Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
bioRxiv ; 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38328086

RESUMO

Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned hIL-6 transgenic NSG mice (NSG+hIL6) reliably support the engraftment of malignant and pre-malignant human plasma cells including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and post-relapse myeloma, plasma cell leukemia, and AL amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells, developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single cell RNA sequencing showed non-malignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma engrafted mice given CAR T-cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.

3.
JCI Insight ; 9(10)2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38713510

RESUMO

Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.


Assuntos
Modelos Animais de Doenças , Interleucina-6 , Mieloma Múltiplo , Animais , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Humanos , Camundongos , Interleucina-6/metabolismo , Camundongos Transgênicos , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Masculino , Feminino , Plasmócitos/imunologia , Gamopatia Monoclonal de Significância Indeterminada/imunologia , Gamopatia Monoclonal de Significância Indeterminada/patologia
4.
Sci Transl Med ; 14(660): eabo6135, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36044599

RESUMO

T cell receptor (TCR)-based immunotherapy has emerged as a promising therapeutic approach for the treatment of patients with solid cancers. Identifying peptide-human leukocyte antigen (pHLA) complexes highly presented on tumors and rarely expressed on healthy tissue in combination with high-affinity TCRs that when introduced into T cells can redirect T cells to eliminate tumor but not healthy tissue is a key requirement for safe and efficacious TCR-based therapies. To discover promising shared tumor antigens that could be targeted via TCR-based adoptive T cell therapy, we employed population-scale immunopeptidomics using quantitative mass spectrometry across ~1500 tumor and normal tissue samples. We identified an HLA-A*02:01-restricted pan-cancer epitope within the collagen type VI α-3 (COL6A3) gene that is highly presented on tumor stroma across multiple solid cancers due to a tumor-specific alternative splicing event that rarely occurs outside the tumor microenvironment. T cells expressing natural COL6A3-specific TCRs demonstrated only modest activity against cells presenting high copy numbers of COL6A3 pHLAs. One of these TCRs was affinity-enhanced, enabling transduced T cells to specifically eliminate tumors in vivo that expressed similar copy numbers of pHLAs as primary tumor specimens. The enhanced TCR variants exhibited a favorable safety profile with no detectable off-target reactivity, paving the way to initiate clinical trials using COL6A3-specific TCRs to target an array of solid tumors.


Assuntos
Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T , Linfócitos T , Antígenos de Neoplasias , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Imunoterapia Adotiva/métodos , Proteômica , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/uso terapêutico
5.
J Orthop Res ; 33(5): 675-84, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25626998

RESUMO

Type III collagen (Col3) has been proposed to play a key role in tissue repair based upon its temporospatial expression during the healing process of many tissues, including bone. Given our previous finding that Col3 regulates the quality of cutaneous repair, as well as our recent data supporting its role in regulating osteoblast differentiation and trabecular bone quantity, we hypothesized that mice with diminished Col3 expression would exhibit altered long-bone fracture healing. To determine the role of Col3 in bone repair, young adult wild-type (Col3+/+) and haploinsufficent (Col3+/-) mice underwent bilateral tibial fractures. Healing was assessed 7, 14, 21, and 28 days following fracture utilizing microcomputed tomography (microCT), immunohistochemistry, and histomorphometry. MicroCT analysis revealed a small but significant increase in bone volume fraction in Col3+/- mice at day 21. However, histological analysis revealed that Col3+/- mice have less bone within the callus at days 21 and 28, which is consistent with the established role for Col3 in osteogenesis. Finally, a reduction in fracture callus osteoclastic activity in Col3+/- mice suggests Col3 also modulates callus remodeling. Although Col3 haploinsufficiency affected biological aspects of bone repair, it did not affect the regain of mechanical function in the young mice that were evaluated in this study. These findings provide evidence for a modulatory role for Col3 in fracture repair and support further investigations into its role in impaired bone healing.


Assuntos
Regeneração Óssea , Colágeno Tipo III/metabolismo , Consolidação da Fratura , Animais , Calo Ósseo/patologia , Calo Ósseo/fisiologia , Proliferação de Células , Feminino , Camundongos , Osteoclastos/fisiologia , Fraturas da Tíbia/diagnóstico por imagem , Fraturas da Tíbia/patologia , Microtomografia por Raio-X
6.
J Orthop Trauma ; 28 Suppl 1: S20-3, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24378431

RESUMO

OBJECTIVES: Morbidity associated with geriatric fractures may be attributed, in part, to compromised mesenchymal stem cell (MSC) function within the fracture callus. The Notch signaling pathway is important for the healing of nonskeletal tissues in an age-dependent manner, but the effect of Notch on age-dependent fracture healing and MSC dysfunction has not been substantiated. The objective of this study was to examine Notch signaling in MSCs obtained from young and geriatric mice. METHODS: Marrow-derived MSCs were harvested from the femora of 5- and 25-month-old C57BL/6 mice. We assessed in vivo MSC number using CFU-F, proliferation using an Alamar Blue assay, osteoblast differentiation by Alizarin Red S staining, and adipogenic differentiation using Oil Red O staining. Notch receptor and ligand expression was assessed using quantitative PCR, and Notch signaling was assessed by evaluating Notch target gene expression (Hey and HES) under basal conditions and when cells were plated to Jagged-1 ligand. RESULTS: MSC from geriatric mice exhibit reduced MSC number (CFU-F), proliferation, adipogenesis, and inconsistent osteogenesis. The highest expressed Notch receptor is Notch 2, and the highest expressed ligand is Jagged-1, but there were no differences in ligand and receptor gene expression between young and old MSCs. Interestingly, geriatric MSCs show decreased basal Notch signaling activity but are fully responsive to Jagged-1 stimulation. CONCLUSIONS: These data suggest that therapeutic targeting of Notch signaling should be explored in clinical therapies to improve geriatric fracture healing.


Assuntos
Calo Ósseo/metabolismo , Consolidação da Fratura/fisiologia , Células-Tronco Mesenquimais/metabolismo , Receptor Notch2/biossíntese , Fatores Etários , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Contagem de Células , Diferenciação Celular , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Masculino , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serrate-Jagged , Transdução de Sinais
7.
J Orthop Res ; 31(6): 935-43, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23280580

RESUMO

Thrombospondin-2 (TSP2) is a matricellular protein that is highly up-regulated during fracture healing. TSP2 negatively regulates vascularity, vascular reperfusion following ischemia, and cutaneous wound healing. As well, TSP2-null mice show increased endocortical bone formation due to an enhanced number of mesenchymal progenitor cells and show increased cortical thickness. Mice deficient in TSP2 (TSP2-null) show an alteration in fracture healing, that is unrelated to their cortical bone phenotype, which is characterized by enhanced vascularization with a shift towards an intramembranous healing phenotype; thus, we hypothesized that there would be enhanced ischemic fracture healing in the absence of TSP2. We investigated whether an absence of TSP2 would enhance ischemic fracture healing utilizing Laser doppler, µCT and histological analysis. Ischemic tibial fractures were created in wildtype (WT) and TSP2-null mice and harvested 10, 20, or 40 days post-fracture. TSP2-null mice show enhanced vascular perfusion following ischemic fracture. At day 10 post-fracture, TSP2-null mice have 115% greater bone volume than WT mice. This is associated with a 122% increase in vessel density, 20% increase in cell proliferation, and 15% decrease in apoptosis compared to WT. At day 20, TSP2-null mice have 34% more bone volume, 51% greater bone volume fraction, and 37% more bone tissue mineral density than WT. By 40 days after fracture the TSP2-null mice have a 24% increase in bone volume fraction, but other parameters show no significant differences. These findings indicate TSP2 is a negative regulator of ischemic fracture healing and that in the absence of TSP2 bone regeneration is enhanced.


Assuntos
Extremidades/irrigação sanguínea , Consolidação da Fratura , Neovascularização Fisiológica , Fluxo Sanguíneo Regional , Trombospondinas/fisiologia , Animais , Apoptose , Calo Ósseo/irrigação sanguínea , Antígenos CD36/metabolismo , Antígeno CD47/metabolismo , Cartilagem/crescimento & desenvolvimento , Proliferação de Células , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Trombospondina 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA