Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Immunol ; 25(5): 764-777, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38609546

RESUMEN

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.


Asunto(s)
Síndromes de Inmunodeficiencia , Proteínas del Tejido Nervioso , Ubiquitinas , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Femenino , Masculino , FN-kappa B/metabolismo , Ubiquitina-Proteína Ligasas/genética , Inflamación/inmunología , Inflamación/genética , Linfocitos B/inmunología , Mutación con Pérdida de Función , Fibroblastos/metabolismo , Fibroblastos/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Ratones , Alelos
2.
N Engl J Med ; 388(24): 2241-2252, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37256972

RESUMEN

BACKGROUND: Disabling pansclerotic morphea (DPM) is a rare systemic inflammatory disorder, characterized by poor wound healing, fibrosis, cytopenias, hypogammaglobulinemia, and squamous-cell carcinoma. The cause is unknown, and mortality is high. METHODS: We evaluated four patients from three unrelated families with an autosomal dominant pattern of inheritance of DPM. Genomic sequencing independently identified three heterozygous variants in a specific region of the gene that encodes signal transducer and activator of transcription 4 (STAT4). Primary skin fibroblast and cell-line assays were used to define the functional nature of the genetic defect. We also assayed gene expression using single-cell RNA sequencing of peripheral-blood mononuclear cells to identify inflammatory pathways that may be affected in DPM and that may respond to therapy. RESULTS: Genome sequencing revealed three novel heterozygous missense gain-of-function variants in STAT4. In vitro, primary skin fibroblasts showed enhanced interleukin-6 secretion, with impaired wound healing, contraction of the collagen matrix, and matrix secretion. Inhibition of Janus kinase (JAK)-STAT signaling with ruxolitinib led to improvement in the hyperinflammatory fibroblast phenotype in vitro and resolution of inflammatory markers and clinical symptoms in treated patients, without adverse effects. Single-cell RNA sequencing revealed expression patterns consistent with an immunodysregulatory phenotype that were appropriately modified through JAK inhibition. CONCLUSIONS: Gain-of-function variants in STAT4 caused DPM in the families that we studied. The JAK inhibitor ruxolitinib attenuated the dermatologic and inflammatory phenotype in vitro and in the affected family members. (Funded by the American Academy of Allergy, Asthma, and Immunology Foundation and others.).


Asunto(s)
Enfermedades Autoinmunes , Fármacos Dermatológicos , Quinasas Janus , Esclerodermia Sistémica , Quinasas Janus/antagonistas & inhibidores , Nitrilos , Pirazoles/uso terapéutico , Pirazoles/farmacología , Pirimidinas , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/genética , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/genética , Mutación Missense , Mutación con Ganancia de Función , Fármacos Dermatológicos/uso terapéutico , Antiinflamatorios/uso terapéutico
3.
J Virol ; 97(6): e0049323, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37255439

RESUMEN

Influenza defective interfering (DI) viruses have long been considered promising antiviral candidates because of their ability to interfere with replication-competent viruses and induce antiviral immunity. However, the mechanisms underlying DI-mediated antiviral immunity have not been extensively explored. Here, we demonstrated the interferon (IFN)-independent protection conferred by the influenza DI virus against homologous virus infection in mice deficient in type I and III IFN signaling. We identified unique host signatures responding to DI coinfection by integrating transcriptional and posttranscriptional regulatory data. DI-treated mice exhibited reduced viral transcription, less intense inflammatory and innate immune responses, and primed multiciliated cell differentiation in their lungs at an early stage of infection, even in the absence of type I or III IFNs. This increased multiciliogenesis could also be detected at the protein level via the immunofluorescence staining of lung tissue from DI-treated mice. Overall, our study provides mechanistic insight into the protection mediated by DIs, implying a unifying theme involving inflammation and multiciliogenesis in maintaining respiratory homeostasis and revealing their IFN-independent antiviral activity. IMPORTANCE During replication, the influenza virus generates genetically defective viruses. These are found in natural infections as part of the virus population within the infected host. Some versions of these defective viruses are thought to have protective effects through their interference with replication-competent viruses and induction of antiviral immunity. To better determine the mechanisms underlying the protective effects of these defective interfering (DI) viruses, we tested a DI that we previously identified in vitro with mice. Mice that were infected with a mix of wild-type influenza and DI viruses had less intense inflammatory and innate immune responses than did mice that were infected with the wild-type virus only, even when type I or III interferons, which are cytokines that play a prominent role in defending the respiratory epithelial barrier, were absent. More interestingly, the DI-infected mice had primed multiciliated cell differentiation in their lungs, indicating the potential promotion of epithelial repair by DIs.


Asunto(s)
Diferenciación Celular , Virus Interferentes Defectuosos , Infecciones por Orthomyxoviridae , Animales , Ratones , Interferones , Replicación Viral , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Orthomyxoviridae
4.
Ann Rheum Dis ; 83(6): 787-798, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38408849

RESUMEN

OBJECTIVES: To study the molecular pathogenesis of PAPA (pyogenic arthritis, pyoderma gangrenosum and acne) syndrome, a debilitating hereditary autoinflammatory disease caused by dominant mutation in PSTPIP1. METHODS: Gene knock-out and knock-in mice were generated to develop an animal model. THP1 and retrovirally transduced U937 human myeloid leukaemia cell lines, peripheral blood mononuclear cells, small interfering RNA (siRNA) knock-down, site-directed mutagenesis, cytokine immunoassays, coimmunoprecipitation and immunoblotting were used to study inflammasome activation. Cytokine levels in the skin were evaluated by immunohistochemistry. Responsiveness to Janus kinase (JAK) inhibitors was evaluated ex vivo with peripheral blood mononuclear cells and in vivo in five treatment-refractory PAPA patients. RESULTS: The knock-in mouse model of PAPA did not recapitulate the human disease. In a human myeloid cell line model, PAPA-associated PSTPIP1 mutations activated the pyrin inflammasome, but not the NLRP3, NLRC4 or AIM2 inflammasomes. Pyrin inflammasome activation was independent of the canonical pathway of pyrin serine dephosphorylation and was blocked by the p.W232A PSTPIP1 mutation, which disrupts pyrin-PSTPIP1 interaction. IFN-γ priming of monocytes from PAPA patients led to IL-18 release in a pyrin-dependent manner. IFN-γ was abundant in the inflamed dermis of PAPA patients, but not patients with idiopathic pyoderma gangrenosum. Ex vivo JAK inhibitor treatment attenuated IFN-γ-mediated pyrin induction and IL-18 release. In 5/5 PAPA patients, the addition of JAK inhibitor therapy to IL-1 inhibition was associated with clinical improvement. CONCLUSION: PAPA-associated PSTPIP1 mutations trigger a pyrin-IL-18-IFN-γ positive feedback loop that drives PAPA disease activity and is a target for JAK inhibition.


Asunto(s)
Acné Vulgar , Artritis Infecciosa , Modelos Animales de Enfermedad , Inflamasomas , Interferón gamma , Piodermia Gangrenosa , Piodermia Gangrenosa/genética , Humanos , Animales , Ratones , Acné Vulgar/inmunología , Inflamasomas/metabolismo , Inflamasomas/inmunología , Interferón gamma/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Inhibidores de las Cinasas Janus/farmacología , Ratones Noqueados , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Retroalimentación Fisiológica , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Pirina/genética , Mutación , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Técnicas de Sustitución del Gen , Interleucina-18/metabolismo , Células THP-1
5.
Dev Dyn ; 252(12): 1428-1448, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37435833

RESUMEN

BACKGROUND: Fibroblast growth factor receptor-3 (FGFR3) gain-of-function mutations are linked to achondroplasia. Infigratinib, a FGFR1-3 tyrosine kinase inhibitor, improves skeletal growth in an achondroplasia mouse model. FGFs and their receptors have critical roles in developing teeth, yet effects of infigratinib on tooth development have not been assessed. Dentoalveolar and craniofacial phenotype of Wistar rats dosed with low (0.1 mg/kg) and high (1.0 mg/kg) dose infigratinib were evaluated using micro-computed tomography, histology, and immunohistochemistry. RESULTS: Mandibular third molars were reduced in size and exhibited aberrant crown and root morphology in 100% of female rats and 80% of male rats at high doses. FGFR3 and FGF18 immunolocalization and extracellular matrix protein expression were unaffected, but cathepsin K (CTSK) was altered by infigratinib. Cranial vault bones exhibited alterations in dimension, volume, and density that were more pronounced in females. In both sexes, interfrontal sutures were significantly more patent with high dose vs vehicle. CONCLUSIONS: High dose infigratinib administered to rats during early stages affects dental and craniofacial development. Changes in CTSK from infigratinib in female rats suggest FGFR roles in bone homeostasis. While dental and craniofacial disruptions are not expected at therapeutic doses, our findings confirm the importance of dental monitoring in clinical studies.


Asunto(s)
Acondroplasia , Ratones , Masculino , Ratas , Femenino , Animales , Microtomografía por Rayos X , Ratas Wistar , Receptores de Factores de Crecimiento de Fibroblastos/genética
6.
J Infect Dis ; 223(6): 981-984, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33367731

RESUMEN

Neutrophil extracellular traps (NETs) contribute to immunothrombosis and have been associated with mortality in coronavirus disease 2019 (COVID-19). We stimulated donor neutrophils with plasma from patients with COVID-19 and demonstrated that R406 can abrogate the release of NETs. These data provide evidence for how fostamatinib may mitigate neutrophil-associated mechanisms contributing to COVID-19 immunopathogenesis.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Trampas Extracelulares/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Oxazinas/farmacología , Piridinas/farmacología , Aminopiridinas , COVID-19/sangre , COVID-19/patología , Humanos , Morfolinas , Pirimidinas , SARS-CoV-2/aislamiento & purificación
7.
Hum Mol Genet ; 28(7): 1117-1135, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535187

RESUMEN

In healthy adult skeletal muscle fibers microtubules form a three-dimensional grid-like network. In the mdx mouse, a model of Duchenne muscular dystrophy (DMD), microtubules are mostly disordered, without periodicity. These microtubule defects have been linked to the mdx mouse pathology. We now report that increased expression of the beta 6 class V ß-tubulin (tubb6) contributes to the microtubule changes of mdx muscles. Wild-type muscle fibers overexpressing green fluorescent protein (GFP)-tubb6 (but not GFP-tubb5) have disorganized microtubules whereas mdx muscle fibers depleted of tubb6 (but not of tubb5) normalize their microtubules, suggesting that increasing tubb6 is toxic. However, tubb6 increases spontaneously during differentiation of mouse and human muscle cultures. Furthermore, endogenous tubb6 is not uniformly expressed in mdx muscles but is selectively increased in fiber clusters, which we identify as regenerating. Similarly, mdx-based rescued transgenic mice that retain a higher than expected tubb6 level show focal expression of tubb6 in subsets of fibers. Tubb6 is also upregulated in cardiotoxin-induced mouse muscle regeneration, in human myositis and DMD biopsies, and the tubb6 level correlates with that of embryonic myosin heavy chain, a regeneration marker. In conclusion, modulation of a ß-tubulin isotype plays a role in muscle differentiation and regeneration. Increased tubb6 expression and microtubule reorganization are not pathological per se but reflect a return to an earlier developmental stage. However, chronic elevation of tubb6, as occurs in the mdx mouse, may contribute to the repeated cycles of regeneration and to the pathology of the disease.


Asunto(s)
Músculo Esquelético/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/fisiología , Animales , Distrofina/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Microtúbulos/metabolismo , Microtúbulos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Mioblastos , Cultivo Primario de Células , Regeneración , Activación Transcripcional , Regulación hacia Arriba
8.
Ann Rheum Dis ; 80(2): 209-218, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32988843

RESUMEN

OBJECTIVES: Low-density granulocytes (LDGs) are a distinct subset of proinflammatory and vasculopathic neutrophils expanded in systemic lupus erythematosus (SLE). Neutrophil trafficking and immune function are intimately linked to cellular biophysical properties. This study used proteomic, biomechanical and functional analyses to further define neutrophil heterogeneity in the context of SLE. METHODS: Proteomic/phosphoproteomic analyses were performed in healthy control (HC) normal density neutrophils (NDNs), SLE NDNs and autologous SLE LDGs. The biophysical properties of these neutrophil subsets were analysed by real-time deformability cytometry and lattice light-sheet microscopy. A two-dimensional endothelial flow system and a three-dimensional microfluidic microvasculature mimetic (MMM) were used to decouple the contributions of cell surface mediators and biophysical properties to neutrophil trafficking, respectively. RESULTS: Proteomic and phosphoproteomic differences were detected between HC and SLE neutrophils and between SLE NDNs and LDGs. Increased abundance of type 1 interferon-regulated proteins and differential phosphorylation of proteins associated with cytoskeletal organisation were identified in SLE LDGs relative to SLE NDNs. The cell surface of SLE LDGs was rougher than in SLE and HC NDNs, suggesting membrane perturbances. While SLE LDGs did not display increased binding to endothelial cells in the two-dimensional assay, they were increasingly retained/trapped in the narrow channels of the lung MMM. CONCLUSIONS: Modulation of the neutrophil proteome and distinct changes in biophysical properties are observed alongside differences in neutrophil trafficking. SLE LDGs may be increasingly retained in microvasculature networks, which has important pathogenic implications in the context of lupus organ damage and small vessel vasculopathy.


Asunto(s)
Granulocitos/patología , Lupus Eritematoso Sistémico/inmunología , Proteínas de la Membrana/análisis , Neutrófilos/patología , Proteoma/análisis , Estudios de Casos y Controles , Heterogeneidad Genética , Granulocitos/fisiología , Humanos , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/sangre , Microvasos/metabolismo , Neutrófilos/fisiología , Fosforilación , Proteómica
10.
J Muscle Res Cell Motil ; 36(6): 501-15, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26374336

RESUMEN

The sarcoplasmic reticulum (SR) of striated muscles is specialized for releasing Ca(2+) following sarcolemma depolarization in order to activate muscle contraction. To this end, the SR forms a network of longitudinal tubules and cisternae that surrounds the myofibrils and, at the same time, participates to the assembly of the triadic junctional membrane complexes formed by the close apposition of one t-tubule, originated from the sarcolemma, and two SR terminal cisternae. Advancements in understanding the molecular basis of the SR structural organization have identified an interaction between sAnk1, a transmembrane protein located on the longitudinal SR (l-SR) tubules, and obscurin, a myofibrillar protein. The direct interaction between these two proteins results in molecular contacts that have the overall effect to stabilize the l-SR tubules along myofibrils in skeletal muscle fibers. Less known is the structural organization of the sites in the SR that are specialized for Ca(2+) release and are positioned at the junctional SR (j-SR), i.e. the region of the terminal cisternae that faces the t-tubule at triads. At the j-SR, several trans-membrane proteins like triadin, junctin, or intra-luminal SR proteins like calsequestrin, are assembled together with the ryanodine receptor, the SR Ca(2+) release channel, into a macromolecular complex specialized in releasing Ca(2+). At triads, the 12 nm-wide gap between the t-tubule and the j-SR allows the ryanodine receptor on the j-SR to be functionally coupled with the voltage-gated L-type calcium channel on the t-tubule in order to allow the transduction of the voltage-induced signal into Ca(2+) release through the ryanodine receptor channels. The muscle-specific junctophilin isoforms (JPH1 and JPH2) are anchored to the j-SR with a trans-membrane segment present at the C-terminus and are capable to bind the sarcolemma with a series of phospholipid-binding motifs localized at the N-terminus. Accordingly, through this dual interaction, JPH1 and JPH2 are responsible for the assembly of the triadic junctional membrane complexes. Recent data indicate that junctophilins seem also to interact with other proteins of the excitation-contraction machinery, suggesting that they may contribute to hold excitation-contraction coupling proteins to the sites where the j-SR is being organized.


Asunto(s)
Proteínas Musculares/metabolismo , Retículo Sarcoplasmático/metabolismo , Canales de Calcio Tipo L/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Contracción Muscular/fisiología , Miofibrillas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
11.
Sci Rep ; 14(1): 12293, 2024 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811719

RESUMEN

HLA-B27 is a major risk factor for spondyloarthritis (SpA), yet the underlying mechanisms remain unclear. HLA-B27 misfolding-induced IL-23, which is mediated by endoplasmic reticulum (ER) stress has been hypothesized to drive SpA pathogenesis. Expression of HLA-B27 and human ß2m (hß2m) in rats (HLA-B27-Tg) recapitulates key SpA features including gut inflammation. Here we determined whether deleting the transcription factor CHOP (Ddit3-/-), which mediates ER-stress induced IL-23, affects gut inflammation in HLA-B27-Tg animals. ER stress-mediated Il23a overexpression was abolished in CHOP-deficient macrophages. Although CHOP-deficiency also reduced Il23a expression in immune cells isolated from the colon of B27+ rats, Il17a levels were not affected, and gut inflammation was not reduced. Rather, transcriptome analysis revealed increased expression of pro-inflammatory genes, including Il1a, Ifng and Tnf in HLA-B27-Tg colon tissue in the absence of CHOP, which was accompanied by higher histological Z-scores. RNAScope localized Il17a mRNA to the lamina propria of the HLA-B27-Tg rats and revealed similar co-localization with Cd3e (CD3) in the presence and absence of CHOP. This demonstrates that CHOP-deficiency does not improve, but rather exacerbates gut inflammation in HLA-B27-Tg rats, indicating that HLA-B27 is not promoting gut disease through ER stress-induced IL-23. Hence, CHOP may protect rats from more severe HLA-B27-induced gut inflammation.


Asunto(s)
Colitis , Estrés del Retículo Endoplásmico , Antígeno HLA-B27 , Espondiloartritis , Factor de Transcripción CHOP , Animales , Antígeno HLA-B27/genética , Antígeno HLA-B27/metabolismo , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Colitis/metabolismo , Colitis/genética , Colitis/inducido químicamente , Colitis/patología , Ratas , Espondiloartritis/metabolismo , Espondiloartritis/patología , Espondiloartritis/genética , Modelos Animales de Enfermedad , Interleucina-23/metabolismo , Interleucina-23/genética , Humanos , Subunidad p19 de la Interleucina-23/genética , Subunidad p19 de la Interleucina-23/metabolismo , Ratas Transgénicas , Interleucina-17/metabolismo , Interleucina-17/genética , Colon/patología , Colon/metabolismo , Macrófagos/metabolismo , Macrófagos/inmunología
12.
JCI Insight ; 8(16)2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37463048

RESUMEN

Gene therapy is under advanced clinical development for several lysosomal storage disorders. Pompe disease, a debilitating neuromuscular illness affecting infants, children, and adults with different severity, is caused by a deficiency of lysosomal glycogen-degrading enzyme acid α-glucosidase (GAA). Here, we demonstrated that adeno-associated virus-mediated (AAV-mediated) systemic gene transfer reversed glycogen storage in all key therapeutic targets - skeletal and cardiac muscles, the diaphragm, and the central nervous system - in both young and severely affected old Gaa-knockout mice. Furthermore, the therapy reversed secondary cellular abnormalities in skeletal muscle, such as those in autophagy and mTORC1/AMPK signaling. We used an AAV9 vector encoding a chimeric human GAA protein with enhanced uptake and secretion to facilitate efficient spread of the expressed protein among multiple target tissues. These results lay the groundwork for a future clinical development strategy in Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , alfa-Glucosidasas , Niño , Ratones , Humanos , Animales , alfa-Glucosidasas/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética , Ratones Noqueados , Glucógeno/metabolismo
13.
Structure ; 31(10): 1233-1246.e5, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37572662

RESUMEN

HIV-1 Rev is an essential regulatory protein that transports unspliced and partially spliced viral mRNAs from the nucleus to the cytoplasm for the expression of viral structural proteins. During its nucleocytoplasmic shuttling, Rev interacts with several host proteins to use the cellular machinery for the advantage of the virus. Here, we report the 3.5 Å cryo-EM structure of a 4.8 MDa Rev-tubulin ring complex. Our structure shows that Rev's arginine-rich motif (ARM) binds to both the acidic surfaces and the C-terminal tails of α/ß-tubulin. The Rev-tubulin interaction is functionally homologous to that of kinesin-13, potently destabilizing microtubules at sub-stoichiometric levels. Expression of Rev in astrocytes and HeLa cells shows that it can modulate the microtubule cytoskeleton within the cellular environment. These results show a previously undefined regulatory role of Rev.


Asunto(s)
VIH-1 , Humanos , Células HeLa , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , VIH-1/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Tubulina (Proteína)/metabolismo
14.
Dev Cell ; 58(12): 1052-1070.e10, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37105173

RESUMEN

Organismal homeostasis and regeneration are predicated on committed stem cells that can reside for long periods in a mitotically dormant but reversible cell-cycle arrest state defined as quiescence. Premature escape from quiescence is detrimental, as it results in stem cell depletion, with consequent defective tissue homeostasis and regeneration. Here, we report that Polycomb Ezh1 confers quiescence to murine muscle stem cells (MuSCs) through a non-canonical function. In the absence of Ezh1, MuSCs spontaneously exit quiescence. Following repeated injuries, the MuSC pool is progressively depleted, resulting in failure to sustain proper muscle regeneration. Rather than regulating repressive histone H3K27 methylation, Ezh1 maintains gene expression of the Notch signaling pathway in MuSCs. Selective genetic reconstitution of the Notch signaling corrects stem cell number and re-establishes quiescence of Ezh1-/- MuSCs.


Asunto(s)
Transducción de Señal , Células Madre , Ratones , Animales , División Celular , Puntos de Control del Ciclo Celular , Músculos
15.
J Clin Med ; 11(18)2022 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-36142969

RESUMEN

Italy was the first Western country to face a massive SARS-CoV-2 outbreak. The limited information initially available on the natural course of the disease required caution in the discharge of patients accessing health facilities. This resulted in overcrowded health facilities and emergency services. In this context, improvements of other forms of hospital care assistance were needed. This study shows the results of the first Italian remote monitoring program for COVID-19 patients. The program was implemented by the Azienda Socio Sanitaria (ASST) Lodi (Italy) by using the innovative Zcare software®. Data generated during patient recruitment, monitoring, and discharge were extracted from the Zcare software and statistically analysed. Data refer to a sample of 1196 patients enrolled in the remote monitoring program in 2020. Patients reported symptoms mainly during the first week. The most frequently reported symptoms were general fatigue, cough, and loss of taste and smell (dysosmia). More than 80% of patients reported a saturation level below 96% at least once, and more than 70% had a temperature above 37 °C. Active monitoring of reported symptoms provided valuable insights into the disease's natural history during its less severe acute phase. Only 109 individuals visited the emergency department at least once in the first 100 days of monitoring. Of these, 101 had COVID-19 infection, 69 of whom were hospitalized following a first clinical assessment at the emergency department. The ASST Lodi's telemedicine strategy for COVID patients appears to be a viable alternative to hospitalization. This strategy enables the provision of proper care while making resources available for more critically ill patients, and enhances the availability of resources available for more critically ill patients.

16.
Front Cell Dev Biol ; 7: 176, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31620435

RESUMEN

Differentiated mammalian cells and tissues, such as skeletal muscle fibers, acquire an organization of Golgi complex and microtubules profoundly different from that in proliferating cells and still poorly understood. In adult rodent skeletal muscle, the multinucleated muscle fibers have hundreds of Golgi elements (GE), small stacks of cisternae that serve as microtubule-organizing centers. We are interested in the role of the GE in organizing a peculiar grid of microtubules located in the fiber cortex, against the sarcolemma. Modifications of this grid in the mdx mouse model of Duchenne muscular dystrophy have led to identifying dystrophin, the protein missing in both human disease and mouse model, as a microtubule guide. Compared to wild-type (WT), mdx microtubules are disordered and more dense and they have been linked to the dystrophic pathology. GE themselves are disordered in mdx. Here, to identify the causes of GE and microtubule alterations in the mdx muscle, we follow GFP-tagged microtubule markers in live mdx fibers and investigate the recovery of GE and microtubules after treatment with nocodazole. We find that mdx microtubules grow 10% faster but in 30% shorter bouts and that they begin to form a tangled network, rather than an orthogonal grid, right after nucleation from GE. Strikingly, a large fraction of microtubules in mdx muscle fibers seem to dissociate from GE after nucleation. Moreover, we report that mdx GE are mispositioned and increased in number and size. These results were replicated in WT fibers overexpressing the beta-tubulin tubb6, which is elevated in Duchenne muscular dystrophy, in mdx and in regenerating muscle. Finally, we examine the association of GE with ER exit sites and ER-to-Golgi intermediate compartment, which starts during muscle differentiation, and find it persisting in mdx and tubb6 overexpressing fibers. We conclude that GE are full, small, Golgi complexes anchored, and positioned through ER Exit Sites. We propose a model in which GE mispositioning, together with the absence of microtubule guidance due to the lack of dystrophin, determines the differences in GE and microtubule organization between WT and mdx muscle fibers.

18.
Biochim Biophys Acta ; 1773(12): 1774-85, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17996313

RESUMEN

Alpha-enolase is a key glycolytic enzyme that plays a functional role in several physiological processes depending on the cellular localization. The enzyme is mainly localized in the cytoplasm whereas an alternative translated form, named MBP-1, is predominantly nuclear. The MBP-1 protein has been characterized as a c-Myc promoter binding protein that negatively controls transcription. In the present study, we identified the kelch protein NS1-BP as one of the alpha-enolase/MBP-1 partners by using a yeast two-hybrid screening. Although NS1-BP has been originally described as a protein mainly localized in the nucleus, we provide evidence that NS1-BP also interacts with actin in human cells, as reported for most kelch-containing proteins. Here we showed that alpha-enolase and MBP-1 associate with NS1-BP in vitro and in vivo by GST pull-down assays and coimmunoprecipitation experiments; subsequent immunofluorescent staining confirmed colocalization of the proteins within the cells. Furthermore, functional analyses performed by cotransfection assays revealed that NS1-BP enhances the inhibitory effect exerted by MBP-1 on c-Myc promoter. In mammalian cells, the overexpression of both proteins resulted in an increased repression of basal c-Myc transcription and consistently affected the steady state levels of endogenous c-Myc mRNA. These findings further support the distinct roles of alpha-enolase and its MBP-1 variant in maintaining cell homeostasis. Moreover, our data suggest a novel function for NS1-BP in the control of cell proliferation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Actinas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/aislamiento & purificación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Proteínas de Unión al ARN , Fracciones Subcelulares , Factores de Transcripción/química , Factores de Transcripción/aislamiento & purificación
19.
BMC Ecol ; 7: 6, 2007 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-17663776

RESUMEN

BACKGROUND: The endemic seagrass Posidonia oceanica (L.) Delile colonizes soft bottoms producing highly productive meadows that play a crucial role in coastal ecosystems dynamics. Human activities and natural events are responsible for a widespread meadows regression; to date the identification of "diagnostic" tools to monitor conservation status is a critical issue. In this study the feasibility of a novel tool to evaluate ecological impacts on Posidonia meadows has been tested. Quantification of a putative stress indicator, i.e. phenols content, has been coupled to 2-D electrophoretic protein analysis of rhizome samples. RESULTS: The overall expression pattern from Posidonia rhizome was determined using a preliminary proteomic approach, 437 protein spots were characterized by pI and molecular weight. We found that protein expression differs in samples belonging to sites with high or low phenols: 22 unique protein spots are peculiar of "low phenols" and 27 other spots characterize "high phenols" samples. CONCLUSION: Posidonia showed phenols variations within the meadow, that probably reflect the heterogeneity of environmental pressures. In addition, comparison of the 2-D electrophoresis patterns allowed to highlight qualitative protein expression differences in response to these pressures. These differences may account for changes in metabolic/physiological pathways as adaptation to stress. A combined approach, based on phenols content determination and 2-D electrophoresis protein pattern, seems a promising tool to monitor Posidonia meadows health state.


Asunto(s)
Alismatales/crecimiento & desarrollo , Monitoreo del Ambiente , Fenoles/análisis , Proteínas de Plantas/biosíntesis , Alismatales/química , Electroforesis en Gel Bidimensional , Proteómica , Rizoma/química , Rizoma/crecimiento & desarrollo
20.
Expert Opin Ther Targets ; 21(9): 897-910, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28756711

RESUMEN

INTRODUCTION: Obscurin, a giant protein of striated muscles, is emerging as an important player in a wide range of processes including myofibril assembly and maintenance, muscle protein degradation and intracellular signaling. Accordingly, obscurin participates to the mechanisms by which muscles adapt to physiological requirements or to pathological cues associated with cardiac and skeletal muscle diseases. Areas covered: The structure of the different obscurin isoforms identified so far, their tissue distribution and the most recent findings on obscurin in invertebrates and mammals will be reviewed. We will provide a synopsis of known molecular interactions between obscurin and other proteins and the biological relevance of these interactions for striated muscle function. The involvement of obscurin in protein degradation mechanisms and intracellular signaling will be also discussed along with initial evidence of a role of obscurin in the pathophysiology of human diseases. Expert opinion: Although still much remains to be discovered about the role of obscurin either as a structural component of the sarcomere or as a mediator of signaling pathways within muscle cells, it can be envisioned that this protein represents an interesting novel pharmacological target for the prevention and treatment of cardiac and skeletal muscle diseases.


Asunto(s)
Diseño de Fármacos , Enfermedades Musculares/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Animales , Humanos , Terapia Molecular Dirigida , Músculo Esquelético/patología , Enfermedades Musculares/fisiopatología , Miocardio/patología , Proteínas Serina-Treonina Quinasas , Sarcómeros/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda