Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
3.
Mol Cancer ; 14: 190, 2015 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-26545365

RESUMEN

BACKGROUND: MicroRNAs (miR, miRNAs) play pivotal roles in numerous physiological and pathophysiological contexts. We investigated whether miR-362-5p act as an oncogene in chronic myeloid leukaemia (CML) and aimed to understand its potential underlying mechanisms. METHODS: We compared the miR-362-5p expression levels between CML and non-CML cell lines, and between fresh blood samples from CML patients and normal healthy controls using quantitative real-time PCR (qPCR). Cell counting kit-8 (CCK-8) and Annexin V-FITC/PI analyses were used to measure the effects of miR-362-5p on proliferation and apoptosis, and Transwell assays were used to evaluate migration and invasion. A xenograft model was used to examine in vivo tumourigenicity. The potential target of miR-362-5p was confirmed by a luciferase reporter assay, qPCR and western blotting. Involvement of the JNK1/2 and P38 pathways was investigated by western blotting. RESULTS: miR-362-5p was up-regulated in CML cell lines and fresh blood samples from CML patients, and was associated with Growth arrest and DNA damage-inducible (GADD)45α down-regulation. Inhibition of miR-362-5p simultaneously repressed tumour growth and up-regulated GADD45α expression in a xenograft model. Consistently, the knockdown of GADD45α expression partially neutralized the effects of miR-362-5p inhibition. Furthermore study suggested that GADD45α mediated downstream the effects of miR-362-5p, which might indirectly regulates the activation of the JNK1/2 and P38 signalling pathways. CONCLUSION: miR-362-5p acts as an oncomiR that down-regulates GADD45α, which consequently activates the JNK1/2 and P38 signalling. This finding provides novel insights into CML leukaemogenesis and may help identify new diagnostic and therapeutic targets.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , MicroARNs/fisiología , Proteínas Nucleares/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Células HEK293 , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Proteínas Nucleares/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Mol Cancer ; 14: 10, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25622857

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, is the third leading cause of cancer-related death in human. Alcohol is a known risk factor for HCC. However it is still unclear whether and how alcohol enhances the progression and metastasis of existing HCC. METHODS AND RESULTS: We first retrospectively investigated 52 HCC patients (24 alcohol-drinkers and 28 non-drinkers), and found a positive correlation between alcohol consumption and advanced Tumor-Node-Metastasis (TNM) stages, higher vessel invasion and poorer prognosis. In vitro and in vivo experiments further indicated that alcohol promoted the progression and migration/invasion of HCC. Specifically, in a 3-D tumor/endothelial co-culture system, we found that alcohol enhanced the migration/invasion of HepG2 cells and increased tumor angiogenesis. Consistently, higher expression of VEGF, MCP-1 and NF-κB was observed in HCC tissues of alcohol-drinkers. Alcohol induced the accumulation of intracellular reactive oxygen species (ROS) and the activation of NF-κB signaling in HepG2 cells. Conversely, blockage of alcohol-mediated ROS accumulation and NF-κB signaling inhibited alcohol-induced expression of VEGF and MCP-1, the tumor growth, angiogenesis and metastasis. CONCLUSION: This study suggested that chronic moderate alcohol consumption may promote the progression and metastasis of HCC; the oncogenic effect may be at least partially mediated by the ROS accumulation and NF-ĸB-dependent VEGF and MCP-1 up-regulation.


Asunto(s)
Carcinoma Hepatocelular/genética , Movimiento Celular/genética , Etanol/efectos adversos , FN-kappa B/genética , Invasividad Neoplásica/genética , Transducción de Señal/genética , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Quimiocina CCL2/genética , Progresión de la Enfermedad , Femenino , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Células 3T3 NIH , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Estudios Retrospectivos , Factor A de Crecimiento Endotelial Vascular/genética
5.
BMC Cancer ; 15: 525, 2015 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-26187637

