Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2023 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-37090621

RESUMO

Collagen plays a critical role in regulating breast cancer progression and therapeutic resistance. An improved understanding of both the features and drivers of tumor-permissive and -restrictive collagen matrices are critical to improve prognostication and develop more effective therapeutic strategies. In this study, using a combination of in vitro, in vivo and in silico experiments, we show that type III collagen (Col3) plays a tumor-restrictive role in human breast cancer. We demonstrate that Col3-deficient, human fibroblasts produce tumor-permissive collagen matrices that drive cell proliferation and suppress apoptosis in noninvasive and invasive breast cancer cell lines. In human TNBC biopsy samples, we demonstrate elevated deposition of Col3 relative to type I collagen (Col1) in noninvasive compared to invasive regions. Similarly, in silico analyses of over 1000 breast cancer patient biopsies from The Cancer Genome Atlas BRCA cohort revealed that patients with higher Col3:Col1 bulk tumor expression had improved overall, disease-free and progression-free survival relative to those with higher Col1:Col3 expression. Using an established 3D culture model, we show that Col3 increases spheroid formation and induces formation of lumen-like structures that resemble non-neoplastic mammary acini. Finally, our in vivo study shows co-injection of murine breast cancer cells (4T1) with rhCol3-supplemented hydrogels limits tumor growth and decreases pulmonary metastatic burden compared to controls. Taken together, these data collectively support a tumor-suppressive role for Col3 in human breast cancer and suggest that strategies that increase Col3 may provide a safe and effective modality to limit recurrence in breast cancer patients.

2.
Matrix Biol ; 109: 19-33, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35339637

RESUMO

TGFß is a key regulator of the dynamic reciprocity between cells and the extracellular matrix that drives physiologic and pathologic responses in both tissue repair and tumor microenvironments. Our studies define type III Collagen (Col3) as a suppressor of scar formation and desmoplasia through its effects, in part, on myofibroblasts. TGFß stimulates activation of myofibroblasts, and here, we demonstrate that cultured Col3-deficient fibroblasts have increased TGFß signaling compared to wild-type fibroblasts. Moreover, kinetic binding studies show that a synthetic peptide containing a Col3 cysteine-rich (CR) domain found within its N-propeptide binds in a dose-dependent manner to TGFß1, while a CR control peptide with mutated cysteines does not, suggesting that Col3 attenuates TGFß signaling in part through the N-propeptide CR domain. Consistent with this hypothesis, the CR peptide attenuates TGFß signaling in fibroblasts and 4T1 breast cancer cells and suppresses fibroblast activation and contraction, as assessed by α-smooth-muscle actin staining, cell wrinkling of deformable silicone, and stressed-fibroblast populated collagen lattice contraction assays. Finally, CR peptide treatment of orthotopically injected breast cancer cells (4T1) suppresses intratumoral fibroblast activation and inhibits primary tumor growth compared to CR control. Treatment with the CR peptide decreases both intratumoral canonical and non-canonical downstream TGFß signaling targets, consistent with its extracellular binding to TGFß. Taken together, our results suggest that the Col3 N-propeptide CR domain binds TGFß1 and attenuates (but importantly does not eliminate) TGFß signaling in fibroblasts and cancer cells. Expanding on our previous work, this study demonstrates an additional mechanism by which Col3 regulates cell behaviors in post-injury and tumor microenvironments and suggests that novel Col3-targeted strategies could effectively control biologic responses in vivo and improve anti-scarring/fibrosis and oncologic therapies.


Assuntos
Neoplasias da Mama , Colágeno Tipo III , Actinas/metabolismo , Neoplasias da Mama/metabolismo , Células Cultivadas , Cicatriz/metabolismo , Colágeno/metabolismo , Colágeno Tipo III/metabolismo , Cisteína , Feminino , Fibroblastos/metabolismo , Humanos , Miofibroblastos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral
3.
Can Vet J ; 62(8): 872-876, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34341603

RESUMO

An 8-month-old spayed female Labrador retriever dog was evaluated for regurgitation 6 months after surgery for a suspected vascular ring anomaly. The dog had a history of regurgitation and slow development as a puppy. An initial left-sided exploratory thoracotomy was unsuccessful in identifying and treating a vascular ring anomaly. The dog was subsequently presented to the PennVet Emergency Service for regurgitation. Thoracic radiography showed cranial thoracic esophageal dilation and an esophageal foreign body that was then removed endoscopically. Subsequent computed tomographic (CT) angiography revealed a double aortic arch. A left 4th intercostal space thoracotomy was performed. The smaller left aortic arch and a left ligamentum arteriosum were ligated and transected. The dog recovered uneventfully and was healthy at the 1-month follow-up visit. This is the 5th reported successful surgical correction of a double aortic arch in a dog. Computed tomographic angiography was essential in diagnosis and surgical planning. Key clinical message: Although uncommon, double aortic arches can occur and present a diagnostic and surgical challenge when a persistent right aortic arch is suspected. Computed tomographic angiography provides an accurate preoperative diagnosis and allows for surgical planning.


Traitement chirurgical d'un double arc aortique chez un chien. Une chienne Labrador retriever femelle stérilisée âgée de 8 mois a été évaluée pour régurgitation 6 mois après une chirurgie pour une anomalie suspectée de l'anneau vasculaire. Le chien avait des antécédents de régurgitation et de développement lent en tant que chiot. Une première thoracotomie exploratrice gauche n'a pas permis d'identifier et de traiter une anomalie de l'anneau vasculaire. Le chien a ensuite été présenté au service d'urgence PennVet pour régurgitation. La radiographie thoracique a montré une dilatation de l'oesophage thoracique crânien et un corps étranger oesophagien qui a ensuite été retiré par endoscopie. L'angiographie tomodensitométrique (TDM) subséquente a révélé un double arc aortique. Une thoracotomie du 4e espace intercostal gauche a été réalisée. Le plus petit arc aortique gauche et un ligament artériel gauche ont été ligaturés et sectionnés. Le chien s'est rétabli sans incident et était en bonne santé lors de la visite de suivi à 1 mois. Il s'agit de la cinquième correction chirurgicale réussie d'un double arc aortique chez un chien. L'angiographie tomodensitométrique était essentielle dans le diagnostic et la planification chirurgicale.Message clinique clé :Bien que rares, des arcs aortiques doubles peuvent survenir et présenter un défi diagnostique et chirurgical lorsqu'un arc aortique droit persistant est suspecté. L'angiographie tomodensitométrique fournit un diagnostic préopératoire précis et permet une planification chirurgicale.(Traduit par Dr Serge Messier).


Assuntos
Doenças do Cão , Anel Vascular , Angiografia , Animais , Aorta Torácica/diagnóstico por imagem , Aorta Torácica/cirurgia , Doenças do Cão/diagnóstico por imagem , Doenças do Cão/cirurgia , Cães , Esôfago , Feminino , Toracotomia/veterinária , Anel Vascular/veterinária
4.
Sci Rep ; 11(1): 8659, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33883562

RESUMO

In spite of major advances over the past several decades in diagnosis and treatment, breast cancer remains a global cause of morbidity and premature death for both human and veterinary patients. Due to multiple shared clinicopathological features, dogs provide an excellent model of human breast cancer, thus, a comparative oncology approach may advance our understanding of breast cancer biology and improve patient outcomes. Despite an increasing awareness of the critical role of fibrillar collagens in breast cancer biology, tumor-permissive collagen features are still ill-defined. Here, we characterize the molecular and morphological phenotypes of type I collagen in canine mammary gland tumors. Canine mammary carcinoma samples contained longer collagen fibers as well as a greater population of wider fibers compared to non-neoplastic and adenoma samples. Furthermore, the total number of collagen cross-links enriched in the stable hydroxylysine-aldehyde derived cross-links was significantly increased in neoplastic mammary gland samples compared to non-neoplastic mammary gland tissue. The mass spectrometric analyses of type I collagen revealed that in malignant mammary tumor samples, lysine residues, in particular those in the telopeptides, were markedly over-hydroxylated in comparison to non-neoplastic mammary tissue. The extent of glycosylation of hydroxylysine residues was comparable among the groups. Consistent with these data, expression levels of genes encoding lysyl hydroxylase 2 (LH2) and its molecular chaperone FK506-binding protein 65 were both significantly increased in neoplastic samples. These alterations likely lead to an increase in the LH2-mediated stable collagen cross-links in mammary carcinoma that may promote tumor cell metastasis in these patients.


Assuntos
Colágeno/metabolismo , Doenças do Cão/metabolismo , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/metabolismo , Aminoácidos/metabolismo , Animais , Colágeno Tipo I/metabolismo , Doenças do Cão/patologia , Cães , Feminino , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/patologia , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real
5.
Sci Rep ; 11(1): 5950, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723274

RESUMO

Cells interpret cues from and interact with fibrous microenvironments through the body based on the mechanics and organization of these environments and the phenotypic state of the cell. This in turn regulates mechanoactive pathways, such as the localization of mechanosensitive factors. Here, we leverage the microscale heterogeneity inherent to engineered fiber microenvironments to produce a large morphologic data set, across multiple cells types, while simultaneously measuring mechanobiological response (YAP/TAZ nuclear localization) at the single cell level. This dataset describing a large dynamic range of cell morphologies and responses was coupled with a machine learning approach to predict the mechanobiological state of individual cells from multiple lineages. We also noted that certain cells (e.g., invasive cancer cells) or biochemical perturbations (e.g., modulating contractility) can limit the predictability of cells in a universal context. Leveraging this finding, we developed further models that incorporate biochemical cues for single cell prediction or identify individual cells that do not follow the established rules. The models developed here provide a tool for connecting cell morphology and signaling, incorporating biochemical cues in predictive models, and identifying aberrant cell behavior at the single cell level.


Assuntos
Fenômenos Fisiológicos Celulares , Células/citologia , Microambiente Celular , Mecanotransdução Celular , Modelos Biológicos , Algoritmos , Animais , Linhagem Celular Tumoral , Núcleo Celular , Células/patologia , Fibroblastos , Humanos , Camundongos , Redes Neurais de Computação , Microambiente Tumoral
6.
Biomaterials ; 269: 120667, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33450585

RESUMO

Aberrant extracellular matrix (ECM) assembly surrounding implanted biomaterials is the hallmark of the foreign body response, in which implants become encapsulated in thick fibrous tissue that prevents their proper function. While macrophages are known regulators of fibroblast behavior, how their phenotype influences ECM assembly and the progression of the foreign body response is poorly understood. In this study, we used in vitro models with physiologically relevant macrophage phenotypes, as well as controlled release of macrophage-modulating cytokines from gelatin hydrogels implanted subcutaneously in vivo to investigate the role of macrophages in ECM assembly. Primary human macrophages were polarized to four distinct phenotypes, which have each been associated with fibrosis, including pro-inflammatory M1, pro-healing M2, and a hybrid M1/M2, generated by exposing macrophages to M1-and M2-promoting stimuli simultaneously. Additionally, macrophages were first polarized to M1 and then to M2 (M1→M2) to generate a phenotype typically observed during normal wound healing. Human dermal fibroblasts that were cultured in macrophage-conditioned media upregulated numerous genes involved in regulation of ECM assembly, especially in M2-conditioned media. Hybrid M1/M2 macrophage-conditioned media caused fibroblasts to produce a matrix with thicker and less aligned fibers, while M2 macrophage-conditioned media caused the formation of a more aligned matrix with thinner fibers. Gelatin methacrylate hydrogels containing interleukin-4 (IL4) and IL13-loaded poly(lactic-co-glycolic acid) (PLGA) microparticles were designed to promote the M2 phenotype in a murine subcutaneous in vivo model. NanoString multiplex gene expression analysis of hydrogel explants showed that hydrogels without cytokines caused mostly M1 phenotype markers to be highly expressed at an early time point (3 days), but the release of IL4+IL13 promoted upregulation of M2 markers and genes associated with regulation of ECM assembly, such as Col5a1 and Col6a1. Biochemical analysis and second harmonic generation microscopy showed that the release of IL4+IL13 increased total sulfated glycosaminoglycan content and decreased fibril alignment, which is typically associated with less fibrotic tissue. Together, these results show that hybrid M1/M2 macrophages regulate ECM assembly, and that shifting the balance towards M2 may promote architectural and compositional changes in ECM with enhanced potential for downstream remodeling.


Assuntos
Hidrogéis , Macrófagos , Animais , Materiais Biocompatíveis , Citocinas , Matriz Extracelular , Humanos , Camundongos , Fenótipo
7.
PLoS One ; 15(8): e0236516, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32776970

RESUMO

Breast cancer is the most common cause of cancer-related deaths in women worldwide. Identification of reliable prognostic indicators and therapeutic targets is critical for improving patient outcome. Cancer in companion animals often strongly resembles human cancers and a comparative approach to identify prognostic markers can improve clinical care across species. Feline mammary tumors (FMT) serve as models for extremely aggressive triple negative breast cancer (TNBC) in humans, with high rates of local and distant recurrence after resection. Despite the aggressive clinical behavior of most FMT, current prognostic indicators are insufficient for accurately predicting outcome, similar to human patients. Given significant heterogeneity of mammary tumors, there has been a recent focus on identification of universal tumor-permissive stromal features that can predict biologic behavior and provide therapeutic targets to improve outcome. As in human and canine patients, collagen signatures appear to play a key role in directing mammary tumor behavior in feline patients. We find that patients bearing FMTs with denser collagen, as well as longer, thicker and straighter fibers and less identifiable tumor-stromal boundaries had poorer outcomes, independent of the clinical variables grade and surgical margins. Most importantly, including the collagen parameters increased the predictive power of the clinical model. Thus, our data suggest that similarities with respect to the stromal microenvironment between species may allow this model to predict outcome and develop novel therapeutic targets within the tumor stroma that would benefit both veterinary and human patients with aggressive mammary tumors.


Assuntos
Colágeno/metabolismo , Neoplasias Mamárias Animais/cirurgia , Prognóstico , Neoplasias de Mama Triplo Negativas/cirurgia , Animais , Gatos , Colágeno/genética , Modelos Animais de Doenças , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/cirurgia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral/genética
8.
Cancer Cell ; 35(1): 33-45.e6, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30645975

RESUMO

Tumor-derived extracellular vesicles (TEV) "educate" healthy cells to promote metastases. We found that melanoma TEV downregulated type I interferon (IFN) receptor and expression of IFN-inducible cholesterol 25-hydroxylase (CH25H). CH25H produces 25-hydroxycholesterol, which inhibited TEV uptake. Low CH25H levels in leukocytes from melanoma patients correlated with poor prognosis. Mice incapable of downregulating the IFN receptor and Ch25h were resistant to TEV uptake, TEV-induced pre-metastatic niche, and melanoma lung metastases; however, ablation of Ch25h reversed these phenotypes. An anti-hypertensive drug, reserpine, suppressed TEV uptake and disrupted TEV-induced formation of the pre-metastatic niche and melanoma lung metastases. These results suggest the importance of CH25H in defense against education of normal cells by TEV and argue for the use of reserpine in adjuvant melanoma therapy.


Assuntos
Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma/patologia , Receptor de Interferon alfa e beta/metabolismo , Esteroide Hidroxilases/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Humanos , Interferons/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Melanoma/metabolismo , Camundongos , Metástase Neoplásica , Oxisteróis/metabolismo , Reserpina/administração & dosagem , Reserpina/farmacologia , Esteroide Hidroxilases/genética , Células THP-1
9.
Vet Pathol ; 55(5): 622-633, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29788797

RESUMO

The epithelial-mesenchymal transition (EMT) is a dynamic process linked to metastasis in many tumor types, including mammary tumors. In this study, we evaluated E-cadherin and vimentin immunolocalization in primary canine mammary carcinomas (20 cases) and their respective metastases, as well as their relationship with the core regulators SNAIL/SLUG. To assess the number of cells undergoing the process of EMT, we quantitated double-positive (E-cadherin+/vimentin+) cells using immunofluorescence, via cell counting and image analysis. In addition, SNAIL/SLUG expression was evaluated by established immunohistochemical methods. Primary tumors had significantly more E-cadherin+/vimentin+ co-expression than their paired respective lymph node or distant metastasis, respectively. Furthermore, the percentage of E-cadherin+/vimentin+ cells in grade II and III carcinomas was significantly higher than in grade I tumors. Primary tumors had significantly higher SNAIL/SLUG expression when analyzed based on the percentage of positive cells compared with their respective distant metastases in pairwise comparisons. An inverse correlation was noted between SNAIL/SLUG immunoreactivity and percentage of E-cadherin+/vimentin+ immunopositive cells in primary tumor samples when SNAIL/SLUG immunoreactivity was grouped into 2 categories (high versus low) based on percentage-positive staining. These results show a positive correlation between E-cadherin+/vimentin+ cells and higher tumor grade, establish differences between primary tumor and their respective metastases, and provide further support that EMT plays a critical role in the metastasis of canine mammary carcinoma. Furthermore, these data suggest that modulation of this process could provide greater therapeutic control and provide support for further research to determine if E-cadherin+/vimentin+ co-immunoreactivity imparts predictive value in the clinical outcome of patients with canine mammary carcinomas.


Assuntos
Carcinoma/veterinária , Doenças do Cão/patologia , Transição Epitelial-Mesenquimal , Neoplasias Mamárias Animais/patologia , Animais , Caderinas/metabolismo , Carcinoma/patologia , Cães , Feminino , Imunofluorescência/veterinária , Glândulas Mamárias Animais/patologia , Vimentina/metabolismo
10.
PLoS One ; 12(7): e0180448, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28683102

RESUMO

Increasing evidence indicates that the tumor microenvironment plays a critical role in regulating the biologic behavior of breast cancer. In veterinary oncology, there is a need for improved prognostic markers to accurately identify dogs at risk for local and distant (metastatic) recurrence of mammary gland carcinoma and therefore would benefit from adjuvant therapy. Collagen density and fiber organization have been shown to regulate tumor progression in both mouse and human mammary tumors, with certain collagen signatures predicting poor outcomes in women with breast cancer. We hypothesized that collagen signatures in canine mammary tumor biopsies can serve as prognostic biomarkers and potential targets for treatment. We used second harmonic generation imaging to evaluate fibrillar collagen density, the presence of a tumor-stromal boundary, tumor associated collagen signatures (TACS) and individual collagen fiber characteristics (width, length and straightness) in grade I/II and grade III canine mammary tumors. Collagen density, as well as fiber width, length and straightness, were inversely correlated with patient overall survival time. Notably, grade III cases were less likely to have a tumor-stromal boundary and the lack of a boundary predicted poor outcome. Importantly, a lack of a defined tumor-stromal boundary and an increased collagen fiber width were associated with decreased survival even when tumor grade, patient stage, ovariohysterectomy status at the time of mammary tumor excision, and histologic evidence of lymphovascular invasion were considered in a multivariable model, indicating that these parameters could augment current methods to identify patients at high risk for local or metastatic progression/recurrence. Furthermore, these data, which identify for the first time, prognostic collagen biomarkers in naturally occurring mammary gland neoplasia in the dog, support the use of the dog as a translational model for tumor-stromal interactions in breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Colágeno/metabolismo , Glândulas Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/diagnóstico por imagem , Microambiente Tumoral , Animais , Biópsia , Colágeno/ultraestrutura , Progressão da Doença , Cães , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Feminino , Metástase Linfática , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/cirurgia , Neoplasias Mamárias Animais/mortalidade , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/cirurgia , Microscopia de Fluorescência por Excitação Multifotônica , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Análise de Sobrevida , Resultado do Tratamento
11.
J Invest Dermatol ; 137(3): 737-747, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27856290

RESUMO

Electrical stimulation (ES) is known to promote cutaneous healing; however, its ability to regulate reinnervation remains unclear. First, we show that ES treatment of human acute cutaneous wounds (n = 40) increased reinnervation. Next, to define neurophysiologic mechanisms through which ES affects repair, microarray analysis of wound biopsy samples was performed on days 3, 7, 10, and 14 after wounding. This identified neural differentiation biomarkers TUBB3 (melanocyte development and neuronal marker) and its upstream molecule FIG4 (phosphatidylinositol (3,5)-bisphosphate 5-phosphatase) as significantly up-regulated after ES treatment. To demonstrate a functional ES-TUBB3 axis in cutaneous healing, we showed increased TUBB3+ melanocytes and melanogenesis plus FIG4 and nerve growth factor expression, suggesting higher cellular differentiation. In support of this role of ES to regulate neural crest-derived cell fate and differentiation in vivo, knockdown of FIG4 in neuroblastoma cells resulted in vacuologenesis and cell degeneration, whereas ES treatment after FIG4-small interfering RNA transfection enhanced neural differentiation, survival, and integrity. Further characterization showed increased TUBB3+ and protein gene product 9.5+ Merkel cells during in vivo repair, after ES. We demonstrate that ES contributes to increased expression of neural differentiation biomarkers, reinnervation, and expansion of melanocyte and Merkel cell pool during repair. Targeted ES-assisted acceleration of healing has significant clinical implications.


Assuntos
Biomarcadores/metabolismo , Estimulação Elétrica , Pele/inervação , Pele/metabolismo , Cicatrização/fisiologia , Biópsia , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Flavoproteínas/metabolismo , Humanos , Melanócitos/metabolismo , Células de Merkel/metabolismo , Fator de Crescimento Neural/metabolismo , Crista Neural/metabolismo , Neurofisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Tubulina (Proteína)/metabolismo
12.
J Invest Dermatol ; 136(2): 487-496, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26802241

RESUMO

Breaches in the skin barrier initiate an inflammatory immune response that is critical for successful wound healing. Innate lymphoid cells (ILCs) are a recently identified population of immune cells that reside at epithelial barrier surfaces such as the skin, lung, and gut, and promote proinflammatory or epithelial repair functions after exposure to allergens, pathogens, or chemical irritants. However, the potential role of ILCs in regulating cutaneous wound healing remains undefined. Here, we demonstrate that cutaneous injury promotes an IL-33-dependent group 2 ILC (ILC2) response and that abrogation of this response impairs re-epithelialization and efficient wound closure. In addition, we provide evidence suggesting that an analogous ILC2 response is operational in acute wounds of human skin. Together, these results indicate that IL-33-responsive ILC2s are an important link between the cutaneous epithelium and the immune system, acting to promote the restoration of skin integrity after injury.


Assuntos
Imunidade Inata/fisiologia , Interleucina-33/imunologia , Pele/lesões , Cicatrização/imunologia , Ferimentos e Lesões/imunologia , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Humanos , Imunidade Inata/imunologia , Imuno-Histoquímica , Interleucina-33/metabolismo , Linfócitos/citologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pele/patologia , Cicatrização/fisiologia , Ferimentos e Lesões/patologia
13.
Vet Microbiol ; 182: 102-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26711035

RESUMO

Ebolaviruses and marburgviruses belong to the Filoviridae family and often cause severe, fatal hemorrhagic fever in humans and non-human primates. The magnitude of the 2014 outbreak in West Africa and the unprecedented emergence of Ebola virus disease (EVD) in the United States underscore the urgency to better understand the dynamics of Ebola virus infection, transmission and spread. To date, the susceptibility and possible role of domestic animals and pets in the transmission cycle and spread of EVD remains unclear. We utilized infectious VSV recombinants and lentivirus pseudotypes expressing the EBOV surface glycoprotein (GP) to assess the permissiveness of canine and feline cells to EBOV GP-mediated entry. We observed a general restriction in EBOV-mediated infection of primary canine and feline cells. To address the entry mechanism, we used cells deficient in NPC1, a host protein implicated in EBOV entry, and a pharmacological blockade of cholesterol transport, to show that an NPC1-dependent mechanism of EBOV entry is conserved in canine and feline cells. These data demonstrate that cells of canine and feline origin are susceptible to EBOV GP mediated infection; however, infectivity of these cells is reduced significantly compared to controls. Moreover, these data provide new insights into the mechanism of EBOV GP mediated entry into cells of canine and feline origin.


Assuntos
Ebolavirus/fisiologia , Fibroblastos/virologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Gatos , Células Cultivadas , Cães , Ebolavirus/classificação , Fibroblastos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Especificidade da Espécie , Proteínas Virais de Fusão , Internalização do Vírus
14.
J Invest Dermatol ; 135(10): 2519-2529, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26358387

RESUMO

Currently available skin grafts and skin substitutes for healing following third-degree burn injuries are fraught with complications, often resulting in long-term physical and psychological sequelae. Synthetic treatment that can promote wound healing in a regenerative manner would provide an off-the-shelf, non-immunogenic strategy to improve clinical care of severe burn wounds. Here, we demonstrate the vulnerary efficacy and accelerated healing mechanism of a dextran-based hydrogel in a third-degree porcine burn model. The model was optimized to allow examination of the hydrogel treatment for clinical translation and its regenerative response mechanisms. Hydrogel treatment accelerated third-degree burn wound healing by rapid wound closure, improved re-epithelialization, enhanced extracellular matrix remodeling, and greater nerve reinnervation, compared with the dressing-treated group. These effects appear to be mediated through the ability of the hydrogel to facilitate a rapid but brief initial inflammatory response that coherently stimulates neovascularization within the granulation tissue during the first week of treatment, followed by an efficient vascular regression to promote a regenerative healing process. Our results suggest that the dextran-based hydrogels may substantially improve healing quality and reduce skin grafting incidents and thus pave the way for clinical studies to improve the care of severe burn injury patients.


Assuntos
Queimaduras/terapia , Hidrogéis/farmacologia , Pele Artificial , Cicatrização/efeitos dos fármacos , Animais , Biópsia por Agulha , Queimaduras/patologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Distribuição Aleatória , Regeneração/efeitos dos fármacos , Sensibilidade e Especificidade , Higiene da Pele/métodos , Transplante de Pele/métodos , Sus scrofa , Suínos
15.
Am J Pathol ; 185(5): 1471-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25795282

RESUMO

Breast cancer metastasis is the leading cause of cancer-related deaths in women worldwide. Collagen in the tumor microenvironment plays a crucial role in regulating tumor progression. We have shown that type III collagen (Col3), a component of tumor stroma, regulates myofibroblast differentiation and scar formation after cutaneous injury. During the course of these wound-healing studies, we noted that tumors developed at a higher frequency in Col3(+/-) mice compared to wild-type littermate controls. We, therefore, examined the effect of Col3 deficiency on tumor behavior, using the murine mammary carcinoma cell line 4T1. Notably, tumor volume and pulmonary metastatic burden after orthotopic injection of 4T1 cells were increased in Col3(+/-) mice compared to Col3(+/+) littermates. By using murine (4T1) and human (MDA-MB-231) breast cancer cells grown in Col3-poor and Col3-enriched microenvironments in vitro, we found that several major events of the metastatic process were suppressed by Col3, including adhesion, invasion, and migration. In addition, Col3 deficiency increased proliferation and decreased apoptosis of 4T1 cells both in vitro and in primary tumors in vivo. Mechanistically, Col3 suppresses the procarcinogenic microenvironment by regulating stromal organization, including density and alignment of fibrillar collagen and myofibroblasts. We propose that Col3 plays an important role in the tumor microenvironment by suppressing metastasis-promoting characteristics of the tumor-associated stroma.


Assuntos
Colágeno Tipo III/metabolismo , Neoplasias Mamárias Experimentais/patologia , Invasividade Neoplásica/patologia , Microambiente Tumoral/fisiologia , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
16.
J Invest Dermatol ; 135(4): 1166-1174, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25431847

RESUMO

Cutaneous wounds establish endogenous "wound current" upon injury until re-epithelialization is complete. Keratinocyte proliferation, regulated partly by p53, is required for epidermal closure. SIVA1 promotes human double minute 2 homolog (HDM2)-mediated p53 regulation. However, the role of SIVA1 in wound healing is obscure. Here, we report that electrical stimulation (ES) accelerates wound healing by upregulating SIVA1 and its subsequent ability to modulate p53 activities. Cultured donut-shaped human skin explants, subjected to ES, exhibited better epidermal stratification, increased proliferation, and upregulation of gene and protein expression of HDM2/SIVA1, compared with non-ES-treated explants. ES significantly increased in vitro keratinocyte proliferation and phospho-p53-SIVA1 interaction; however, this showed stable expression of phospho-p53, which increased significantly in the absence of SIVA1. Here, HDM2 alone was unable to downregulate nuclear-accumulated phospho-p53, which was evident from decreased proliferation and increased sub-G1 population seen by flow cytometry. Further examination of the epidermis of human cutaneous wounds showed higher p53-SIVA1 coexpression and proliferation 7 days after injury in ES-treated wounds compared with control wounds. In summary, ES-inducible SIVA1 modulates p53 activities in proliferating keratinocytes, and exogenous ES affects p53/HDM2/SIVA1 axis leading to increased proliferation during re-epithelialization. This highlights ES as a potential strategy for enhancing cutaneous repair.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Estimulação Elétrica , Proteína Supressora de Tumor p53/metabolismo , Cicatrização/fisiologia , Ciclo Celular , Divisão Celular , Proliferação de Células , Epiderme/metabolismo , Citometria de Fluxo , Fase G1 , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Queratinócitos/citologia , Técnicas de Cultura de Órgãos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Pele/metabolismo , Pele/patologia , Dermatopatias/metabolismo , Regulação para Cima
17.
Wound Repair Regen ; 21(3): 382-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23627495

RESUMO

Veterinarians and veterinary medicine have been integral to the development of stem cell therapies. The contributions of large animal experimental models to the development and refinement of modern hematopoietic stem cell transplantation were noted nearly five decades ago. More recent advances in adult stem cell/regenerative cell therapies continue to expand knowledge of the basic biology and clinical applications of stem cells. A relatively liberal legal and ethical regulation of stem cell research in veterinary medicine has facilitated the development and in some instances clinical translation of a variety of cell-based therapies involving hematopoietic stem cells and mesenchymal stem cells, as well as other adult regenerative cells and recently embryonic stem cells and induced pluripotent stem cells. In fact, many of the pioneering developments in these fields of stem cell research have been achieved through collaborations of veterinary and human scientists. This review aims to provide an overview of the contribution of large animal veterinary models in advancing stem cell therapies for both human and clinical veterinary applications. Moreover, in the context of the "One Health Initiative," the role veterinary patients may play in the future evolution of stem cell therapies for both human and animal patients will be explored.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Medicina Regenerativa/métodos , Pesquisa com Células-Tronco , Transplante de Células-Tronco , Pesquisa Translacional Biomédica/métodos , Ferimentos e Lesões/terapia , Animais , Humanos
18.
Cell Transplant ; 21(10): 2189-200, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22472645

RESUMO

Clinical trials utilizing bone marrow-derived mesenchymal stem cell (BM-MSC) therapies show promise for treating a variety of pathologic conditions. Paramount to optimization of such cell-based therapies is a thorough understanding of MSC biology. Despite the tremendous potential that exists for the clinical use of canine BM-MSCs in veterinary medicine, as well as in preclinical studies for human medicine, relatively little information exists regarding basic biological properties of the cells. In this study, we compared the importance of donor characteristics (age and harvest site) and ex vivo expansion on canine BM-MSC frequency (CFU-f) and differentiation potential. Advancing age was found to have a negative effect on CFU-f as well as osteogenic potential. Site of harvest was also found to have significant effects on MSC properties. MSCs obtained from the humerus were found at the lowest frequency and were least osteogenic compared to those harvested from the tibia, femur, and ilium. Osteogenic potential diminished significantly by the third passage. These results suggest important donor parameters and culture effects to consider in translational studies examining MSC-based regenerative medical strategies.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Transplante de Células-Tronco Mesenquimais/veterinária , Células-Tronco Mesenquimais/fisiologia , Doadores de Tecidos , Fatores Etários , Animais , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Cães , Humanos , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa/métodos , Engenharia Tecidual
19.
Biol Blood Marrow Transplant ; 15(3): 293-305, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19203720

RESUMO

In the murine model, in utero hematopoietic cell transplantation (IUHCT) has been shown to achieve low levels of allogeneic chimerism and associated donor-specific tolerance permitting minimal conditioning postnatal hematopoietic stem cell transplantation (HSCT). In this pilot study, we investigated IUHCT in the canine leukocyte adhesion deficiency (CLAD) model. Haploidentical IUHCT resulted in stable low-level donor cell chimerism in all dogs that could be analyzed by sensitive detection methodology (4 of 10) through 18 months of follow-up. In the 2 CLAD recipients, low-level chimerism resulted in amelioration and complete reversal of the CLAD phenotype, respectively. Six recipients of IUHCT (5 carriers and 1 CLAD) subsequently received postnatal HSCT from the same haploidentical prenatal donor after minimal conditioning with busulfan 10 mg/kg. Chimerism in 2 of 5 CLAD carriers that underwent HSCT increased from < 1% pre-HSCT to sustained levels of 35% to 45%. Control animals undergoing postnatal haploidentical HSCT without IUHCT had no detectable donor chimerism. These results demonstrate that haploidentical IUHCT in the CLAD model can result in low-level donor chimerism that can prevent the lethal phenotype in CLAD dogs, and can result in donor-specific tolerance that can facilitate postnatal minimal conditioning HSCT.


Assuntos
Terapias Fetais/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Síndrome da Aderência Leucocítica Deficitária/terapia , Animais , Bussulfano/administração & dosagem , Inibidores da Dipeptidil Peptidase IV , Modelos Animais de Doenças , Cães , Feminino , Doença Enxerto-Hospedeiro/imunologia , Haploidia , Tolerância Imunológica , Imunossupressores/administração & dosagem , Síndrome da Aderência Leucocítica Deficitária/imunologia , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Gravidez , Quimeras de Transplante , Condicionamento Pré-Transplante/métodos
20.
Wound Repair Regen ; 15(5): 736-47, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17971020

RESUMO

Chronic wounds create a formidable clinical problem resulting in considerable morbidity and healthcare expenditure. The etiology for wound healing impairment appears to be multifactorial; however, ischemia is a common factor in most types of chronic wounds. Ideal therapy for such wounds would be to correct deficiencies in growth factors and matrix components and provide cellular precursors required for timely wound closure. We hypothesized that stromal progenitor cell (SPC) therapy could correct the ischemic wound-healing defect through both direct and indirect mechanisms. To test this hypothesis, we used the ischemic rabbit ear model of chronic wound healing. We found that treatment of the wounds with SPCs was able to reverse the ischemic wound-healing impairment, with improved granulation tissue formation and reepithelialization compared with vehicle or bone marrow mononuclear cell controls. In vitro, SPCs were found to produce factors involved in angiogenesis and reepithelialization, and extracellular matrix components, providing evidence for both direct and indirect mechanisms for the observed correction of the healing impairment in these wounds. Treatment of ischemic wounds with SPCs can dramatically improve wound healing and provides a rationale for further studies focused on SPCs as a potential cellular therapy in impaired wound healing.


Assuntos
Isquemia/terapia , Transplante de Células-Tronco Mesenquimais , Cicatrização/fisiologia , Animais , Ensaios de Migração Celular , Primers do DNA , Modelos Animais de Doenças , Orelha Externa/irrigação sanguínea , Isquemia/fisiopatologia , Queratinócitos , Masculino , Neovascularização Fisiológica/fisiologia , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA