Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Transl Autoimmun ; 5: 100162, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36097634

RESUMEN

Gasdermin D (GSDMD) and gasdermin E (GSDME) perpetuate inflammation by mediating the release of cytokines such as interleukin-1ß (IL-1ß) and IL-18. However, not only are the actions of GSDMD in colitis still controversial, but its interplay with GSDME in the pathogenesis of this disease has not been investigated. We sought to fill these knowledge gaps using the dextran sodium sulfate (DSS) experimental mouse colitis model. DSS ingestion by wild-type mice caused body weight loss as the result of severe gut inflammation, outcomes that were significantly attenuated in Gsdmd -/- or Gsdme -/- mice and nearly fully prevented in Gsdmd -/- ;Gsdme -/- animals. To assess the translational implications of these findings, we tested the efficacy of the active metabolite of US Food and Drug Administration (FDA)-approved disulfiram, which inhibits GSDMD and GSDME function. The severe DSS-induced gut toxicity was significantly decreased in mice treated with the inhibitor. Collectively, our findings indicate that disruption of the function of both GSDMD and GSDME is necessary to achieve maximal therapeutic effect in colitis.

2.
Elife ; 112022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35244027

RESUMEN

Amino-terminal fragments from proteolytically cleaved gasdermins (GSDMs) form plasma membrane pores that enable the secretion of interleukin-1ß (IL-1ß) and IL-18. Excessive GSDM-mediated pore formation can compromise the integrity of the plasma membrane thereby causing the lytic inflammatory cell death, pyroptosis. We found that GSDMD and GSDME were the only GSDMs that were readily expressed in bone microenvironment. Therefore, we tested the hypothesis that GSDMD and GSDME are implicated in fracture healing owing to their role in the obligatory inflammatory response following injury. We found that bone callus volume and biomechanical properties of injured bones were significantly reduced in mice lacking either GSDM compared with wild-type (WT) mice, indicating that fracture healing was compromised in mutant mice. However, compound loss of GSDMD and GSDME did not exacerbate the outcomes, suggesting shared actions of both GSDMs in fracture healing. Mechanistically, bone injury induced IL-1ß and IL-18 secretion in vivo, a response that was mimicked in vitro by bone debris and ATP, which function as inflammatory danger signals. Importantly, the secretion of these cytokines was attenuated in conditions of GSDMD deficiency. Finally, deletion of IL-1 receptor reproduced the phenotype of Gsdmd or Gsdme deficient mice, implying that inflammatory responses induced by the GSDM-IL-1 axis promote bone healing after fracture.


Asunto(s)
Inflamasomas , Interleucina-18 , Animales , Curación de Fractura , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Proteínas de Unión a Fosfato/genética , Piroptosis/genética
3.
BMC Med Genomics ; 14(1): 264, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753502

RESUMEN

Osteopetrosis is a genetically heterogenous, fatal bone disorder characterized by increased bone density. Globally, various genetic causes are reported for osteopetrosis with all forms of inheritance patterns. A precise molecular diagnosis is necessary for prognosis and for prescribing treatment paradigms in osteopetrosis. Here we report on thirteen individuals diagnosed with infantile malignant osteopetrosis coming from ten unrelated Pakistani families; nine of whom are consanguineous. We performed whole exome sequencing and Sanger sequencing in all families and identified homozygous variants in genes previously reported for autosomal recessive inheritance of osteopetrosis. All the identified variants are expected to affect the stability or length of gene products except one nonsynonymous missense variant. TCIRG1 was found as a candidate causal gene in majority of the families. We report six novel variants; four in TCIRG1 and one each in CLCN7 and OSTM1. Our combined findings will be helpful in molecular diagnosis and genetic counselling of patients with osteopetrosis particularly in populations with high consanguinity.


Asunto(s)
Osteopetrosis/genética , Canales de Cloruro/genética , Femenino , Homocigoto , Humanos , Masculino , Proteínas de la Membrana/genética , Mutación Missense , Pakistán , Linaje , Ubiquitina-Proteína Ligasas/genética , ATPasas de Translocación de Protón Vacuolares/genética , Secuenciación del Exoma
4.
Arthritis Res Ther ; 23(1): 286, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784954

RESUMEN

BACKGROUND: Gasdermin D (GSDMD) is cleaved by several proteases including by caspase-1, a component of intracellular protein complexes called inflammasomes. Caspase-1 also converts pro-interleukin-1ß (pro-IL-1ß) and pro-IL-18 into bioactive IL-1ß and IL-18, respectively. GSDMD amino-terminal fragments form plasma membrane pores, which mediate the secretion of IL-1ß and IL-18 and cause the inflammatory form of cell death pyroptosis. Here, we tested the hypothesis that GSDMD contributes to joint degeneration in the K/BxN serum transfer-induced arthritis (STIA) model in which autoantibodies against glucose-6-phosphate isomerase promote the formation of pathogenic immune complexes on the surface of myeloid cells, which highly express the inflammasomes. The unexpected outcomes with the STIA model prompted us to determine the role of GSDMD in the post-traumatic osteoarthritis (PTOA) model caused by meniscus ligamentous injury (MLI) based on the hypothesis that this pore-forming protein is activated by signals released from damaged joint tissues. METHODS: Gsdmd +/+ and Gsdmd-/- mice were injected with K/BxN mouse serum or subjected to MLI to cause STIA or PTOA, respectively. Paw and ankle swelling and DXA scanning were used to assess the outcomes in the STIA model whereas histopathology and micro-computed tomography (µCT) were utilized to monitor joints in the PTOA model. Murine and human joint tissues were also examined for GSDMD, IL-1ß, and IL-18 expression by qPCR, immunohistochemistry, or immunoblotting. RESULTS: GSDMD levels were higher in serum-inoculated paws compared to PBS-injected paws. Unexpectedly, ablation of GSDMD failed to reduce joint swelling and osteolysis, suggesting that GSDMD was dispensable for the pathogenesis of STIA. GSDMD levels were also higher in MLI compared to sham-operated joints. Importantly, ablation of GSDMD attenuated MLI-associated cartilage degradation (p = 0.0097), synovitis (p = 0.014), subchondral bone sclerosis (p = 0.0006), and subchondral bone plate thickness (p = 0.0174) based on histopathological and µCT analyses. CONCLUSION: GSDMD plays a key role in the pathogenesis of PTOA, but not STIA, suggesting that its actions in experimental arthropathy are tissue context-specific.


Asunto(s)
Complejo Antígeno-Anticuerpo , Artritis , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Unión a Fosfato/genética , Heridas y Lesiones/complicaciones , Animales , Artritis/etiología , Autoanticuerpos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Microtomografía por Rayos X
5.
Sci Immunol ; 6(64): eabj3859, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34678046

RESUMEN

NOD-like receptor (NLR), family pyrin domain containing 3 (NLRP3) assembles a protein complex known as the NLRP3 inflammasome upon sensing certain pathogen products or sterile danger signals. Gain-of-function mutations such as the D301N substitution in NLRP3, which cause its constitutive activation (NLRP3CA) also results in inflammasome assembly. This inflammasome processes pro­interleukin-1 ß (pro­IL-1ß) and pro­IL-18 into bioactive IL-1ß and IL-18, respectively, and cleaves gasdermin D (GSDMD). GSDMD amino-terminal fragments form plasma membrane pores that facilitate the secretion of IL-1ß and IL-18 and lead to the inflammatory cell death pyroptosis. Accordingly, GSDMD inactivation results in negligible spontaneous inflammation in various experimental models such as in Nlrp3CA/+ mice lacking GSDMD (Nlrp3CA/+;Gsdmd−/− mice). Here, we found that Nlrp3CA/+;Gsdmd−/− mice, when challenged with LPS or TNF-α, still secreted IL-1ß and IL-18, indicating inflammasome activation independent of GSDMD. Accordingly, Gsdmd−/− macrophages failed to secrete IL-1ß and undergo pyroptosis when briefly exposed to NLRP3 inflammasome activators but released these cytokines when persistently activated. Sustained NLRP3 inflammasome induced caspase-8/-3 and GSDME cleavage and IL-1ß maturation in vitro in Gsdmd−/− macrophages. Thus, a salvage inflammatory pathway involving caspase-8/-3­GSDME was activated after NLRP3 activation when the canonical NLRP3-GSDMD signaling was blocked. Consistent with genetic data, the active metabolite of FDA-approved disulfiram CuET, which inhibited GSDMD and GSDME cleavage in macrophages, reduced the severe inflammation and tissue damage that occurred in the Nlrp3CA/+ mice. Thus, NLRP3 inflammasome activation overwhelms the protection afforded by GSDMD deficiency, rewiring signaling cascades through mechanisms that include GSDME to propagate inflammation.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteínas de Unión a Fosfato/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Animales , Células Cultivadas , Inflamación/patología , Ratones , Ratones Congénicos , Ratones Noqueados , Ratones Transgénicos , Proteínas de Unión a Fosfato/deficiencia , Proteínas Citotóxicas Formadoras de Poros/deficiencia
6.
FASEB J ; 35(9): e21837, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34383985

RESUMEN

Overwhelming evidence indicates that excessive stimulation of innate immune receptors of the NOD-like receptor (NLR) family causes significant damage to multiple tissues, yet the role of these proteins in bone metabolism is not well known. Here, we studied the interaction between the NLRP3 and NLRC4 inflammasomes in bone homeostasis and disease. We found that loss of NLRP3 or NLRC4 inflammasome attenuated osteoclast differentiation in vitro. At the tissue level, lack of NLRP3, or NLRC4 to a lesser extent, resulted in higher baseline bone mass compared to wild-type (WT) mice, and conferred protection against LPS-induced inflammatory osteolysis. Bone mass accrual in mutant mice correlated with lower serum IL-1ß levels in vivo. Unexpectedly, the phenotype of Nlrp3-deficient mice was reversed upon loss of NLRC4 as bone mass was comparable between WT mice and Nlrp3;Nlrc4 knockout mice. Thus, although bone homeostasis is perturbed to various degrees by the lack of NLRP3 or NLRC4, this tissue appears to function normally upon compound loss of the inflammasomes assembled by these receptors.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Resorción Ósea/metabolismo , Huesos/metabolismo , Proteínas de Unión al Calcio/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Diferenciación Celular/fisiología , Homeostasis/fisiología , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/metabolismo , Osteólisis/metabolismo
7.
Am J Transl Res ; 13(3): 1750-1756, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841698

RESUMEN

OBJECTIVE: To investigate the effects of vitamin D drops on immune function in children with recurrent respiratory tract infections (RRTI). METHODS: The clinical data of 119 children with RRTI in our hospital were retrospectively retrieved, and they were divided into group A (n=59, receiving routine treatment) and group B (n=60, receiving vitamin D drops) based on their treatment modality. The clinical efficacy, symptom disappearance time, immune function index, insulin-like growth factor (IGF-1), 25-hydroxyvitamin D3 [25-(OH)D3], serum y-interferon (INF-y), and the number of episodes of respiratory tract infections were compared between the two groups. RESULTS: The total effective rate of treatment in group B was 96.67%, which was significantly higher than 71.19% in group A (P<0.05). Children in group B had shorter time to disappearance of lung rales, cough, and fever than group A (P<0.05). Group B had higher IgA, IgG, and IgM levels, higher CD4+, CD3+ levels and lower CD8+ levels as well as higher IGF-1, 25-(OH)D3, INF-y levels, and fewer respiratory infections after treatment than group A (P<0.05). CONCLUSION: Vitamin D drops are effective in the treatment of children with RRTI, which is beneficial to the improvement of clinical symptoms and immune function.

8.
PLoS Biol ; 18(8): e3000807, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32760056

RESUMEN

Radiotherapy is a commonly used conditioning regimen for bone marrow transplantation (BMT). Cytotoxicity limits the use of this life-saving therapy, but the underlying mechanisms remain poorly defined. Here, we use the syngeneic mouse BMT model to test the hypothesis that lethal radiation damages tissues, thereby unleashing signals that indiscriminately activate the inflammasome pathways in host and transplanted cells. We find that a clinically relevant high dose of radiation causes severe damage to bones and the spleen through mechanisms involving the NLRP3 and AIM2 inflammasomes but not the NLRC4 inflammasome. Downstream, we demonstrate that gasdermin D (GSDMD), the common effector of the inflammasomes, is also activated by radiation. Remarkably, protection against the injury induced by deadly ionizing radiation occurs only when NLRP3, AIM2, or GSDMD is lost simultaneously in both the donor and host cell compartments. Thus, this study reveals a continuum of the actions of lethal radiation relayed by the inflammasome-GSDMD axis, initially affecting recipient cells and ultimately harming transplanted cells as they grow in the severely injured and toxic environment. This study also suggests that therapeutic targeting of inflammasome-GSDMD signaling has the potential to prevent the collateral effects of intense radiation regimens.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Proteínas de Unión al ADN/genética , Inflamasomas/efectos de la radiación , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Unión a Fosfato/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteínas de Unión al ADN/deficiencia , Femenino , Fémur/citología , Fémur/metabolismo , Regulación de la Expresión Génica , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteínas de Unión a Fosfato/deficiencia , Piroptosis/genética , Piroptosis/efectos de la radiación , Transducción de Señal , Bazo/metabolismo , Bazo/patología , Bazo/efectos de la radiación , Trasplante Isogénico , Irradiación Corporal Total , Rayos X
9.
J Bone Miner Res ; 35(4): 776-788, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31793068

RESUMEN

Induction of nuclear factor of activated T cell cytoplasmic 1 (NFATc1) by macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) is essential for macrophage differentiation into osteoclasts (OCs), but the underlying mechanisms remain unclear. The ability of poly(ADP-ribose) polymerase 1 (PARP1) to poly-ADP-ribosylate NFATc1 in T cells prompted us to investigate the PARP1 and NFATc1 interaction during osteoclastogenesis. However, extensive studies failed to directly link PARP1 to NFATc1. A combination of transcriptomics and proteomics studies was then used to identify PARP1 targets under these conditions. These unbiased approaches in conjunction with site-directed mutagenesis studies revealed that PARP1 inhibited NFATc1 expression and OC formation by ADP-ribosylating histone H2B at serine 7 and decreasing the occupancy of this histone variant at the NFATc1 promoter. The anti-osteoclastogenic function of PARP1 was confirmed in vivo in several mouse models of PARP1 loss-of-function or gain-of-function, including a novel model in which PARP1 was conditionally ablated in myeloid cells. Thus, PARP1 ADP-ribosylates H2B to negatively regulate NFATc1 expression and OC differentiation. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Histonas , Osteoclastos , Animales , Diferenciación Celular , Ratones , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFI , Osteoclastos/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Linfocitos T/metabolismo
10.
PLoS Biol ; 16(11): e3000047, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30388107

RESUMEN

Mutated NLRP3 assembles a hyperactive inflammasome, which causes excessive secretion of interleukin (IL)-1ß and IL-18 and, ultimately, a spectrum of autoinflammatory disorders known as cryopyrinopathies of which neonatal-onset multisystem inflammatory disease (NOMID) is the most severe phenotype. NOMID mice phenocopy several features of the human disease as they develop severe systemic inflammation driven by IL-1ß and IL-18 overproduction associated with damage to multiple organs, including spleen, skin, liver, and skeleton. Secretion of IL-1ß and IL-18 requires gasdermin D (GSDMD), which-upon activation by the inflammasomes-translocates to the plasma membrane where it forms pores through which these cytokines are released. However, excessive pore formation resulting from sustained activation of GSDMD compromises membrane integrity and ultimately causes a pro-inflammatory form of cell death, termed pyroptosis. In this study, we first established a strong correlation between NLRP3 inflammasome activation and GSDMD processing and pyroptosis in vitro. Next, we used NOMID mice to determine the extent to which GSDMD-driven pyroptosis influences the pathogenesis of this disorder. Remarkably, all NOMID-associated inflammatory symptoms are prevented upon ablation of GSDMD. Thus, GSDMD-dependent actions are required for the pathogenesis of NOMID in mice.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Síndromes Periódicos Asociados a Criopirina/metabolismo , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Membrana Celular/metabolismo , Síndromes Periódicos Asociados a Criopirina/genética , Síndromes Periódicos Asociados a Criopirina/fisiopatología , Inflamasomas/metabolismo , Inflamación , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Unión a Fosfato , Piroptosis/fisiología
11.
Neurosci Lett ; 687: 88-93, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30243884

RESUMEN

Iron metabolism dysfunction and redox-active iron-induced oxidative stress in the brain may contribute to the pathogenesis of Parkinson's disease. We have previously demonstrated that reduced serum ceruloplasmin level exacerbates nigral iron deposition in Parkinson's disease, although the underlying cause of the low serum ceruloplasmin level in Parkinson's disease remains unknown. Fluorescent quantitative real-time polymerase chain reaction analysis revealed that patients with Parkinson's disease had higher serum levels of microRNA (miR)-520d-5p than controls (p = 0.0011). Patients with Alzheimer's disease or multiple system atrophy did not have significantly elevated miR-520d-5p levels. Expression of miR-520d-5p did not correlate with disease severity or the motor phenotype of Parkinson's disease. Luciferase assays confirmed that miR-520d-5p was associated with ceruloplasmin gene expression, as predicted by the TargetScan tool and miRBase. In vitro experiments showed that miR-520d-5p reduced ceruloplasmin gene expression in the U251 astrocyte cell line. Our data suggest that miR-520d-5p may be a potential regulator of ceruloplasmin gene expression in vitro.


Asunto(s)
Ceruloplasmina/biosíntesis , MicroARNs/sangre , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/diagnóstico , Anciano , Astrocitos/metabolismo , Biomarcadores/sangre , Línea Celular , Ceruloplasmina/genética , Estudios Transversales , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad
12.
J Am Soc Nephrol ; 29(8): 2110-2122, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30002222

RESUMEN

BACKGROUND: We previously reported that mutations in the anillin (ANLN) gene cause familial forms of FSGS. ANLN is an F-actin binding protein that modulates podocyte cell motility and interacts with the phosphoinositide 3-kinase (PI3K) pathway through the slit diaphragm adaptor protein CD2-associated protein (CD2AP). However, it is unclear how the ANLN mutations cause the FSGS phenotype. We hypothesized that the R431C mutation exerts its pathogenic effects by uncoupling ANLN from CD2AP. METHODS: We conducted in vivo complementation assays in zebrafish to determine the effect of the previously identified missense ANLN variants, ANLNR431C and ANLNG618C during development. We also performed in vitro functional assays using human podocyte cell lines stably expressing wild-type ANLN (ANLNWT ) or ANLNR431C . RESULTS: Experiments in anln-deficient zebrafish embryos showed a loss-of-function effect for each ANLN variant. In human podocyte lines, expression of ANLNR431C increased cell migration, proliferation, and apoptosis. Biochemical characterization of ANLNR431C -expressing podocytes revealed hyperactivation of the PI3K/AKT/mTOR/p70S6K/Rac1 signaling axis and activation of mTOR-driven endoplasmic reticulum stress in ANLNR431C -expressing podocytes. Inhibition of mTOR, GSK-3ß, Rac1, or calcineurin ameliorated the effects of ANLNR431C . Additionally, inhibition of the calcineurin/NFAT pathway reduced the expression of endogenous ANLN and mTOR. CONCLUSIONS: The ANLNR431C mutation causes multiple derangements in podocyte function through hyperactivation of PI3K/AKT/mTOR/p70S6K/Rac1 signaling. Our findings suggest that the benefits of calcineurin inhibition in FSGS may be due, in part, to the suppression of ANLN and mTOR. Moreover, these studies illustrate that rational therapeutic targets for familial FSGS can be identified through biochemical characterization of dysregulated podocyte phenotypes.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/genética , Movimiento Celular/genética , Células Cultivadas , Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Humanos , Mutación Missense , Podocitos/metabolismo , Sensibilidad y Especificidad , Transducción de Señal , Pez Cebra , Proteína de Unión al GTP rac1/genética
13.
Sci Rep ; 7(1): 6630, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747793

RESUMEN

The NLRP3 inflammasome senses a variety of signals referred to as danger associated molecular patterns (DAMPs), including those triggered by crystalline particulates or degradation products of extracellular matrix. Since some DAMPs confer tissue-specific activation of the inflammasomes, we tested the hypothesis that bone matrix components function as DAMPs for the NLRP3 inflammasome and regulate osteoclast differentiation. Indeed, bone particles cause exuberant osteoclastogenesis in the presence of RANKL, a response that correlates with NLRP3 abundance and the state of inflammasome activation. To determine the relevance of these findings to bone homeostasis, we studied the impact of Nlrp3 deficiency on bone using pre-clinical mouse models of high bone turnover, including estrogen deficiency and sustained exposure to parathyroid hormone or RANKL. Despite comparable baseline indices of bone mass, bone loss caused by hormonal or RANKL perturbations is significantly reduced in Nlrp3 deficient than in wild type mice. Consistent with the notion that osteolysis releases DAMPs from bone matrix, pharmacologic inhibition of bone resorption by zoledronate attenuates inflammasome activation in mice. Thus, signals originating from bone matrix activate the NLRP3 inflammasome in the osteoclast lineage, and may represent a bone-restricted positive feedback mechanism that amplifies bone resorption in pathologic conditions of accelerated bone turnover.


Asunto(s)
Matriz Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Inflamasomas/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Receptores de Superficie Celular/metabolismo , Animales , Estrógenos/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Hormona Paratiroidea/metabolismo , Ligando RANK/metabolismo
14.
Sci Rep ; 7(1): 4880, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687790

RESUMEN

Skeletal complications are common features of neonatal-onset multisystem inflammatory disease (NOMID), a disorder caused by NLRP3-activating mutations. NOMID mice in which NLRP3 is activated globally exhibit several characteristics of the human disease, including systemic inflammation and cartilage dysplasia, but the mechanisms of skeletal manifestations remain unknown. In this study, we find that activation of NLRP3 in myeloid cells, but not mesenchymal cells triggers chronic inflammation, which ultimately, causes growth plate and epiphyseal dysplasia in mice. These responses are IL-1 signaling-dependent, but independent of PARP1, which also functions downstream of NLRP3 and regulates skeletal homeostasis. Mechanistically, inflammation causes severe anemia and hypoxia in the bone environment, yet down-regulates the HIF-1α pathway in chondrocytes, thereby promoting the demise of these cells. Thus, activation of NLRP3 in hematopoietic cells initiates IL-1ß-driven paracrine cascades, which promote abnormal growth plate development in NOMID mice.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/patología , Placa de Crecimiento/patología , Inflamasomas/metabolismo , Inflamación/fisiopatología , Células Mieloides/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-1/metabolismo , Ratones , Transducción de Señal
15.
Hum Mol Genet ; 26(10): 1927-1941, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334874

RESUMEN

Genomic disorders are the clinical conditions manifested by submicroscopic genomic rearrangements including copy number variants (CNVs). The CNVs can be identified by array-based comparative genomic hybridization (aCGH), the most commonly used technology for molecular diagnostics of genomic disorders. However, clinical aCGH only informs CNVs in the probe-interrogated regions. Neither orientational information nor the resulting genomic rearrangement structure is provided, which is a key to uncovering mutational and pathogenic mechanisms underlying genomic disorders. Long-range polymerase chain reaction (PCR) is a traditional approach to obtain CNV breakpoint junction, but this method is inefficient when challenged by structural complexity such as often found at the PLP1 locus in association with Pelizaeus-Merzbacher disease (PMD). Here we introduced 'capture and single-molecule real-time sequencing' (cap-SMRT-seq) and newly developed 'asymmetry linker-mediated nested PCR walking' (ALN-walking) for CNV breakpoint sequencing in 49 subjects with PMD-associated CNVs. Remarkably, 29 (94%) of the 31 CNV breakpoint junctions unobtainable by conventional long-range PCR were resolved by cap-SMRT-seq and ALN-walking. Notably, unexpected CNV complexities, including inter-chromosomal rearrangements that cannot be resolved by aCGH, were revealed by efficient breakpoint sequencing. These sequence-based structures of PMD-associated CNVs further support the role of DNA replicative mechanisms in CNV mutagenesis, and facilitate genotype-phenotype correlation studies. Intriguingly, the lengths of gained segments by CNVs are strongly correlated with clinical severity in PMD, potentially reflecting the functional contribution of other dosage-sensitive genes besides PLP1. Our study provides new efficient experimental approaches (especially ALN-walking) for CNV breakpoint sequencing and highlights their importance in uncovering CNV mutagenesis and pathogenesis in genomic disorders.


Asunto(s)
Hibridación Genómica Comparativa/métodos , Variaciones en el Número de Copia de ADN/genética , Variaciones en el Número de Copia de ADN/fisiología , Enfermedad de Pelizaeus-Merzbacher/genética , Secuencia de Bases , Replicación del ADN , Femenino , Dosificación de Gen/genética , Duplicación de Gen/genética , Reordenamiento Génico/genética , Estudios de Asociación Genética/métodos , Genoma Humano , Genómica/métodos , Humanos , Masculino , Mutación , Enfermedad de Pelizaeus-Merzbacher/sangre , Análisis de Secuencia de ADN/métodos
16.
Sci Rep ; 7: 44271, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28333167

RESUMEN

Cockayne syndrome is an autosomal recessive disorder principally characterized by postnatal growth failure and progressive neurological dysfunction, due primarily to mutations in ERCC6 and ERCC8. Here, we report our diagnostic experience for two patients in a Chinese family suspected on clinical grounds to have Cockayne syndrome. Using multiple molecular techniques, including whole exome sequencing, array comparative genomic hybridization and quantitative polymerase chain reaction, we identified compound heterozygosity for a maternal splicing variant (chr5:60195556, NM_000082:c.618-2A > G) and a paternal complex deletion/inversion/deletion rearrangement removing exon 4 of ERCC8, confirming the suspected pathogenesis in these two subjects. Microhomology (TAA and AGCT) at the breakpoints indicated that microhomology-mediated FoSTeS events were involved in this complex ERCC8 rearrangement. This diagnostic experience illustrates the value of high-throughput genomic technologies combined with detailed phenotypic assessment in clinical genetic diagnosis.


Asunto(s)
Secuencia de Bases , Síndrome de Cockayne/genética , Enzimas Reparadoras del ADN/genética , Eliminación de Secuencia , Factores de Transcripción/genética , Adolescente , Pueblo Asiatico , Preescolar , Síndrome de Cockayne/diagnóstico , Síndrome de Cockayne/etnología , Síndrome de Cockayne/patología , Hibridación Genómica Comparativa , Enzimas Reparadoras del ADN/deficiencia , Exones , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Empalme del ARN , Hermanos , Factores de Transcripción/deficiencia , Secuenciación del Exoma
17.
Nat Neurosci ; 19(11): 1442-1453, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27668389

RESUMEN

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, ∼20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.


Asunto(s)
Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Herencia Multifactorial/genética , Esquizofrenia/genética , Encéfalo/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Riesgo
18.
Nature ; 530(7588): 98-102, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26808898

RESUMEN

Methyl-CpG binding protein 2 (MeCP2) has crucial roles in transcriptional regulation and microRNA processing. Mutations in the MECP2 gene are found in 90% of patients with Rett syndrome, a severe developmental disorder with autistic phenotypes. Duplications of MECP2-containing genomic segments cause the MECP2 duplication syndrome, which shares core symptoms with autism spectrum disorders. Although Mecp2-null mice recapitulate most developmental and behavioural defects seen in patients with Rett syndrome, it has been difficult to identify autism-like behaviours in the mouse model of MeCP2 overexpression. Here we report that lentivirus-based transgenic cynomolgus monkeys (Macaca fascicularis) expressing human MeCP2 in the brain exhibit autism-like behaviours and show germline transmission of the transgene. Expression of the MECP2 transgene was confirmed by western blotting and immunostaining of brain tissues of transgenic monkeys. Genomic integration sites of the transgenes were characterized by a deep-sequencing-based method. As compared to wild-type monkeys, MECP2 transgenic monkeys exhibited a higher frequency of repetitive circular locomotion and increased stress responses, as measured by the threat-related anxiety and defensive test. The transgenic monkeys showed less interaction with wild-type monkeys within the same group, and also a reduced interaction time when paired with other transgenic monkeys in social interaction tests. The cognitive functions of the transgenic monkeys were largely normal in the Wisconsin general test apparatus, although some showed signs of stereotypic cognitive behaviours. Notably, we succeeded in generating five F1 offspring of MECP2 transgenic monkeys by intracytoplasmic sperm injection with sperm from one F0 transgenic monkey, showing germline transmission and Mendelian segregation of several MECP2 transgenes in the F1 progeny. Moreover, F1 transgenic monkeys also showed reduced social interactions when tested in pairs, as compared to wild-type monkeys of similar age. Together, these results indicate the feasibility and reliability of using genetically engineered non-human primates to study brain disorders.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/psicología , Modelos Animales de Enfermedad , Mutación de Línea Germinal/genética , Herencia/genética , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Animales Modificados Genéticamente , Ansiedad/genética , Ansiedad/psicología , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Encéfalo/metabolismo , Cognición/fisiología , Femenino , Humanos , Locomoción/genética , Locomoción/fisiología , Macaca fascicularis , Masculino , Fenotipo , Conducta Social , Inyecciones de Esperma Intracitoplasmáticas , Transgenes/genética
19.
Neurosci Bull ; 31(2): 245-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25758665

RESUMEN

Our previous studies have demonstrated that ceruloplasmin (CP) dysmetabolism is correlated with Parkinson's disease (PD). However, the causes of decreased serum CP levels in PD patients remain to be clarified. This study aimed to explore the potential association between genetic variants of the CP gene and PD. Clinical features, serum CP levels, and the CP gene (both promoter and coding regions) were analyzed in 60 PD patients and 50 controls. A luciferase reporter system was used to investigate the function of promoter single-nucleotide polymorphisms (SNPs). High-density comparative genomic hybridization microarrays were also used to detect large-scale copy-number variations in CP and an additional 47 genes involved in PD and/or copper/iron metabolism. The frequencies of eight SNPs (one intronic SNP and seven promoter SNPs of the CP gene) and their haplotypes were significantly different between PD patients, especially those with lowered serum CP levels, and controls. However, the luciferase reporter system revealed no significant effect of the risk haplotype on promoter activity of the CP gene. Neither these SNPs nor their haplotypes were correlated with the Hoehn and Yahr staging of PD. The results of this study suggest that common genetic variants of CP are associated with PD and further investigation is needed to explore their functions in PD.


Asunto(s)
Ceruloplasmina/genética , Predisposición Genética a la Enfermedad/genética , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple/genética , Anciano , Estudios de Casos y Controles , Línea Celular , Hibridación Genómica Comparativa , Femenino , Genotipo , Haplotipos , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Transfección
20.
Mol Genet Genomics ; 289(5): 755-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24737421

RESUMEN

Rare copy number variations (CNVs) generated by human genomic rearrangements have been shown to play an important role in pathogenesis of human diseases and cancers. CNV breakpoint analysis can help define genomic location, genetic content and sequence structure of pathogenic CNVs. This process is vital to elucidate CNV mutational mechanism and etiology of CNV-associated disorders. However, it is technically challenging to map CNV breakpoints at base-pair level, especially in the genomic regions with sequence complexity. In this study, we developed a new method of capture and breakpoint approaching sequencing (CBAS) to efficiently obtain CNV breakpoint sequences. This strategy is independent of CNV structures and applicable to various CNV types. As was demonstrated in CNV-associated patients with neurological disorders, CBAS achieved fine mapping of breakpoint sequences for compound deletion, complex duplication, and translocation. Intriguingly, CBAS also revealed unexpected CNV complexity involving long-range DNA rearrangement. Our observations showed that CBAS is an efficient method for obtaining CNV breakpoint sequence and mapping insertional events as well. This method can facilitate the researches on CNV-associated human diseases and cancers. CBAS is also applicable to mapping the integration sites of retrovirus (such as HIV) and transgenes in model organisms.


Asunto(s)
Puntos de Rotura del Cromosoma , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Secuencia de Bases , Cromosomas Humanos X/genética , Hibridación Genómica Comparativa , Duplicación de Gen , Reordenamiento Génico , Humanos , Discapacidad Intelectual/genética , Proteína Proteolipídica de la Mielina/genética , Enfermedad de Parkinson/genética , Enfermedad de Pelizaeus-Merzbacher/genética , Eliminación de Secuencia , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...