the spinal cord of most patients with amyotrophic lateral sclerosis (ALS). To test the hypothesis that STMN2 loss contributes to ALS pathogenesis, we generated constitutive and conditional STMN2 knockout mice. Constitutive STMN2 loss results in early-onset sensory and motor neuropathy featuring impaired motor behavior and dramatic distal neuromuscular junction (NMJ) denervation of fast-fatigable motor units, which are selectively vulnerable in ALS, without axon or motoneuron degeneration. Selective excision of STMN2 in motoneurons leads to similar NMJ pathology. STMN2 knockout heterozygous mice, which better model the partial loss of STMN2 protein found in patients with ALS, display a slowly progressive, motor-selective neuropathy with functional deficits and NMJ denervation. Thus, our findings strongly support the hypothesis that STMN2 reduction owing to TDP-43 pathology contributes to ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , DNA-Binding Proteins , Stathmin , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Mice, Knockout , Motor Neurons/metabolism , Motor Neurons/pathology , Stathmin/deficiency , Stathmin/genetics , Stathmin/metabolism
2.
Cancer Res ; 79(2): 397-409, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30478213

ABSTRACT

Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer. SIGNIFICANCE: Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/metabolism , Receptor, ErbB-2/metabolism , Stathmin/metabolism , Animals , Carcinogenesis , Female , HEK293 Cells , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Prolactin/metabolism , Receptor, ErbB-2/genetics , Receptors, Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Stathmin/deficiency , Stathmin/genetics
3.
Cell Cycle ; 13(19): 3100-11, 2014.
Article in English | MEDLINE | ID: mdl-25486569

ABSTRACT

The CDK inhibitor p27(kip1) is a critical regulator of cell cycle progression, but the mechanisms by which p27(kip1) controls cell proliferation in vivo are still not fully elucidated. We recently demonstrated that the microtubule destabilizing protein stathmin is a relevant p27(kip1) binding partner. To get more insights into the in vivo significance of this interaction, we generated p27(kip1) and stathmin double knock-out (DKO) mice. Interestingly, thorough characterization of DKO mice demonstrated that most of the phenotypes of p27(kip1) null mice linked to the hyper-proliferative behavior, such as the increased body and organ weight, the outgrowth of the retina basal layer and the development of pituitary adenomas, were reverted by co-ablation of stathmin. In vivo analyses showed a reduced proliferation rate in DKO compared to p27(kip1) null mice, linked, at molecular level, to decreased kinase activity of CDK4/6, rather than of CDK1 and CDK2. Gene expression profiling of mouse thymuses confirmed the phenotypes observed in vivo, showing that DKO clustered with WT more than with p27 knock-out tissue. Taken together, our results demonstrate that stathmin cooperates with p27(kip1) to control the early phase of G1 to S phase transition and that this function may be of particular relevance in the context of tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Stathmin/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Female , G1 Phase , Gene Expression Profiling , Gigantism/metabolism , Gigantism/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pituitary Gland/metabolism , Pituitary Gland/pathology , S Phase , Stathmin/deficiency , Thymus Gland/metabolism , Thymus Gland/pathology
4.
PLoS One ; 9(10): e109750, 2014.
Article in English | MEDLINE | ID: mdl-25285524

ABSTRACT

Stathmin 1 performs a critical function in cell proliferation by regulating microtubule polymerization. This proliferative function is thought to explain the frequent overexpression of stathmin in human cancer and its correlation with a bad prognosis. Whether stathmin also functions in cell death pathways is unclear. Stathmin regulates microtubules in part by binding free tubulin, a process inhibited by stathmin phosphorylation from kinases including c-Jun N-terminal kinase (JNK). The involvement of JNK activation both in stathmin phosphorylation, and in hepatocellular resistance to oxidative stress, led to an examination of the role of stathmin/JNK crosstalk in oxidant-induced hepatocyte death. Oxidative stress from menadione-generated superoxide induced JNK-dependent stathmin phosphorylation at Ser-16, Ser-25 and Ser-38 in hepatocytes. A stathmin knockdown sensitized hepatocytes to both apoptotic and necrotic cell death from menadione without altering levels of oxidant generation. The absence of stathmin during oxidative stress led to JNK overactivation that was the mechanism of cell death as a concomitant knockdown of JNK1 or JNK2 blocked death. Hepatocyte death from JNK overactivation was mediated by the effects of JNK on mitochondria. Mitochondrial outer membrane permeabilization occurred in stathmin knockdown cells at low concentrations of menadione that triggered apoptosis, whereas mitochondrial ß-oxidation and ATP homeostasis were compromised at higher, necrotic menadione concentrations. Stathmin therefore mediates hepatocyte resistance to death from oxidative stress by down regulating JNK and maintaining mitochondrial integrity. These findings demonstrate a new mechanism by which stathmin promotes cell survival and potentially tumor growth.


Subject(s)
Down-Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Oxidative Stress , Stathmin/metabolism , Animals , Apoptosis/drug effects , Cell Line , Down-Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Hepatocytes/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Necrosis/chemically induced , Oxidative Stress/drug effects , Rats , Signal Transduction/drug effects , Stathmin/deficiency , Stathmin/genetics , Vitamin K 3/pharmacology
5.
Nat Commun ; 5: 4389, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25007915

ABSTRACT

Changes in the stability of microtubules regulate many biological processes, but their role in memory remains unclear. Here we show that learning causes biphasic changes in the microtubule-associated network in the hippocampus. In the early phase, stathmin is dephosphorylated, enhancing its microtubule-destabilizing activity by promoting stathmin-tubulin binding, whereas in the late phase these processes are reversed leading to an increase in microtubule/KIF5-mediated localization of the GluA2 subunit of AMPA receptors at synaptic sites. A microtubule stabilizer paclitaxel decreases or increases memory when applied at the early or late phases, respectively. Stathmin mutations disrupt changes in microtubule stability, GluA2 localization, synaptic plasticity and memory. Aged wild-type mice show impairments in stathmin levels, changes in microtubule stability and GluA2 localization. Blocking GluA2 endocytosis rescues memory deficits in stathmin mutant and aged wild-type mice. These findings demonstrate a role for microtubules in memory in young adult and aged individuals.


Subject(s)
Aging/physiology , Learning/physiology , Memory Disorders/physiopathology , Memory/physiology , Microtubules/physiology , Stathmin/physiology , Animals , Hippocampus/physiology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Microtubule Proteins/physiology , Mutation/genetics , Neuronal Plasticity/physiology , Receptors, AMPA/physiology , Signal Transduction/physiology , Stathmin/deficiency , Stathmin/genetics , Tubulin/physiology
6.
PLoS One ; 9(2): e90141, 2014.
Article in English | MEDLINE | ID: mdl-24587245

ABSTRACT

Stathmin is a prognostic marker in many cancers, including endometrial cancer. Preclinical studies, predominantly in breast cancer, have suggested that stathmin may additionally be a predictive marker for response to paclitaxel. We first evaluated the response to paclitaxel in endometrial cancer cell lines before and after stathmin knock-down. Subsequently we investigated the clinical response to paclitaxel containing chemotherapy in metastatic endometrial cancer in relation to stathmin protein level in tumors. Stathmin level was also determined in metastatic lesions, analyzing changes in biomarker status on disease progression. Knock-down of stathmin improved sensitivity to paclitaxel in endometrial carcinoma cell lines with both naturally higher and lower sensitivity to paclitaxel. In clinical samples, high stathmin level was demonstrated to be associated with poor response to paclitaxel containing chemotherapy and to reduced disease specific survival only in patients treated with such combination. Stathmin level increased significantly from primary to metastatic lesions. This study suggests, supported by both preclinical and clinical data, that stathmin could be a predictive biomarker for response to paclitaxel treatment in endometrial cancer. Re-assessment of stathmin level in metastatic lesions prior to treatment start may be relevant. Also, validation in a randomized clinical trial will be important.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Bridged-Ring Compounds/pharmacology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Stathmin/metabolism , Taxoids/pharmacology , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Bridged-Ring Compounds/therapeutic use , Cell Line, Tumor , Endometrial Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Neoplasm Metastasis , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Stathmin/deficiency , Stathmin/genetics , Taxoids/therapeutic use , Treatment Outcome
7.
J Immunol ; 188(11): 5421-7, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529300

ABSTRACT

Polarization of T cells involves reorientation of the microtubule organizing center (MTOC). Because activated ERK is localized at the immunological synapse, we investigated its role by showing that ERK activation is important for MTOC polarization. Suspecting that ERK phosphorylates a regulator of microtubules, we next focused on stathmin, a known ERK substrate. Our work indicates that during T cell activation, ERK is recruited to the synapse, allowing it to phosphorylate stathmin molecules near the immunological synapse. Supporting an important role of stathmin phosphorylation in T cell activation, we showed that T cell activation results in increased microtubule growth rate dependent on the presence of stathmin. The significance of this finding was demonstrated by results showing that CTLs from stathmin(-/-) mice displayed defective MTOC polarization and defective target cell cytolysis. These data implicate stathmin as a regulator of the microtubule network during T cell activation.


Subject(s)
Cell Polarity/immunology , Lymphocyte Activation/immunology , Microtubule-Organizing Center/metabolism , Stathmin/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Enlargement , Cells, Cultured , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Organizing Center/immunology , Phosphorylation/immunology , Stathmin/deficiency , Stathmin/metabolism , T-Lymphocyte Subsets/cytology
8.
PLoS One ; 7(2): e30942, 2012.
Article in English | MEDLINE | ID: mdl-22312434

ABSTRACT

Extinction is an integral part of normal healthy fear responses, while it is compromised in several fear-related mental conditions in humans, such as post-traumatic stress disorder (PTSD). Although much research has recently been focused on fear extinction, its molecular and cellular underpinnings are still unclear. The development of animal models for extinction will greatly enhance our approaches to studying its neural circuits and the mechanisms involved. Here, we describe two gene-knockout mouse lines, one with impaired and another with enhanced extinction of learned fear. These mutant mice are based on fear memory-related genes, stathmin and gastrin-releasing peptide receptor (GRPR). Remarkably, both mutant lines showed changes in fear extinction to the cue but not to the context. We performed indirect imaging of neuronal activity on the second day of cued extinction, using immediate-early gene c-Fos. GRPR knockout mice extinguished slower (impaired extinction) than wildtype mice, which was accompanied by an increase in c-Fos activity in the basolateral amygdala and a decrease in the prefrontal cortex. By contrast, stathmin knockout mice extinguished faster (enhanced extinction) and showed a decrease in c-Fos activity in the basolateral amygdala and an increase in the prefrontal cortex. At the same time, c-Fos activity in the dentate gyrus was increased in both mutant lines. These experiments provide genetic evidence that the balance between neuronal activities of the amygdala and prefrontal cortex defines an impairment or facilitation of extinction to the cue while the hippocampus is involved in the context-specificity of extinction.


Subject(s)
Amygdala/physiology , Cues , Extinction, Psychological/physiology , Fear/psychology , Prefrontal Cortex/physiology , Receptors, Bombesin/metabolism , Stathmin/metabolism , Amygdala/cytology , Amygdala/metabolism , Animals , Conditioning, Psychological/physiology , Fear/physiology , Gene Knockout Techniques , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Receptors, Bombesin/deficiency , Receptors, Bombesin/genetics , Stathmin/deficiency , Stathmin/genetics
9.
J Bone Miner Res ; 26(9): 2052-67, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21557310

ABSTRACT

Cytoskeleton microtubules regulate various cell signaling pathways that are involved in bone cell function. We recently reported that inhibition of microtubule assembly by microtubule-targeting drugs stimulates osteoblast differentiation and bone formation. To further elucidate the role of microtubules in bone homeostasis, we characterized the skeletal phenotype of mice null for stathmin, an endogenous protein that inhibits microtubule assembly. In vivo micro-computed tomography (µCT) and histology revealed that stathmin deficiency results in a significant reduction of bone mass in adult mice concurrent with decreased osteoblast and increased osteoclast numbers in bone tissues. Phenotypic analyses of primary calvarial cells and bone marrow cells showed that stathmin deficiency inhibited osteoblast differentiation and induced osteoclast formation. In vitro overexpression studies showed that increased stathmin levels enhanced osteogenic differentiation of preosteoblast MC3T3-E1 cells and mouse bone marrow-derived cells and attenuated osteoclast formation from osteoclast precursor Raw264.7 cells and bone marrow cells. Results of immunofluorescent studies indicated that overexpression of stathmin disrupted radial microtubule filaments, whereas deficiency of stathmin stabilized the microtubule network structure in these bone cells. In addition, microtubule-targeting drugs that inhibit microtubule assembly and induce osteoblast differentiation lost these effects in the absence of stathmin. Collectively, these results suggest that stathmin, which alters microtubule dynamics, plays an essential role in maintenance of postnatal bone mass by regulating both osteoblast and osteoclast functions in bone. \


Subject(s)
Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone and Bones/pathology , Microtubules/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Stathmin/deficiency , Animals , Bone Diseases, Metabolic/diagnostic imaging , Bone Morphogenetic Protein 2/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Cell Differentiation , Kruppel-Like Transcription Factors/metabolism , Mice , Organ Size , Osteoblasts/pathology , Osteoclasts/pathology , Phenotype , Stathmin/metabolism , X-Ray Microtomography , Zinc Finger Protein Gli2
10.
Mol Cell Biol ; 30(9): 2229-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20194624

ABSTRACT

p27(kip1) (p27) is an inhibitor of cyclin/cyclin-dependent kinase complexes, whose nuclear loss indicates a poor prognosis in various solid tumors. When located in the cytoplasm, p27 binds Op18/stathmin (stathmin), a microtubule (MT)-destabilizing protein, and restrains its activity. This leads to MT stabilization, which negatively affects cell migration. Here, we demonstrate that this p27 function also influences morphology and motility of cells immersed in three-dimensional (3D)matrices. Cells lacking p27 display a decrease in MT stability, a rounded shape when immersed in 3D environments, and a mesenchymal-amoeboid conversion in their motility mode. Upon cell contact to extracellular matrix, the decreased MT stability observed in p27 null cells results in accelerated lipid raft trafficking and increased RhoA activity. Importantly, cell morphology, motility, MT network composition, and distribution of p27 null cells were rescued by the concomitant genetic ablation of Stathmin, implicating that the balanced expression of p27 and stathmin represents a crucial determinant for cytoskeletal organization and cellular behavior in 3D contexts.


Subject(s)
Cell Movement , Cell Shape , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endocytosis , Membrane Microdomains/metabolism , Microtubules/metabolism , 3T3 Cells , Animals , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cytoplasmic Vesicles/metabolism , Embryo, Mammalian/cytology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Stathmin/deficiency , Stathmin/metabolism , rhoA GTP-Binding Protein/metabolism
11.
Am J Physiol Renal Physiol ; 290(6): F1559-67, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16434570

ABSTRACT

In kidneys subjected to ischemic reperfusion injury (IRI) stathmin, a tubulin-binding protein involved in the regulation of mitosis, is expressed in dedifferentiated and proliferating renal tubule cells during the recovery phase. To ascertain the role of stathmin in the recovery from ischemic kidney injury, stathmin-deficient (OP18-/-) and wild-type (WT) animals were subjected to experimental IRI. At 3, 7, and 14 days after reperfusion serum samples and kidneys were collected for the examination of parameters of renal function, morphology, and recovery. Our studies indicate that on day 14 after reperfusion OP18-/- mice have significant renal failure, whereas the creatinine levels of WT animals have returned to baseline. Compared with WT animals OP18-/- mice had more extensive tubular fibrosis. The examination of proliferating cell nuclear antigen expression indicated that OP18-/- animals have increased proliferative or DNA repair activity for a more prolonged duration. The OP18-/- animals also had an increased number of tubules with apoptotic cells. These results suggest that in stathmin-deficient mice subjected to IRI, the aberrant regulation of cell cycle progression, not observed under normal conditions, impairs or at least delays the process of tubular repair and recovery after acute renal injury.


Subject(s)
Kidney/blood supply , Reperfusion Injury/physiopathology , Stathmin/deficiency , Stathmin/physiology , Animals , Apoptosis , Cell Division , DNA Repair , Epithelial Cells/chemistry , Epithelial Cells/pathology , Fibrosis , Inbreeding , Kidney/pathology , Kidney/physiopathology , Kidney Tubules/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proliferating Cell Nuclear Antigen/analysis , Renal Insufficiency/epidemiology , Renal Insufficiency/etiology , Reperfusion Injury/mortality , Reperfusion Injury/pathology , Stathmin/genetics , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...