RESUMEN

BACKGROUND: The lymph node metastasis is a key early step of the tumor metastatic process. VEGFD-mediated tumor lymphangiogenesis plays a key role, since down-regulation of p-VEGFR-3 could block the lymph node metastasis. YL529 has been reported to possess potent anti-angiogenesis and antitumor activities; however, its roles in tumor-associated lymphangiogenesis and lymphatic metastasis remain unclear. METHOD: We investigated the effect of YL529 on tumor-associated lymphangiogenesis and lymph node metastasis using in vitro lymph node metastasis models and in vivo subcutaneous tumor models in C57 BL/6 mice. RESULT: We found that YL529 inhibited VEGF-D-induced survival, proliferation and tube-formation of Human Lymphatic Endothelial Cells. Furthermore, in established in vitro and in vivo lymph node metastasis models using VEGF-D-LL/2 cells, YL529 significantly inhibited the tumor-associated lymphangiogenesis and metastasis. At molecular level, YL529 down-regulated p-VEGFR-3, p-JNK and Bax while up-regulated Bcl-2. CONCLUSION: YL529 provided the therapeutic benefits by both direct effects on tumor cells and inhibiting lymphangiogenesis and metastasis via the VEGFR-3 signaling pathway, which may have significant direct clinical implications.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Bencenosulfonatos/administración & dosificación , Linfangiogénesis/efectos de los fármacos , Metástasis Linfática/prevención & control , Picolinas/administración & dosificación , Factor D de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Picolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Inflamm Res ; 63(3): 207-15, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327087

RESUMEN

INTRODUCTION: Previous studies found that neuron specific enolase promoter (Nse-BMP4) transgenic mice have increased expression of the nociceptive mediator, substance P and exaggerated local injury responses associated with heterotopic ossification (HO). It is of interest great to know the pain responses in these mice and how the opioid signaling is involved in the downstream events such as mast cell (MC) activation. MATERIALS AND METHODS: This study utilized a transgenic mouse model of HO in which BMP4 is expressed under the control of the Nse-BMP4. The tactile sensitivity and the cold sensitivity of the mice were measured in a classic inflammatory pain model (carrageenan solution injected into the plantar surface of the left hind paw). The MC activation and the expression profiles of different components in the opioid signaling were demonstrated through routine histology and immunohistochemistry and Western blotting, in the superficial and deep muscle injury models. RESULTS: We found that the pain responses in these mice were paradoxically attenuated or unchanged, and we also found increased expression of both Methionine Enkephalin (Met-Enk), and the µ-opioid receptor (MOR). Met-Enk and MOR both co-localized within activated MCs in limb tissues. Further, Nse-BMP4;MOR(-/-) double mutant mice showed attenuated MC activation and had a significant reduction in HO formation in response to injuries. CONCLUSIONS: These observations suggest that opioid signaling may play a key role in MC activation and the downstream inflammatory responses associated with HO. In addition to providing insight into the role of MC activation and associated injury responses in HO, these findings suggest opioid signaling as a potential therapeutic target in HO and possibly others disorders involving MC activation.


Asunto(s)
Encefalina Metionina/fisiología , Mastocitos/fisiología , Osificación Heterotópica/fisiopatología , Animales , Proteína Morfogenética Ósea 4/genética , Frío , Inmunohistoquímica , Inflamación/complicaciones , Inflamación/patología , Mastocitos/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/lesiones , Mutación/genética , Mutación/fisiología , Nocicepción/fisiología , Osificación Heterotópica/patología , Dimensión del Dolor , Fosfopiruvato Hidratasa/genética , Estimulación Física , Receptores Opioides mu/fisiología , Transducción de Señal/fisiología
7.
Blood ; 116(23): 4968-77, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20724542

RESUMEN

FLIP is a well-established suppressor of death receptor-mediated apoptosis. To define its essential in vivo role in myeloid cells, we generated and characterized mice with Flip conditionally deleted in the myeloid lineage. Myeloid specific Flip-deficient mice exhibited growth retardation, premature death, and splenomegaly with altered architecture and extramedullary hematopoiesis. They also displayed a dramatic increase of circulating neutrophils and multiorgan neutrophil infiltration. In contrast, although circulating inflammatory monocytes were also significantly increased, macrophages in the spleen, lymph nodes, and the peritoneal cavity were reduced. In ex vivo cultures, bone marrow progenitor cells failed to differentiate into macrophages when Flip was deleted. Mixed bone marrow chimera experiments using cells from Flip-deficient and wild-type mice did not demonstrate an inflammatory phenotype. These observations demonstrate that FLIP is necessary for macrophage differentiation and the homeostatic regulation of granulopoiesis.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Diferenciación Celular/genética , Granulocitos/citología , Homeostasis/genética , Macrófagos/citología , Mielopoyesis/genética , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Granulocitos/metabolismo , Inmunohistoquímica , Inmunofenotipificación , Ganglios Linfáticos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología
8.
J Cell Biochem ; 112(10): 2759-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21748788

RESUMEN

Heterotopic ossification (HO) is a disabling condition associated with neurologic injury, inflammation, and overactive bone morphogenetic protein (BMP) signaling. The inductive factors involved in lesion formation are unknown. We found that the expression of the neuro-inflammatory factor Substance P (SP) is dramatically increased in early lesional tissue in patients who have either fibrodysplasia ossificans progressiva (FOP) or acquired HO, and in three independent mouse models of HO. In Nse-BMP4, a mouse model of HO, robust HO forms in response to tissue injury; however, null mutations of the preprotachykinin (PPT) gene encoding SP prevent HO. Importantly, ablation of SP(+) sensory neurons, treatment with an antagonist of SP receptor NK1r, deletion of NK1r gene, or genetic down-regulation of NK1r-expressing mast cells also profoundly inhibit injury-induced HO. These observations establish a potent neuro-inflammatory induction and amplification circuit for BMP-dependent HO lesion formation, and identify novel molecular targets for prevention of HO.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Osificación Heterotópica/metabolismo , Sustancia P/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Morfogenéticas Óseas/genética , Femenino , Humanos , Inmunohistoquímica , Isoindoles/farmacología , Masculino , Ratones , Ratones Transgénicos , Miositis Osificante/genética , Miositis Osificante/metabolismo , Antagonistas del Receptor de Neuroquinina-1 , Osificación Heterotópica/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores de Neuroquinina-1/metabolismo , Células Receptoras Sensoriales/metabolismo , Taquicininas/genética , Taquicininas/metabolismo
9.
J Neurosci Res ; 89(3): 299-309, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21259317

RESUMEN

Members of the Hes and Hey families of basic helix-loop-helix transcription factors are regarded as Notch target genes that generally inhibit neuronal differentiation of neural progenitor cells. We found that HeyL, contrary to the classic function of Hes and Hey factors, promotes neuronal differentiation of neural progenitor cells both in culture and in the embryonic brain in vivo. Furthermore, null mutation of HeyL decreased the rate of neuronal differentiation of cultured neural progenitor cells. HeyL binds to and activates the promoter of the proneural gene neurogenin2, which is inhibited by other Hes and Hey family members, and HeyL is a weak inhibitor of the Hes1 promoter. HeyL is able to bind other Hes and Hey family members, but it cannot bind the Groucho/Tle1 transcriptional corepressor, which mediates the inhibitory effects of the Hes family of factors. Furthermore, although HeyL expression is only weakly augmented by Notch signaling, we found that bone morphogenic protein signaling increases HeyL expression by neural progenitor cells. These observations suggest that HeyL promotes neuronal differentiation of neural progenitor cells by activating proneural genes and by inhibiting the actions of other Hes and Hey family members.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Animales , Animales Recién Nacidos , Antígenos/genética , Astrocitos/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Proteína Morfogenética Ósea 4/metabolismo , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunoprecipitación/métodos , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteoglicanos/genética , Transfección/métodos , Tubulina (Proteína)/metabolismo
10.
J Biomed Sci ; 18: 92, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22168923

RESUMEN

BACKGROUND: Skin stem cells contribute to all three major lineages of epidermal appendages, i.e., the epidermis, the hair follicle, and the sebaceous gland. In hair follicles, highly proliferative committed progenitor cells, called matrix cells, are located at the base of the follicle in the hair bulb. The differentiation of these early progenitor cells leads to specification of a central hair shaft surrounded by an inner root sheath (IRS) and a companion layer. Multiple signaling molecules, including bone morphogenetic proteins (BMPs), have been implicated in this process. METHODS: To further probe the contribution of BMP signaling to hair follicle development and maintenance we employed a transgenic mouse that expresses the BMP inhibitor, Noggin, to disrupt BMP signaling specifically in subset of hair follicle progenitors under the control of neuron specific enolase (Nse) promoter. We then studied the skin tumor phenotypes of the transgenic mice through histology, immunohistochemistry and Western Blotting to delineate the underlying mechanisms. Double transgenic mice expressing BMP as well as noggin under control of the Nse promoter were used to rescue the skin tumor phenotypes. RESULTS: We found that the transgene is expressed specifically in a subpopulation of P-cadherin positive progenitor cells in Nse-Noggin mice. Blocking BMP signaling in this cell population led to benign hair follicle-derived neoplasias resembling human trichofolliculomas, associated with down-regulation of E-cadherin expression and dynamic regulation of CD44. CONCLUSIONS: These observations further define a critical role for BMP signaling in maintaining the homeostasis of hair follicles, and suggest that dysregulation of BMP signaling in hair follicle progenitors may contribute to human trichofolliculoma.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Cadherinas/genética , Quiste Folicular/metabolismo , Enfermedades del Cabello/metabolismo , Folículo Piloso/metabolismo , Neoplasias Basocelulares/metabolismo , Neoplasias Cutáneas/metabolismo , Células Madre/patología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Quiste Folicular/patología , Enfermedades del Cabello/patología , Folículo Piloso/patología , Receptores de Hialuranos/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Basocelulares/patología , Transducción de Señal , Neoplasias Cutáneas/patología , Células Madre/metabolismo
11.
J Biomed Biotechnol ; 2011: 309287, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20981294

RESUMEN

Heterotopic ossification (HO) is the formation of marrow-containing bone outside of the normal skeleton. Acquired HO following traumatic events is a common and costly clinical complication. In contrast, hereditary HO is rarer, progressive, and life-threatening. Substantial effort has been directed towards understanding the mechanisms underlying HO and finding efficient treatments. However, one crucial limiting factor has been the lack of relevant animal models. This article reviews the major currently available animal models, summarizes some of the insights gained from these studies, and discusses the potential future challenges and directions in HO research.


Asunto(s)
Modelos Animales de Enfermedad , Osificación Heterotópica/patología , Animales , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Osificación Heterotópica/genética
12.
Stroke ; 41(2): 357-62, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20019326

RESUMEN

BACKGROUND AND PURPOSE: Bone morphogenetic proteins and their receptors are expressed in adult brains, and their expression levels increase after cerebral ischemia. The brain also expresses an inhibitor of bone morphogenetic protein signaling, noggin, but the role of noggin in ischemic disease outcome has not been studied. METHODS: We used transgenic mice overexpressing noggin to assess whether inhibition of bone morphogenetic protein signaling affects ischemic injury responses after permanent middle cerebral artery occlusion. RESULTS: Transgenic mice overexpressing noggin mice had significantly smaller infarct volumes and lower motor deficits compared to wild-type mice. CD11b(+) and IBA1(+) microglia along with oligodendroglial progenitors were significantly increased in transgenic mice overexpressing noggin mice at 14 days after permanent middle cerebral artery occlusion. CONCLUSIONS: These results provide genetic evidence that overexpression of noggin reduces ischemic brain injury after permanent middle cerebral artery occlusion via enhanced activation of microglia and oligodendrogenesis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Proteínas Portadoras/genética , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/terapia , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Infarto Encefálico/terapia , Isquemia Encefálica/fisiopatología , Proliferación Celular , Citoprotección/fisiología , Modelos Animales de Enfermedad , Terapia Genética/métodos , Gliosis/patología , Gliosis/fisiopatología , Gliosis/terapia , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Regeneración Nerviosa/fisiología , Oligodendroglía/metabolismo , Paresia/patología , Paresia/fisiopatología , Paresia/terapia , Recuperación de la Función/fisiología , Células Madre/metabolismo
13.
Stem Cells ; 27(1): 150-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18832590

RESUMEN

Heterotopic ossification (HO), the abnormal formation of true marrow-containing bone within extraskeletal soft tissues, is a serious bony disorder that may be either acquired or hereditary. We utilized an animal model of the genetic disorder fibrodysplasia ossificans progressiva to examine the cellular mechanisms underlying HO. We found that HO in these animals was triggered by soft tissue injuries and that the effects were mediated by macrophages. Spreading of HO beyond the initial injury site was mediated by an abnormal adaptive immune system. These observations suggest that dysregulation of local stem/progenitor cells could be a common cellular mechanism for typical HO irrespective of the signal initiating the bone formation.


Asunto(s)
Osificación Heterotópica/patología , Células Madre/patología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Linaje de la Célula , Sistema Inmunológico/inmunología , Integrasas/metabolismo , Macrófagos/inmunología , Ratones , Modelos Biológicos , Músculos/patología , Osificación Heterotópica/inmunología , Fosfopiruvato Hidratasa/metabolismo , Piel/patología
14.
Histol Histopathol ; 34(4): 303-312, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30422303

RESUMEN

Wnt signaling, canonical or non-canonical, plays conserved roles in numerous physiological and pathological processes. However, it is well beyond the scope of this review to cover all functional aspects of Wnt signaling in different contexts at reasonable depth; therefore this review intends to cover only the roles of Wnt signaling in bone biology; more specifically, we intend to first update the roles of Wnt signaling in physiological bone process, including in osteogenesis and chondrogenesis, since recent years have witnessed tremendous progressions in this area, and then we seek to extend our understanding to the pathological bone process, especially to the heterotopic ossification (HO), even though the understanding of Wnt signaling in HO has been limited. We then further clarify the potential crosstalking between Wnt and other conserved signaling pathways, including FGF, GPCR and Hif1α pathways. Overall, our goal is to update the progressions, identify the general theme and the knowledge gaps and discuss the potential promising avenue for future applications in HO prevention and treatment.


Asunto(s)
Condrogénesis/fisiología , Osificación Heterotópica/metabolismo , Osteogénesis/fisiología , Vía de Señalización Wnt/fisiología , Animales , Humanos
15.
J Cosmet Dermatol ; 18(6): 1930-1934, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30859684

RESUMEN

BACKGROUND: Pachyonychia congenita (PC), a rare autosomal dominant disorder, is featured by significant hypertrophic nail, palmoplantar keratoderma, and plantar pain. It is caused by the mutation of KRT6A, KRT6B, KRT6C, KRT16, or KRT17. AIMS: To identify the gene mutation caused the PC in a Chinese family. PATIENTS/METHODS: Genomic DNA was extracted from peripheral blood samples of five patients and six healthy individuals. Genomic DNA of three patients was sequenced by whole-exome sequencing (WES). Then, exons 6 of KRT16 of all samples were amplified by polymerase chain reaction (PCR), and PCR products were sequenced to identify potential mutations. RESULTS: We identified the proline substitution mutation p.Leu421Pro (c.1262T>C) in the 2B domain of K16 that is associated with PC in a Chinese family. The same mutation was not found in the six healthy individuals of the family. CONCLUSIONS: The mutation found in this study is the first report in China. So far, 25 mutations in KRT16 have been reportedly associated with PC. Twenty-one mutations are located on exon 1, and four mutations on exon 6.


Asunto(s)
Queratina-16/genética , Mutación , Paquioniquia Congénita/genética , Pueblo Asiatico/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje
16.
Bone Res ; 7: 33, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31700694

RESUMEN

Heterotopic ossification (HO), true bone formation in soft tissue, is closely associated with abnormal injury/immune responses. We hypothesized that a key underlying mechanism of HO might be injury-induced dysregulation of immune checkpoint proteins (ICs). We found that the earliest stages of HO are characterized by enhanced infiltration of polarized macrophages into sites of minor injuries in an animal model of HO. The non-specific immune suppressants, Rapamycin and Ebselen, prevented HO providing evidence of the central role of the immune responses. We examined the expression pattern of ICs and found that they are dysregulated in HO lesions. More importantly, loss of function of inhibitory ICs (including PD1, PD-L1, and CD152) markedly inhibited HO, whereas loss of function of stimulatory ICs (including CD40L and OX-40L) facilitated HO. These findings suggest that IC inhibitors may provide a therapeutic approach to prevent or limit the extent of HO.

17.
Stem Cell Res Ther ; 10(1): 14, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30635039

RESUMEN

BACKGROUND: Heterotopic ossification (HO), either acquired (aHO) or hereditary, such as fibrodysplasia ossificans progressiva (FOP), is a serious condition without effective treatment. Understanding of the core process of injury-induced HO is still severely limited. METHODS: Double-pulse thymidine analog labeling was used to explore the distinctive domains evolved in injury-induced lesions in an animal model of HO (Nse-BMP4). Histological studies were performed to see whether a similar zonal pattern is also consistently found in biopsies from patients with aHO and FOP. In vivo clonal analysis with Rainbow mice, genetic loss-of-function studies with diphtheria toxin A (DTA)-mediated depletion and lineage tracing with Zsgreen reporter mice were used to obtain further evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells. Immunohistochemistry was used to test whether vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Similar methods also were employed to further understand the signaling pathways that regulate the niche and the resultant HO. RESULTS: We found that distinctive domains evolved in injury-induced lesions, including, from outside-in, a mesenchymal stem cell (MSC) niche, a transient domain and an inner differentiated core in an animal model of HO (Nse-BMP4). A similar zonal structure was found in patients with aHO and FOP. In vivo clonal analysis with Rainbow mice and genetic loss-of-function studies with DTA provided evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells; consistently, vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Further mechanistic study found that BMP and hedgehog (Hh) signaling co-regulate the niche and the resultant HO. CONCLUSIONS: Available data provide evidence of a potential core mechanism in which multiple disease-specific cellular and extracellular molecular elements form a unique local microenvironment, i.e., an injury-induced stem cell niche, which regulates the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs). The implication for HO is that therapeutic approaches must consider several different disease specific factors as parts of a functional unit, instead of treating one factor at a time.


Asunto(s)
Miositis Osificante/genética , Osificación Heterotópica/genética , Osteogénesis/genética , Nicho de Células Madre/genética , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Toxina Diftérica/genética , Modelos Animales de Enfermedad , Transportador 1 de Aminoácidos Excitadores/genética , Humanos , Mutación con Pérdida de Función/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Miositis Osificante/patología , Miositis Osificante/terapia , Osificación Heterotópica/patología , Osificación Heterotópica/terapia , Fragmentos de Péptidos/genética , Receptor TIE-2/genética , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1/genética
18.
Bone ; 109: 43-48, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28455214

RESUMEN

Heterotopic ossification (HO), a serious disorder of extra-skeletal bone formation, occurs as a common complication of trauma or in rare genetic disorders. Many conserved signaling pathways have been implicated in HO; however, the exact underlying molecular mechanisms for many forms of HO are still unclear. The emerging picture is that dysregulation of bone morphogenetic protein (BMP) signaling plays a central role in the process, but that other conserved signaling pathways, such as Hedgehog (HH), Wnt/ß-catenin and Fibroblast growth factors (FGF), are also involved, either through cross-talk with BMP signaling or through other independent mechanisms. Deep understanding of the conserved signaling pathways is necessary for the effective prevention and treatment of HO. In this review, we update and integrate recent progress in this area. Hopefully, our discussion will point to novel promising, druggable loci for further translational research and successful clinical applications.


Asunto(s)
Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Miositis Osificante/genética , Miositis Osificante/metabolismo , Osificación Heterotópica/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
19.
Bone ; 109: 71-79, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28645539

RESUMEN

Heterotopic ossification (HO), acquired or hereditary, endochondral or intramembranous, is the formation of true bone outside the normal skeleton. Since perivascular Gli1+ progenitors contribute to injury induced organ fibrosis, and CD133 is expressed by a variety of populations of adult stem cells, this study utilized Cre-lox based genetic lineage tracing to test the contribution to endochondral HO of adult stem/progenitor cells that expressed either Gli1 or CD133. We found that both lineages contributed broadly to different normal tissues with distinct patterns, but that only Gli1-creERT labeled stem/progenitor cells contributed to all stages of endochondral HO in a BMP dependent, injury induced, transgenic mouse model. Hedgehog (Hh) signaling was abnormal at endochondral HO lesion sites with increased signaling surrounding the lesion but diminished signaling within it. Thus, local dysregulation of Hh signaling participates in the pathophysiology of endochondral HO. However, unlike a previous report of intramembranous HO, systemic inhibition of Hh signaling was insufficient to prevent the initiation of the endochondral HO process or to treat the existing endochondral HO, suggesting that Hh participates in, but is not essential for endochondral HO in this model. This could potentially reflect the underlying difference between intramembranous and endochondral HO. Nevertheless, identification of this novel stem/precursor cell population as a HO-contributing cell population provides a potential drugable target.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Osteogénesis/fisiología , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Transgénicos , Osteogénesis/genética , Pirimidinonas/farmacología , Transducción de Señal/genética , Transducción de Señal/fisiología , Tiofenos/farmacología
20.
Histol Histopathol ; 32(10): 977-985, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28328009

RESUMEN

Bone marrow contains a non-hematopoietic, clonogenic, multipotent population of stromal cells that are later called mesenchymal stem cells (MSC). Similar cells that share many common features with MSC are also found in other organs, which are thought to contribute both to normal tissue regeneration and to pathological processes such as heterotopic ossification (HO), the formation of ectopic bone in soft tissue. Understanding the microenvironmental factors that regulate MSC in vivo is essential both for understanding the biology of the stem cells and for effective translational applications of MSC. Unfortunately, this important aspect has been largely underappreciated. This review tries to raise the attention and highlight this critical issue by updating the relevant literature along with discussions of the key issues in the area.


Asunto(s)
Microambiente Celular , Células Madre Mesenquimatosas/patología , Osificación Heterotópica/patología , Animales , Humanos , Regeneración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA