Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(12)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38781018

RESUMO

We present a transcriptomic analysis that provides a better understanding of regulatory mechanisms within the healthy and injured periosteum. The focus of this work is on characterizing early events controlling bone healing during formation of periosteal callus on day 3 after fracture. Building on our previous findings showing that induced Notch1 signaling in osteoprogenitors leads to better healing, we compared samples in which the Notch 1 intracellular domain is overexpressed by periosteal stem/progenitor cells, with control intact and fractured periosteum. Molecular mechanisms and changes in skeletal stem/progenitor cells (SSPCs) and other cell populations within the callus, including hematopoietic lineages, were determined. Notably, Notch ligands were differentially expressed in endothelial and mesenchymal populations, with Dll4 restricted to endothelial cells, whereas Jag1 was expressed by mesenchymal populations. Targeted deletion of Dll4 in endothelial cells using Cdh5CreER resulted in negative effects on early fracture healing, while deletion in SSPCs using α-smooth muscle actin-CreER did not impact bone healing. Translating these observations into a clinically relevant model of bone healing revealed the beneficial effects of delivering Notch ligands alongside the osteogenic inducer, BMP2. These findings provide insights into the regulatory mechanisms within the healthy and injured periosteum, paving the way for novel translational approaches to bone healing.


Assuntos
Células Endoteliais , Consolidação da Fratura , Proteína Jagged-1 , Periósteo , Transdução de Sinais , Animais , Camundongos , Proteína Jagged-1/metabolismo , Proteína Jagged-1/genética , Células Endoteliais/metabolismo , Periósteo/metabolismo , Periósteo/citologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Células-Tronco Mesenquimais/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética , Osteogênese/genética , Receptor Notch1/metabolismo , Receptor Notch1/genética , Masculino , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética
2.
Stem Cells ; 41(5): 493-504, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36888549

RESUMO

Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.


Assuntos
Calo Ósseo , Proteínas RGS , Camundongos , Animais , Osteocalcina/metabolismo , Calo Ósseo/metabolismo , Calo Ósseo/patologia , Osteogênese , Consolidação da Fratura/fisiologia , Condrócitos/metabolismo , Camundongos Transgênicos , Osteoblastos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo
3.
J Orthop Res ; 38(11): 2350-2361, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32141629

RESUMO

Fracture healing involves interactions of different cell types, driven by various growth factors, and signaling cascades. Periosteal mesenchymal progenitor cells give rise to the majority of osteoblasts and chondrocytes in a fracture callus. Notch signaling has emerged as an important regulator of skeletal cell proliferation and differentiation. We investigated the effects of Notch signaling during the fracture healing process. Increased Notch signaling in osteochondroprogenitor cells driven by overexpression of Notch1 intracellular domain (NICD1) (αSMACreERT2 mice crossed with Rosa-NICD1) during fracture resulted in less cartilage, more mineralized callus tissue, and stronger and stiffer bones after 3 weeks. Periosteal cells overexpressing NICD1 showed increased proliferation and migration in vitro. In vivo data confirmed that increased Notch1 signaling caused expansion of alpha-smooth muscle actin (αSMA)-positive cells and their progeny including αSMA-derived osteoblasts in the callus without affecting osteoclast numbers. In contrast, anti-NRR1 antibody treatment to inhibit Notch1 signaling resulted in increased callus cartilage area, reduced callus bone mass, and reduced biomechanical strength. Our study shows a positive effect of induced Notch1 signaling on the fracture healing process, suggesting that stimulating the Notch pathway could be beneficial for fracture repair.


Assuntos
Consolidação da Fratura , Receptor Notch1/metabolismo , Animais , Feminino , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Receptor Notch1/antagonistas & inibidores
4.
Stem Cells ; 38(6): 769-781, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32053258

RESUMO

Bone remodeling and regeneration are dependent on resident stem/progenitor cells with the ability to replenish mature osteoblasts and repair the skeleton. Using lineage tracing approaches, we identified a population of Dmp1+ cells that reside within cortical bone and are distinct from osteocytes. Our aims were to characterize this stromal population of transcortical perivascular cells (TPCs) in their resident niche and evaluate their osteogenic potential. To distinguish this population from osteoblasts/osteocytes, we crossed mice containing inducible DMP1CreERT2/Ai9 Tomato reporter (iDMP/T) with Col2.3GFP reporter (ColGFP), a marker of osteoblasts and osteocytes. We observed iDMP/T+;ColGFP- TPCs within cortical bone following tamoxifen injection. These cells were perivascular and located within transcortical channels. Ex vivo bone outgrowth cultures showed TPCs migrated out of the channels onto the plate and expressed stem cell markers such as Sca1, platelet derived growth factor receptor beta (PDGFRß), and leptin receptor. In a cortical bone transplantation model, TPCs migrate from their vascular niche within cortical bone and contribute to new osteoblast formation and bone tube closure. Treatment with intermittent parathyroid hormone increased TPC number and differentiation. TPCs were unable to differentiate into adipocytes in the presence of rosiglitazone in vitro or in vivo. Altogether, we have identified and characterized a novel stromal lineage-restricted osteoprogenitor that is associated with transcortical vessels of long bones. Functionally, we have demonstrated that this population can migrate out of cortical bone channels, expand, and differentiate into osteoblasts, therefore serving as a source of progenitors contributing to new bone formation.


Assuntos
Osso e Ossos/fisiopatologia , Osteoblastos/metabolismo , Osteócitos/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
5.
J Cell Physiol ; 235(5): 4643-4654, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31650536

RESUMO

Current treatments of large bone defects are based on autologous or allogenic bone transplantation. Human amniotic fluid stem cells (hAFSCs) were evaluated for their potential in bone regenerative medicine. In this study, hAFSCs were transduced with lentiviral vector harboring red fluorescent protein to investigate their role in the regeneration of critical-size bone defects in calvarial mouse model. To distinguish donor versus recipient cells, a transgenic mouse model carrying GFP fluorescent reporter was used as recipient to follow the fate of hAFSCs transplanted in vivo into Healos® scaffold. Our results showed that transduced hAFSCs can be tracked in vivo directly at the site of transplantation. The presence of GFP positive cells in the scaffold at 3 and 6 weeks after transplantation indicates that donor hAFSCs can recruit host cells during the repair process. These observations help clarify the role of hAFSCs in bone tissue repair.


Assuntos
Líquido Amniótico/citologia , Regeneração Óssea , Osteogênese , Crânio/cirurgia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Animais , Comunicação Celular , Linhagem da Célula , Movimento Celular , Rastreamento de Células , Células Cultivadas , Técnicas de Cocultura , Feminino , Genes Reporter , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Fenótipo , Gravidez , Transdução de Sinais , Crânio/metabolismo , Crânio/patologia , Crânio/fisiopatologia , Fatores de Tempo
6.
Stem Cells ; 38(4): 530-541, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31859429

RESUMO

Osteogenesis imperfecta (OI) is a genetic disorder most commonly caused by mutations associated with type I collagen, resulting in a defective collagen bone matrix. Current treatments for OI focus on pharmaceutical strategies to increase the amount of defective bone matrix, but do not address the underlying collagen defect. Introducing healthy donor stem cells that differentiate into osteoblasts producing normal collagen in OI patients has the potential to increase bone mass and correct the mutant collagen matrix. In this study, donor bone marrow stromal cells (BMSCs, also known as bone marrow mesenchymal stem cells) expressing both αSMACreERT2/Ai9 progenitor reporter and osteoblast reporter Col2.3GFP were locally transplanted into the femur of OI murine (OIM) mice. One month post-transplantation, 18% of the endosteal surface was lined by donor Col2.3GFP expressing osteoblasts indicating robust engraftment. Long-term engraftment in the marrow was observed 3 and 6 months post-transplantation. The presence of Col1a2-expressing donor cell-derived cortical bone matrix was detected in transplanted OIM femurs. Local transplantation of BMSCs increased cortical thickness (+12%), the polar moment of inertia (+14%), bone strength (+30%), and stiffness (+30%) 3 months post-transplantation. Engrafted cells expressed progenitor markers CD51 and Sca-1 up to 3 months post-transplantation. Most importantly, 3 months post-transplantation donor cells maintained the ability to differentiate into Col2.3GFP+ osteoblasts in vitro, and in vivo following secondary transplantation into OIM animals. Locally transplanted BMSCs can improve cortical structure and strength, and persist as continued source of osteoblast progenitors in the OIM mouse for at least 6 months.


Assuntos
Osso e Ossos/metabolismo , Osteogênese Imperfeita/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo , Animais , Osso e Ossos/citologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Fenótipo , Células-Tronco/citologia
7.
Acta Clin Croat ; 57(2): 264-277, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30431719

RESUMO

The objective of this study was to determine differential expression of TFF1, TFF2 and TFF3 genes and proteins in breast tumor subtypes. In addition, we investigated the correlation between TFF genes within tumor subgroups, and TFF genes with clinical and pathologic characteristics of the tumor. Study group included 122 patients with surgically removed breast tumors. Samples were investigated using qRT-PCR and immunohistochemistry. TFF1 and TFF3 genes and proteins were expressed in breast tumors, while the levels of TFF2 gene and protein expression were very low or undetectable. TFF1 was significantly more expressed in benign tumors, while TFF3 was more expressed in malignant tumors. Gene and protein expression of both TFF1 and TFF3 was greater in lymph node-negative tumors, hormone positive tumors, tumors with moderate levels of Ki67 expression, and in grade II tumors. A strong positive correlation was found between TFF1 and TFF3 genes, and the expression of both negatively correlated with Ki67 and the level of tumor histologic differentiation. Our results suggest that TFF1 and TFF3, but not TFF2, may have a role in breast tumor pathogenesis and could be used in the assessment of tumor differentiation and malignancy.


Assuntos
Neoplasias da Mama , Fator Trefoil-1 , Fator Trefoil-2 , Fator Trefoil-3 , Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Mucinas , Proteínas Musculares , Peptídeos , Fator Trefoil-1/metabolismo , Fator Trefoil-2/metabolismo , Fator Trefoil-3/metabolismo
8.
Acta Med Croatica ; 65 Suppl 1: 115-9, 2011 Sep.
Artigo em Servo-Croata (Latino) | MEDLINE | ID: mdl-23126038

RESUMO

Inadequate Pap smear by definition is a specimen in which detection of cervical epithelial abnormality is impossible or uncertain. This causes poorer detection of intraepithelial lesions of a mild and more severe grade, including the possible false-negative diagnosis. Sample adequacy is most crucial in the evaluation of the finding, alerting the gynecologist to the limitations of the finding, its possible inaccuracy, and need to repeat the examination in order to obtain as precise results as possible. The aim of the study was to establish the frequency and reasons of unsatisfactory cervicovaginal smear samples in the course of one year, during which 1594 of 12,242 conventionally obtained cervical samples were sorted out as inadequate. These were reassessed with respect to their adequacy. Eight percent of the smears in which the evaluation of cell abnormality failed due to sample inadequacy were identified and these smears were repeated and analyzed for adequacy and presence of abnormality. The most common reasons included insufficient endocervical epithelial cells, excessive smear thickness, cells obscured with numerous inflammatory elements and erythrocytes, and sample inadequacy due to the presence of foreign material, poor fixation or staining. Inadequate equipment, insufficient material for cytologic analysis, and poor preparation technique may lead to failure to observe abnormality and errors in microscopic analysis. This implies each team member's responsibility for the accuracy of the result as well for the assurance of specimen adequacy. Reduction in the frequency of the reasons mentioned above is possible if internal control, performance quality monitoring and continuing education of each team member are conducted on a regular basis. The necessity to repeat sampling adds to the cost of health care with no considerable increase in the detection rate of epithelial abnormalities, inadequate specimens being the most common cause of false-negative cytologic findings.


Assuntos
Teste de Papanicolaou , Esfregaço Vaginal/normas , Reações Falso-Negativas , Feminino , Humanos , Manejo de Espécimes , Esfregaço Vaginal/estatística & dados numéricos
9.
J Immunol Methods ; 359(1-2): 42-6, 2010 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-20570676

RESUMO

Elucidation of molecular pathways involved in development of human lymphoma requires efficient methods for tackling gene expression in lymph nodes. Expression studies of transcription factors in these malignancies facilitate understanding the changes occurring in neoplastic transformation and lymphoma development. Excised lymph nodes are routinely fixed in formalin and embedded in paraffin for diagnosis and stored in many hospitals' pathology archives. These tissues represent a precious resource for research since they allow retrospective studies to cover a broad range of human lymphoma even the less frequent types. Reverse transcription polymerase chain reaction (RT-PCR) is a commonly used method for gene expression analysis and a reproducible protocol for RNA isolation from lymph nodes is an inevitable requirement for these studies. However, formalin fixation and paraffin-embedding interfere with the quality of RNA especially when isolated from lymph nodes being the most fragile lymphatic tissues. We present here a simple and fast method for RNA isolation from formalin-fixed paraffin-embedded lymph nodes that can be successfully applied for RT-PCR as well as for quantitative RT-PCR analysis. We tested diverse isolation reagents and combined a range of factors in order to get a high quality RNA for retrospective studies of gene expression in human lymphoma samples. Our modified method of RNA extraction from FFPE provides superior yields and purity based on qPCR data.


Assuntos
Formaldeído/química , Perfilação da Expressão Gênica/métodos , Linfonodos/metabolismo , Inclusão em Parafina , RNA Mensageiro/análise , Fixação de Tecidos , Humanos , Linfoma/genética , RNA Mensageiro/genética , RNA Neoplásico/análise , RNA Neoplásico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA