Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Physiol Genomics ; 52(10): 492-511, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32926651

RESUMO

Skeletal muscles can undergo atrophy and/or programmed cell death (PCD) during development or in response to a wide range of insults, including immobility, cachexia, and spinal cord injury. However, the protracted nature of atrophy and the presence of multiple cell types within the tissue complicate molecular analyses. One model that does not suffer from these limitations is the intersegmental muscle (ISM) of the tobacco hawkmoth Manduca sexta. Three days before the adult eclosion (emergence) at the end of metamorphosis, the ISMs initiate a nonpathological program of atrophy that results in a 40% loss of mass. The ISMs then generate the eclosion behavior and initiate a nonapoptotic PCD during the next 30 h. We have performed a comprehensive transcriptomics analysis of all mRNAs and microRNAs throughout ISM development to better understand the molecular mechanisms that mediate atrophy and death. Atrophy involves enhanced protein catabolism and reduced expression of the genes involved in respiration, adhesion, and the contractile apparatus. In contrast, PCD involves the induction of numerous proteases, DNA methylases, membrane transporters, ribosomes, and anaerobic metabolism. These changes in gene expression are largely repressed when insects are injected with the insect steroid hormone 20-hydroxyecdysone, which delays death. The expression of the death-associated proteins may be greatly enhanced by reductions in specific microRNAs that function to repress translation. This study not only provides fundamental new insights into basic developmental processes, it may also represent a powerful resource for identifying potential diagnostic markers and molecular targets for therapeutic intervention.


Assuntos
Apoptose/genética , Genes de Insetos , Manduca/genética , Atrofia Muscular/genética , Transcriptoma , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas Contráteis/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , MicroRNAs/genética , Contração Muscular/genética , Músculo Esquelético/crescimento & desenvolvimento , RNA Mensageiro/genética
2.
Am J Physiol Cell Physiol ; 314(5): C534-C544, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351404

RESUMO

Peripheral artery disease is an atherosclerotic occlusive disease that causes limb ischemia and has few effective noninterventional treatments. Stem cell therapy is promising, but concomitant diabetes may limit its effectiveness. We evaluated the therapeutic potential of skeletal muscle pericytes to augment postischemic neovascularization in wild-type and type 2 diabetic (T2DM) mice. Wild-type C57BL/6J and leptin receptor spontaneous mutation db/db T2DM mice underwent unilateral femoral artery excision to induce limb ischemia. Twenty-four hours after ischemia induction, CD45-CD34-CD146+ skeletal muscle pericytes or vehicle controls were transplanted into ischemic hindlimb muscles. At postoperative day 28, pericyte transplantation augmented blood flow recovery in wild-type mice (79.3 ± 5% vs. 61.9 ± 5%; P = 0.04), but not in T2DM mice (48.6% vs. 46.3 ± 5%; P = 0.51). Pericyte transplantation augmented collateral artery enlargement in wild-type (26.7 ± 2 µm vs. 22.3 ± 1 µm, P = 0.03), but not T2DM mice (20.4 ± 1.4 µm vs. 18.5 ± 1.2 µm, P = 0.14). Pericyte incorporation into collateral arteries was higher in wild-type than in T2DM mice ( P = 0.002). Unexpectedly, pericytes differentiated into Schwann cells in vivo. In vitro, Insulin increased Nox2 expression and decreased tubular formation capacity in human pericytes. These insulin-induced effects were reversed by N-acetylcysteine antioxidant treatment. In conclusion, T2DM impairs the ability of pericytes to augment neovascularization via decreased collateral artery enlargement and impaired engraftment into collateral arteries, potentially via hyperinsulinemia-induced oxidant stress. While pericytes show promise as a unique form of stem cell therapy to increase postischemic neovascularization, characterizing the molecular mechanisms by which T2DM impairs their function is essential to achieve their therapeutic potential.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Isquemia/cirurgia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Pericitos/transplante , Animais , Diferenciação Celular , Células Cultivadas , Circulação Colateral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Humanos , Insulina/farmacologia , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Pericitos/patologia , Fenótipo , Receptores para Leptina/genética , Fluxo Sanguíneo Regional , Remodelação Vascular
4.
Am J Physiol Cell Physiol ; 311(4): C607-C615, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27558160

RESUMO

Skeletal muscle mass can increase during hypertrophy or decline dramatically in response to normal or pathological signals that trigger atrophy. Many reports have documented that the number of nuclei within these cells is also plastic. It has been proposed that a yet-to-be-defined regulatory mechanism functions to maintain a relatively stable relationship between the cytoplasmic volume and nuclear number within the cell, a phenomenon known as the "myonuclear domain" hypothesis. While it is accepted that hypertrophy is typically associated with the addition of new nuclei to the muscle fiber from stem cells such as satellite cells, the loss of myonuclei during atrophy has been controversial. The intersegmental muscles from the tobacco hawkmoth Manduca sexta are composed of giant syncytial cells that undergo sequential developmental programs of atrophy and programmed cell death at the end of metamorphosis. Since the intersegmental muscles lack satellite cells or regenerative capacity, the tissue is not "contaminated" by these nonmuscle nuclei. Consequently, we monitored muscle mass, cross-sectional area, nuclear number, and cellular DNA content during atrophy and the early phases of cell death. Despite a ∼75-80% decline in muscle mass and cross-sectional area during the period under investigation, there were no reductions in nuclear number or DNA content, and the myonuclear domain was reduced by ∼85%. These data suggest that the myonuclear domain is not an intrinsic property of skeletal muscle and that nuclei persist through atrophy and programmed cell death.


Assuntos
Morte Celular/fisiologia , Núcleo Celular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Atrofia Muscular/fisiopatologia , Animais , Apoptose/fisiologia , Hipertrofia/fisiopatologia , Manduca/fisiologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/fisiologia
5.
Muscle Nerve ; 47(4): 522-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23364895

RESUMO

INTRODUCTION: Skeletal muscle regeneration following damage relies on proliferation and differentiation of muscle precursor cells (MPCs). We recently observed increased NF-kB activity in vascular-associated muscle resident pericytes following muscle damage in humans. We determined how altered NF-kB activity in human primary pericytes (HPPs) affects their myogenic differentiation (cell-autonomous effects), as well as proliferation and differentiation of co-cultured MPCs (non-cell-autonomous effects). METHODS: HPPs were transfected with vectors that increased or decreased NF-kB activity. Transfected HPPs were co-cultured with C2 C12 myoblasts under differentiation conditions, and HPP fusion to myotubes was measured. We also co-cultured HPPs with C2 C12 myoblasts and measured proliferation and myotube formation. RESULTS: Inhibition of NF-kB activity increased HPP fusion to C2 C12 myotubes. Moreover, enhanced NF-kB activity in HPPs suppressed differentiation and enhanced proliferation of co-cultured myoblasts. CONCLUSIONS: NF-kB activity acts cell-autonomously to inhibit HPP myogenic differentiation and non-cell-autonomously to promote MPC proliferation and suppress MPC differentiation in vitro.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Mioblastos/fisiologia , Subunidade p50 de NF-kappa B/fisiologia , Pericitos/fisiologia , Regeneração/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Humanos , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Pericitos/metabolismo
6.
Cancers (Basel) ; 15(17)2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37686533

RESUMO

The mesenchymal subtype of glioblastoma (mGBM), which is characterized by rigorous vasculature, resists anti-tumor immune therapy. Here, we investigated the mechanistic link between tumor vascularization and the evasion of immune surveillance. Clinical datasets with GBM transcripts showed that the expression of the mesenchymal markers YKL-40 (CHI3L1) and Vimentin is correlated with elevated expression of PD-L1 and poor disease survival. Interestingly, the expression of PD-L1 was predominantly found in vascular endothelial cells. Orthotopic transplantation of glioma cells GL261 over-expressing YKL-40 in mice showed increased angiogenesis and decreased CD8+ T cell infiltration, resulting in a reduction in mouse survival. The exposure of recombinant YKL-40 protein induced PD-L1 and VE-cadherin (VE-cad) expression in endothelial cells and drove VE-cad-mediated nuclear translocation of ß-catenin/LEF, where LEF upregulated PD-L1 expression. YKL-40 stimulated the dissociation of VE-cad from PD-L1, rendering PD-L1 available to interact with PD-1 from CD8+-positive TALL-104 lymphocytes and inhibit TALL-104 cytotoxicity. YKL-40 promoted TALL-104 cell migration and adhesion to endothelial cells via CCR5-dependent chemotaxis but blocked its anti-vascular immunity. Knockdown of VE-cad or the PD-L1 gene ablated the effects of YKL-40 and reinvigorated TALL-104 cell immunity against vessels. In summary, our study demonstrates a novel vascular immune escape mechanism by which mGBM promotes tumor vascularization and malignant transformation.

7.
Int J Cancer ; 130(3): 544-54, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21387291

RESUMO

Acheron (Achn) is a new member of the Lupus antigen family of RNA binding proteins. Previous studies have shown that Achn controls developmental decisions in neurons and muscle. In the human mammary gland, Achn expression is restricted to ductal myoepithelial cells. Microarray analysis and immunohistochemistry have shown that Achn expression is elevated in some basal-like ductal carcinomas. To study the possible role of Achn in breast cancer, we engineered human MDA-MB-231 cells to stably express enhanced green fluorescent protein-tagged wild-type Achn (AchnWT), as well as Achn lacking either its nuclear localization signal (AchnNLS) or its nuclear export signal (AchnNES). In in vitro assays, AchnWT and AchnNES, but not AchnNLS, enhanced cell proliferation, lamellipodia formation, and invasive activity and drove expression of the elevated expression of the metastasis-associated proteins MMP-9 and VEGF. To determine if Achn could alter the behavior of human breast cancer cells in vivo, Achn-engineered MDA-MB-231 cells were injected into athymic SCID/Beige mice. AchnWT and AchnNES-expressing tumors displayed enhanced angiogenesis and an approximately 5-fold increase in tumor size relative to either control cells or those expressing AchnNLS. These data suggest that Achn enhances human breast tumor growth and vascularization and that this activity is dependent on nuclear localization.


Assuntos
Autoantígenos/metabolismo , Neoplasias da Mama/etiologia , Ribonucleoproteínas/metabolismo , Animais , Autoantígenos/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Camundongos , Camundongos SCID , Invasividade Neoplásica/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Antígeno SS-B
8.
Nat Cell Biol ; 4(6): 451-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021772

RESUMO

In Drosophila melanogaster, apoptosis is controlled by the integrated actions of the Grim-Reaper (Grim-Rpr) and Drosophila Inhibitor of Apoptosis (DIAP) proteins (reviewed in refs 1 4). The anti-apoptotic DIAPs bind to caspases and inhibit their proteolytic activities. DIAPs also bind to Grim-Rpr proteins, an interaction that promotes caspase activity and the initiation of apoptosis. Using a genetic modifier screen, we identified four enhancers of grim-reaper-induced apoptosis that all regulate ubiquitination processes: uba-1, skpA, fat facets (faf), and morgue. Strikingly, morgue encodes a unique protein that contains both an F box and a ubiquitin E2 conjugase domain that lacks the active site Cys required for ubiquitin linkage. A reduction of morgue activity suppressed grim-reaper-induced cell death in Drosophila. In cultured cells, Morgue induced apoptosis that was suppressed by DIAP1. Targeted morgue expression downregulated DIAP1 levels in Drosophila tissue, and Morgue and Rpr together downregulated DIAP1 levels in cultured cells. Consistent with potential substrate binding functions in an SCF ubiquitin E3 ligase complex, Morgue exhibited F box-dependent association with SkpA and F box-independent association with DIAP1. Morgue may thus have a key function in apoptosis by targeting DIAP1 for ubiquitination and turnover.


Assuntos
Apoptose/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Ligases/química , Neuropeptídeos/metabolismo , Peptídeos/metabolismo , Proteínas Ligases SKP Culina F-Box , Sequência de Aminoácidos , Animais , Células Cultivadas , Drosophila , Proteínas de Drosophila/química , Proteínas do Olho/química , Regulação Enzimológica da Expressão Gênica , Proteínas Inibidoras de Apoptose , Proteínas de Insetos/metabolismo , Ligases/genética , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina
9.
BMC Physiol ; 11: 7, 2011 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-21518451

RESUMO

BACKGROUND: One of the hallmarks of Alzheimer's disease, and several other degenerative disorders such as Inclusion Body Myositis, is the abnormal accumulation of amyloid precursor protein (APP) and its proteolytic amyloid peptides. To better understand the pathological consequences of inappropriate APP expression on developing tissues, we generated transgenic flies that express wild-type human APP in the skeletal muscles, and then performed anatomical, electrophysiological, and behavioral analysis of the adults. RESULTS: We observed that neither muscle development nor animal longevity was compromised in these transgenic animals. However, human APP expressing adults developed age-dependent defects in both climbing and flying. We could advance or retard the onset of symptoms by rearing animals in vials with different surface properties, suggesting that human APP expression-mediated behavioral defects are influenced by muscle activity. Muscles from transgenic animals did not display protein aggregates or structural abnormalities at the light or transmission electron microscopic levels. In agreement with genetic studies performed with developing mammalian myoblasts, we observed that co-expression of the ubiquitin E3 ligase Parkin could ameliorate human APP-induced defects. CONCLUSIONS: These data suggest that: 1) ectopic expression of human APP in fruit flies leads to age- and activity-dependent behavioral defects without overt changes to muscle development or structure; 2) environmental influences can greatly alter the phenotypic consequences of human APP toxicity; and 3) genetic modifiers of APP-induced pathology can be identified and analyzed in this model.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Modelos Animais de Doenças , Drosophila melanogaster/fisiologia , Debilidade Muscular/etiologia , Junção Neuromuscular/fisiopatologia , Envelhecimento , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Geneticamente Modificados , Exercício Físico , Voo Animal , Gânglios dos Invertebrados/fisiopatologia , Vidro , Abrigo para Animais , Humanos , Óperon Lac , Neurônios Motores/fisiologia , Músculos/ultraestrutura , Plásticos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transgenes
10.
Front Cell Dev Biol ; 9: 656370, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33898457

RESUMO

While cell death is a normal and essential component of development and homeostasis, dysregulation of this process underlies most human diseases, including cancer, autoimmunity and neurodegeneration. The best characterized mechanism for cell death is apoptosis, although some cells die by a distinct process known as autophagy-dependent cell death (ADCD). Autophagy is mediated by the formation of double membrane vesicles that contain protein aggregates, damaged organelles like mitochondria, and bulk cytoplasm, which then fuse with lysosomes to degrade and recycle their contents. Autophagy is typically viewed as an adaptive process that allows cells to survive stresses like nutrient deprivation, although increasing evidence suggests that it may also mediate cell death during development and pathogenesis. An aggressive form of autophagy termed autosis has been described in cells following either ischemia/reperfusion injury or in response to autophagy-inducing proteins like Tat-Beclin 1. Despite an extensive literature on autophagic cell death in a variety of contexts, there are still fundamental gaps in our understanding of this process. As examples: Does autophagy directly kill cells and if so how? Is ADCD activated concurrently when cells are triggered to die via apoptosis? And is ADCD essentially a more protracted version of autosis or a distinct pathway? The goal of this mini-review is to summarize the field and to identify some of the major gaps in our knowledge. Understanding the molecular mechanisms that mediate ADCD will not only provide new insights into development, they may facilitate the creation of better tools for both the diagnostics and treatment of disease.

11.
Front Genet ; 12: 775369, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35003216

RESUMO

PIWI-interacting RNAs (piRNAs) are small single-stranded RNAs that can repress transposon expression via epigenetic silencing and transcript degradation. They have been identified predominantly in the ovary and testis, where they serve essential roles in transposon silencing in order to protect the integrity of the genome in the germline. The potential expression of piRNAs in somatic cells has been controversial. In the present study we demonstrate the expression of piRNAs derived from both genic and transposon RNAs in the intersegmental muscles (ISMs) from the tobacco hawkmoth Manduca sexta. These piRNAs are abundantly expressed, ∼27 nt long, map antisense to transposons, are oxidation resistant, exhibit a 5' uridine bias, and amplify via the canonical ping-pong pathway. An RNA-seq analysis demonstrated that 19 piRNA pathway genes are expressed in the ISMs and are developmentally regulated. The abundance of piRNAs does not change when the muscles initiate developmentally-regulated atrophy, but are repressed coincident with the commitment of the muscles undergo programmed cell death at the end of metamorphosis. This change in piRNA expression is correlated with the repression of several retrotransposons and the induction of specific DNA transposons. The developmentally-regulated changes in the expression of piRNAs, piRNA pathway genes, and transposons are all regulated by 20-hydroxyecdysone, the steroid hormone that controls the timing of ISM death. Taken together, these data provide compelling evidence for the existence of piRNA in somatic tissues and suggest that they may play roles in developmental processes such as programmed cell death.

12.
Am J Physiol Cell Physiol ; 298(1): C46-55, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889961

RESUMO

Acheron (Achn) was originally identified as novel gene that is induced when insect muscles become committed to die at the end of metamorphosis. In separate studies, we have demonstrated that Achn acts upstream of MyoD and is required by mammalian myoblasts to either differentiate or undergo apoptosis following loss of growth factors. In the present study we examined the role of Achn in regulating integrin-extracellular matrix interactions that are required for myogenesis. Both control C2C12 myoblasts and those engineered to express ectopic Achn expressed the fibronectin receptor integrin alpha(5)beta(1) in the presence of growth factors and the laminin receptor alpha(7)beta(1) following growth factor withdrawal. Expression of the laminin receptor was blocked in cells expressing either Achn antisense or an Achn deletion mutant that blocks differentiation. Control cells and those expressing ectopic Achn undergo sequential and transient increases in both substrate adhesion and migration before cell fusion. Blockade of Achn expression reduced these effects on laminin but not on fibronectin. Taken together, these data suggest that Achn may influence differentiation in part via its control of cell adhesion dynamics.


Assuntos
Autoantígenos/fisiologia , Integrinas/genética , Mioblastos/fisiologia , Ribonucleoproteínas/fisiologia , Autoantígenos/genética , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular , Citoesqueleto/fisiologia , Primers do DNA , Engenharia Genética/métodos , Homeostase , Humanos , Microscopia de Fluorescência , Mioblastos/citologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas/deficiência , Ribonucleoproteínas/genética , Transfecção , Antígeno SS-B
13.
Front Cell Dev Biol ; 8: 622, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850788

RESUMO

The term programmed cell death (PCD) was coined in 1965 to describe the loss of the intersegmental muscles (ISMs) of moths at the end of metamorphosis. While it was subsequently demonstrated that this hormonally controlled death requires de novo gene expression, the signal transduction pathway that couples hormone action to cell death is largely unknown. Using the ISMs from the tobacco hawkmoth Manduca sexta, we have found that Acheron/LARP6 mRNA is induced ∼1,000-fold on the day the muscles become committed to die. Acheron functions as a survival protein that protects cells until cell death is initiated at eclosion (emergence), at which point it becomes phosphorylated and degraded in response to the peptide Eclosion Hormone (EH). Acheron binds to a novel BH3-only protein that we have named BBH1 (BAD/BNIP3 homology 1). BBH1 accumulates on the day the ISMs become committed to die and is presumably liberated when Acheron is degraded. This is correlated with the release and rapid degradation of cytochrome c and the subsequent demise of the cell. RNAi experiments in the fruit fly Drosophila confirmed that loss of Acheron results in precocious ecdysial muscle death while targeting BBH1 prevents death altogether. Acheron is highly expressed in neurons and muscles in humans and drives metastatic processes in some cancers, suggesting that it may represent a novel survival protein that protects terminally differentiated cells and some cancers from death.

14.
Methods Mol Biol ; 559: 313-32, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19609766

RESUMO

One of the hallmarks of development is that many more cells are produced than are ultimately needed for organogenesis. In the case of striated skeletal muscle, large numbers of myoblasts are generated in the somites and then migrate to take up residence in the limbs and the trunk. A subset of these cells fuses to form multinucleated skeletal muscle fibers, while a second group, known as satellite cells, exits the cell cycle and persists as a pool of lineage-restricted stem cells that can repair damaged muscle. The remaining cells initiate apoptosis and are rapidly lost. Primary myoblasts and established satellite cell lines are powerful tools for dissecting the regulatory events that mediate differentiative decisions and have proven to be important models. As well, muscle diseases represent debilitating and often fatal disorders. This chapter provides a general background for muscle development and then details a variety of assays for monitoring the differentiation and the death of muscle. While some of these methods are specialized to address the phenotypic properties of skeletal muscle, others can be employed with a wide variety of cell types.


Assuntos
Morte Celular , Diferenciação Celular , Mioblastos/citologia , Mioblastos/metabolismo , Animais , Anexina A5 , Apoptose , Caspase 3/análise , Caspase 3/metabolismo , Caspase 7/análise , Caspase 7/metabolismo , Separação Celular/métodos , Células Cultivadas , Camundongos , Mitocôndrias/metabolismo , Músculo Esquelético/citologia
15.
Cell Mol Biol Lett ; 14(2): 273-87, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19096764

RESUMO

Acheron, a Lupus antigen ortholog, was identified as a novel death-associated transcript from the intersegmental muscles of the moth Manduca sexta. Acheron is phylogenetically-conserved and represents a new sub-family of Lupus antigen proteins. Acheron is expressed predominantly in neurons and muscle in vertebrates, and regulates several developmental events including myogenesis, neurogenesis and possibly metastasis. Using Acheron as bait, we performed a yeast two-hybrid screen with a mouse embryo cDNA library and identified CASK-C, a novel CASK/Lin-2 isoform, as an Acheron binding partner. Acheron and CASK-C bind via the C-terminus of Acheron and the CaMKII-like domain of CASK-C. Co-immunoprecipitation assays verify this interaction and demonstrate that Acheron also forms a complex with all members of the Id (inhibitor of differentiation) proteins. Taken together, these data suggest a mechanism by which Acheron may regulate development and pathology.


Assuntos
Autoantígenos/metabolismo , Guanilato Quinases/metabolismo , Proteínas Inibidoras de Diferenciação/metabolismo , Ribonucleoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Guanilato Quinases/química , Humanos , Proteínas Inibidoras de Diferenciação/química , Proteínas de Insetos/metabolismo , Dados de Sequência Molecular , Mariposas/química , Estrutura Terciária de Proteína , Alinhamento de Sequência , Técnicas do Sistema de Duplo-Híbrido , Antígeno SS-B
16.
Cell Mol Biol Lett ; 14(2): 288, 2009 06.
Artigo em Inglês | MEDLINE | ID: mdl-30534160

RESUMO

[This corrects the article DOI: 10.2478/s11658-008-0046-1.].

18.
Front Physiol ; 9: 1887, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30740060

RESUMO

Skeletal muscles are the largest cells in the body and are one of the few syncytial ones. There is a longstanding belief that a given nucleus controls a defined volume of cytoplasm, so when a muscle grows (hypertrophy) or shrinks (atrophy), the number of myonuclei change accordingly. This phenomenon is known as the "myonuclear domain hypothesis." There is a general agreement that hypertrophy is accompanied by the addition of new nuclei from stem cells to help the muscles meet the enhanced synthetic demands of a larger cell. However, there is a considerable controversy regarding the fate of pre-existing nuclei during atrophy. Many researchers have reported that atrophy is accompanied by the dramatic loss of myonuclei via apoptosis. However, since there are many different non-muscle cell populations that reside within the tissue, these experiments cannot easily distinguish true myonuclei from those of neighboring mononuclear cells. Recently, two independent models, one from rodents and the other from insects, have demonstrated that nuclei are not lost from skeletal muscle fibers when they undergo either atrophy or programmed cell death. These and other data argue against the current interpretation of the myonuclear domain hypothesis and suggest that once a nucleus has been acquired by a muscle fiber it persists.

19.
Neoplasia ; 20(2): 182-192, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29274508

RESUMO

The heparin-binding glycoprotein YKL-40 (CHI3L1) is intimately associated with microvascularization in multiple human diseases including cancer and inflammation. However, the heparin-binding domain(s) pertinent to the angiogenic activity have yet been identified. YKL-40 harbors a consensus heparin-binding motif that consists of positively charged arginine (R) and lysine (K) (RRDK; residues 144-147); but they don't bind to heparin. Intriguingly, we identified a separate KR-rich domain (residues 334-345) that does display strong heparin binding affinity. A short synthetic peptide spanning this KR-rich domain successfully competed with YKL-40 and blocked its ability to bind heparin. Three individual point mutations, where alanine (A) substituted for K or R (K337A, K342A, R344A), led to remarkable decreases in heparin-binding ability and angiogenic activity. In addition, a neutralizing anti-YKL-40 antibody that targets these residues and prevents heparin binding impeded angiogenesis in vitro. MDA-MB-231 breast cancer cells engineered to express ectopic K337A, K342A or R344A mutants displayed reduced tumor development and compromised tumor vessel formation in mice relative to control cells expressing wild-type YKL-40. These data reveal that the KR-rich heparin-binding motif is the functional heparin-binding domain of YKL-40. Our findings shed light on novel molecular mechanisms underlying endothelial cell angiogenesis promoted by YKL-40 in a variety of diseases.


Assuntos
Arginina/metabolismo , Neoplasias da Mama/patologia , Proteína 1 Semelhante à Quitinase-3/química , Proteína 1 Semelhante à Quitinase-3/metabolismo , Heparina/metabolismo , Lisina/metabolismo , Neovascularização Patológica/patologia , Animais , Apoptose , Arginina/química , Arginina/genética , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Proteína 1 Semelhante à Quitinase-3/genética , Feminino , Heparina/química , Humanos , Lisina/química , Lisina/genética , Camundongos , Camundongos SCID , Mutação , Neovascularização Patológica/metabolismo , Ligação Proteica , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Curr Biol ; 12(2): 131-5, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11818064

RESUMO

The Drosophila genes reaper, head involution defective (hid), and grim all reside at 75C on chromosome three and encode related proteins that have crucial functions in programmed cell death (reviewed in ). In this report, we describe a novel grim-reaper gene, termed sickle, that resides adjacent to reaper. The sickle gene, like reaper and grim, encodes a small protein which contains an RHG motif and a Trp-block. In wild-type embryos, sickle expression was detected in cells of the developing central nervous system. Unlike reaper, hid, and grim, the sickle gene is not removed by Df(3L)H99, and strong ectopic sickle expression was detected in the nervous system of this cell death mutant. sickle very effectively induced cell death in cultured Spodoptera Sf-9 cells, and this death was antagonized by the caspase inhibitors p35 or DIAP1. Strikingly, unlike the other grim-reaper genes, targeted sickle expression did not induce cell death in the Drosophila eye. However, sickle strongly enhanced the eye cell death induced by expression of either an r/grim chimera or reaper.


Assuntos
Morte Celular/genética , Proteínas de Drosophila/genética , Drosophila/genética , Neuropeptídeos/genética , Peptídeos/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Drosophila/embriologia , Proteínas de Drosophila/química , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA