Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 39(38): 6129-6137, 2020 09.
Article in English | MEDLINE | ID: mdl-32807917

ABSTRACT

Activation of cancer-associated fibroblasts (CAFs) and ensuing desmoplasia play an important role in the growth and progression of solid tumors. Here we demonstrate that, within colon and pancreatic ductal adenocarcinoma tumors, efficient stromagenesis relies on downregulation of the IFNAR1 chain of the type I interferon (IFN1) receptor. Expression of the fibroblast activation protein (FAP) and accumulation of the extracellular matrix (ECM) was notably impaired in tumors grown in the Ifnar1S526A (SA) knock-in mice, which are deficient in IFNAR1 downregulation. Primary fibroblasts from these mice exhibited elevated levels of Smad7, a negative regulator of the transforming growth factor-ß (TGFß) pathway. Knockdown of Smad7 alleviated deficient ECM production in SA fibroblasts in response to TGFß. Analysis of human colorectal cancers revealed an inverse correlation between IFNAR1 and FAP levels. Whereas growth of tumors in SA mice was stimulated by co-injection of wild type but not SA fibroblasts, genetic ablation of IFNAR1 in fibroblasts also accelerated tumor growth. We discuss how inactivation of IFNAR1 in CAFs acts to stimulate stromagenesis and tumor growth.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Receptor, Interferon alpha-beta/genetics , Tumor Microenvironment , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Humans , Immunohistochemistry , Interferon Type I/metabolism , Mice , Neoplasms/pathology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Tumor Microenvironment/genetics
2.
Am J Pathol ; 190(5): 1118-1136, 2020 05.
Article in English | MEDLINE | ID: mdl-32084369

ABSTRACT

Fibroblast activation protein (FAP) has been established as an inducible and mesenchymal cell-specific mediator of disease progression in cancer and fibrosis. Atherosclerosis is a fibroinflammatory disease, and FAP was previously reported to be up-regulated in human atherosclerotic plaques compared with normal vessel. We investigated the spatial and temporal distribution of Fap-expressing cells in a murine model of atherosclerosis and used a genetic approach to determine if and how Fap affected disease progression. Fap was found to be expressed predominantly on vascular smooth muscle cells in lesions of athero-prone Apoe-/- mice. Global deletion of Fap (Fap-/-) in Apoe-/- mice accelerated atherosclerotic disease progression in both males and females, with the effect observed earlier in males. Sex-specific effects on lesion morphology were observed. Relative levels of extracellular matrix, fibrotic, and inflammatory cell content were comparable in lesions in male mice regardless of Fap status. In contrast, lesions in Fap-/- female mice were characterized by a more fibrotic composition due to a reduction in inflammation, specifically a reduction in Mox macrophages. Combined, these data suggest that Fap restrains the progression of atherosclerosis and may contribute to the sexually dimorphic susceptibility to atherosclerosis by regulating the balance between inflammation (an indicator of vulnerability to plaque rupture) and fibrosis (an indicator of plaque stability).


Subject(s)
Atherosclerosis/metabolism , Fibrosis/metabolism , Gelatinases/metabolism , Inflammation/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Sex Characteristics , Animals , Apolipoproteins E/deficiency , Endopeptidases , Female , Male , Mice , Mice, Knockout, ApoE
3.
Matrix Biol ; 75-76: 314-330, 2019 01.
Article in English | MEDLINE | ID: mdl-29894820

ABSTRACT

Cutaneous wound healing consists of three main phases: inflammation, re-epithelialization, and tissue remodeling. During normal wound healing, these processes are tightly regulated to allow restoration of skin function and biomechanics. In many instances, healing leads to an excess accumulation of fibrillar collagen (the principal protein found in the extracellular matrix - ECM), and the formation of scar tissue, which has compromised biomechanics, tested using ramp to failure tests, compared to normal skin (Corr and Hart, 2013 [1]). Alterations in collagen accumulation and architecture have been attributed to the reduced tensile strength found in scar tissue (Brenda et al., 1999; Eleswarapu et al., 2011). Defining mechanisms that govern cellular functionality and ECM remodeling are vital to understanding normal versus pathological healing and developing approaches to prevent scarring. CD44 is a cell surface adhesion receptor expressed on nearly all cell types present in dermis. Although CD44 has been implicated in an array of inflammatory and fibrotic processes such as leukocyte recruitment, T-cell extravasation, and hyaluronic acid (the principal glycosaminoglycan found in the ECM) metabolism, the role of CD44 in cutaneous wound healing and scarring remains unknown. We demonstrate that in an excisional biopsy punch wound healing model, CD44-null mice have increased inflammatory and reduced fibrogenic responses during early phases of wound healing. At wound closure, CD44-null mice exhibit reduced collagen degradation leading to increased accumulation of fibrillar collagen, which persists after wound closure leading to reduced tensile strength resulting in a more severe scarring phenotype compared to WT mice. These data indicate that CD44 plays a previously unknown role in fibrillar collagen accumulation and wound healing during the injury response.


Subject(s)
Extracellular Matrix/genetics , Hyaluronan Receptors/genetics , Inflammation/genetics , Wound Healing/genetics , Animals , Cell Movement/genetics , Fibrillar Collagens/genetics , Fibrillar Collagens/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/pathology , Humans , Inflammation/pathology , Mice , Skin/growth & development , Skin/metabolism , Tensile Strength
4.
Matrix Biol ; 67: 90-106, 2018 04.
Article in English | MEDLINE | ID: mdl-29248556

ABSTRACT

Activated fibroblasts are key players in the injury response, tumorigenesis, fibrosis, and inflammation. Dichotomous outcomes in response to varied stroma-targeted therapies in cancer emphasize the need to disentangle the roles of heterogeneous fibroblast subsets in physiological and pathophysiological settings. In wound healing, fibrosis, and myriad tumor types, fibroblast activation protein (FAP) and alpha-smooth muscle actin (αSMA) identify distinct, yet overlapping, activated fibroblast subsets. Prior studies established that FAPHi reactive fibroblasts and αSMAHi myofibroblasts can exert opposing influences in tumorigenesis. However, the factors that drive this phenotypic heterogeneity and the unique functional roles of these subsets have not been defined. We demonstrate that a convergence of ECM composition, elasticity, and transforming growth factor beta (TGF-ß) signaling governs activated fibroblast phenotypic heterogeneity. Furthermore, FAPHi reactive fibroblasts and αSMAHi myofibroblasts exhibited distinct gene expression signatures and functionality in vitro, illuminating potentially unique roles of activated fibroblast subsets in tissue remodeling. These insights into activated fibroblast heterogeneity will inform the rational design of stroma-targeted therapies for cancer and fibrosis.


Subject(s)
Actins/metabolism , Extracellular Matrix/metabolism , Fibroblasts/cytology , Gelatinases/metabolism , Membrane Proteins/metabolism , Myofibroblasts/cytology , Serine Endopeptidases/metabolism , Animals , Cell Differentiation , Cells, Cultured , Endopeptidases , Fibroblasts/metabolism , Mice , Myofibroblasts/metabolism , Phenotype , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978805

ABSTRACT

Pancreatic ductal adenocarcinomas (PDAs) are desmoplastic and can undergo epithelial-to-mesenchymal transition to confer metastasis and chemoresistance. Studies have demonstrated that phenotypically and functionally distinct stromal cell populations exist in PDAs. Fibroblast activation protein-expressing (FAP-expressing) cells act to enhance PDA progression, while α-smooth muscle actin myofibroblasts can restrain PDA. Thus, identification of precise molecular targets that mediate the protumorigenic activity of FAP+ cells will guide development of therapy for PDA. Herein, we demonstrate that FAP overexpression in the tumor microenvironment correlates with poor overall and disease-free survival of PDA patients. Genetic deletion of FAP delayed onset of primary tumor and prolonged survival of mice in the KPC mouse model of PDA. While genetic deletion of FAP did not affect primary tumor weight in advanced disease, FAP deficiency increased tumor necrosis and impeded metastasis to multiple organs. Lineage-tracing studies unexpectedly showed that FAP is not only expressed by stromal cells, but can also be detected in a subset of CD90+ mesenchymal PDA cells, representing up to 20% of total intratumoral FAP+ cells. These data suggest that FAP may regulate PDA progression and metastasis in cell-autonomous and/or non-cell-autonomous fashions. Together, these data support pursuing FAP as a therapeutic target in PDA.


Subject(s)
Biomarkers, Tumor/physiology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/secondary , Gelatinases/physiology , Membrane Proteins/physiology , Pancreatic Neoplasms/pathology , Serine Endopeptidases/physiology , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Disease Progression , Endopeptidases , Female , Gelatinases/deficiency , Gelatinases/metabolism , Heterografts , Humans , Kaplan-Meier Estimate , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice, Knockout , Middle Aged , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
6.
Front Immunol ; 6: 231, 2015.
Article in English | MEDLINE | ID: mdl-26029216

ABSTRACT

Transition from homeostatic to reactive matrix remodeling is a fundamental adaptive tissue response to injury, inflammatory disease, fibrosis, and cancer. Alterations in architecture, physical properties, and matrix composition result in changes in biomechanical and biochemical cellular signaling. The dynamics of pericellular and extracellular matrices, including matrix protein, proteoglycan, and glycosaminoglycan modification are continually emerging as essential regulatory mechanisms underlying cellular and tissue function. Nevertheless, the impact of matrix organization on inflammation and immunity in particular and the consequent effects on tissue healing and disease outcome are arguably under-studied aspects of adaptive stress responses. Herein, we review how the predominant glycosaminoglycan hyaluronan (HA) contributes to the structure and function of the tissue microenvironment. Specifically, we examine the evidence of HA degradation and the generation of biologically active smaller HA fragments in pathological settings in vivo. We discuss how HA fragments versus nascent HA via alternate receptor-mediated signaling influence inflammatory cell recruitment and differentiation, resident cell activation, as well as tumor growth, survival, and metastasis. Finally, we discuss how HA fragmentation impacts restoration of normal tissue function and pathological outcomes in disease.

SELECTION OF CITATIONS
SEARCH DETAIL
...