Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Bone Oncol ; 47: 100610, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38984147

ABSTRACT

The skeleton is a common site of cancer metastasis and malignancy with the resultant lesions often being incurable. Interactions between metastatic cancer cells and the bone microenvironment are critical for cancer cell survival, outgrowth, and progression. Mesenchymal Stem Cells (MSCs) are an essential stromal cell type in bone that are appreciated for their impacts on cancer-induced bone disease, however, newer evidence suggests that MSCs possess extensive roles in cancer-bone crosstalk, including cancer cell dormancy, metabolic demands, and immune-oncology. Emerging evidence has also identified the importance of MSC tissue source and the influence of ageing when studying MSC biology. Combining these considerations together with developing technologies such as spatial transcriptomics will contribute to defining the molecular mechanisms underlying complex stroma-cancer interactions in bone and assist with identification of therapeutically tractable targets.

2.
Nat Commun ; 15(1): 2458, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503736

ABSTRACT

Multiple myeloma (MM) is an osteolytic malignancy that is incurable due to the emergence of treatment resistant disease. Defining how, when and where myeloma cell intrinsic and extrinsic bone microenvironmental mechanisms cause relapse is challenging with current biological approaches. Here, we report a biology-driven spatiotemporal hybrid agent-based model of the MM-bone microenvironment. Results indicate MM intrinsic mechanisms drive the evolution of treatment resistant disease but that the protective effects of bone microenvironment mediated drug resistance (EMDR) significantly enhances the probability and heterogeneity of resistant clones arising under treatment. Further, the model predicts that targeting of EMDR deepens therapy response by eliminating sensitive clones proximal to stroma and bone, a finding supported by in vivo studies. Altogether, our model allows for the study of MM clonal evolution over time in the bone microenvironment and will be beneficial for optimizing treatment efficacy so as to significantly delay disease relapse.


Subject(s)
Multiple Myeloma , Humans , Bone and Bones/pathology , Chronic Disease , Drug Resistance , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/genetics , Tumor Microenvironment
3.
Sci Adv ; 9(18): eadf0108, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37134157

ABSTRACT

Immune checkpoint blockade has been largely unsuccessful for the treatment of bone metastatic castrate-resistant prostate cancer (mCRPC). Here, we report a combinatorial strategy to treat mCRPC using γδ-enriched chimeric antigen receptor (CAR) T cells and zoledronate (ZOL). In a preclinical murine model of bone mCRPC, γδ CAR-T cells targeting prostate stem cell antigen (PSCA) induced a rapid and significant regression of established tumors, combined with increased survival and reduced cancer-associated bone disease. Pretreatment with ZOL, a U.S. Food and Drug Administration-approved bisphosphonate prescribed to mitigate pathological fracture in mCRPC patients, resulted in CAR-independent activation of γδ CAR-T cells, increased cytokine secretion, and enhanced antitumor efficacy. These data show that the activity of the endogenous Vγ9Vδ2 T cell receptor is preserved in CAR-T cells, allowing for dual-receptor recognition of tumor cells. Collectively, our findings support the use of γδ CAR-T cell therapy for mCRPC treatment.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Chimeric Antigen , United States , Male , Humans , Animals , Mice , Prostatic Neoplasms, Castration-Resistant/therapy , Zoledronic Acid/pharmacology , Receptors, Antigen, T-Cell , Cell- and Tissue-Based Therapy
4.
Nat Commun ; 12(1): 723, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33526787

ABSTRACT

Bone metastatic prostate cancer (PCa) promotes mesenchymal stem cell (MSC) recruitment and their differentiation into osteoblasts. However, the effects of bone-marrow derived MSCs on PCa cells are less explored. Here, we report MSC-derived interleukin-28 (IL-28) triggers prostate cancer cell apoptosis via IL-28 receptor alpha (IL-28Rα)-STAT1 signaling. However, chronic exposure to MSCs drives the selection of prostate cancer cells that are resistant to IL-28-induced apoptosis and therapeutics such as docetaxel. Further, MSC-selected/IL-28-resistant prostate cancer cells grow at accelerated rates in bone. Acquired resistance to apoptosis is PCa cell intrinsic, and is associated with a shift in IL-28Rα signaling via STAT1 to STAT3. Notably, STAT3 ablation or inhibition impairs MSC-selected prostate cancer cell growth and survival. Thus, bone marrow MSCs drive the emergence of therapy-resistant bone metastatic prostate cancer yet this can be disabled by targeting STAT3.


Subject(s)
Adenocarcinoma/secondary , Bone Neoplasms/secondary , Mesenchymal Stem Cells/pathology , Prostatic Neoplasms/pathology , Receptors, Interferon/metabolism , Aminosalicylic Acids/pharmacology , Aminosalicylic Acids/therapeutic use , Animals , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Benzenesulfonates/therapeutic use , Bone Neoplasms/drug therapy , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation , Culture Media, Conditioned/metabolism , Disease Models, Animal , Docetaxel/pharmacology , Docetaxel/therapeutic use , Humans , Interferons/genetics , Interferons/metabolism , Male , Mice, Knockout , Osteoblasts/pathology , Primary Cell Culture , Prostatic Neoplasms/drug therapy , RNA, Small Interfering/metabolism , Receptors, Interferon/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Tibia/pathology
5.
Cancers (Basel) ; 13(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567529

ABSTRACT

BACKGROUND: Bone metastatic prostate cancer (BMPCa), despite the initial responsiveness to androgen deprivation therapy (ADT), inevitably becomes resistant. Recent clinical trials with upfront treatment of ADT combined with chemotherapy or novel hormonal therapies (NHTs) have extended overall patient survival. These results indicate that there is significant potential for the optimization of standard-of-care therapies to delay the emergence of progressive metastatic disease. METHODS: Here, we used data extracted from human bone metastatic biopsies pre- and post-abiraterone acetate/prednisone to generate a mathematical model of bone metastatic prostate cancer that can unravel the treatment impact on disease progression. Intra-tumor heterogeneity in regard to ADT and chemotherapy resistance was derived from biopsy data at a cellular level, permitting the model to track the dynamics of resistant phenotypes in response to treatment from biological first-principles without relying on data fitting. These cellular data were mathematically correlated with a clinical proxy for tumor burden, utilizing prostate-specific antigen (PSA) production as an example. RESULTS: Using this correlation, our model recapitulated the individual patient response to applied treatments in a separate and independent cohort of patients (n = 24), and was able to estimate the initial resistance to the ADT of each patient. Combined with an intervention-decision algorithm informed by patient-specific prediction of initial resistance, we propose to optimize the sequence of treatments for each patient with the goal of delaying the evolution of resistant disease and limit cancer cell growth, offering evidence for an improvement against retrospective data. CONCLUSIONS: Our results show how minimal but widely available patient information can be used to model and track the progression of BMPCa in real time, offering a clinically relevant insight into the patient-specific evolutionary dynamics of the disease and suggesting new therapeutic options for intervention. TRIAL REGISTRATION: NCT # 01953640. FUNDING: Funded by an NCI U01 (NCI) U01CA202958-01 and a Moffitt Team Science Award. CCL and DB were partly funded by an NCI PSON U01 (U01CA244101). AA was partly funded by a Department of Defense Prostate Cancer Research Program (W81XWH-15-1-0184) fellowship. LC was partly funded by a postdoctoral fellowship (PF-13-175-01-CSM) from the American Cancer Society.

6.
Neoplasia ; 22(10): 511-521, 2020 10.
Article in English | MEDLINE | ID: mdl-32896761

ABSTRACT

Bone metastatic prostate cancer significantly impacts patient quality of life and overall survival, and despite available therapies, it is presently incurable with an unmet need for improved treatment options. As mediators of tumor progression, matrix metalloproteinases (MMPs) can degrade extracellular matrix components and regulate growth factor and cytokine bioactivity. Depending on tissue context, MMPs can either promote or inhibit tumorigenesis. Therefore, it is essential to study individual MMPs in specific cancer contexts and microenvironments to support the design and application of selective MMP inhibitors. Here we report that tumor-derived MMP-3 contributes to bone metastatic prostate cancer progression via intrinsic and extrinsic routes. MMP-3 ablation in prostate cancer cell lines significantly reduced in vitro growth combined with lowered AKT and ERK phosphorylation and total VEGFR1 and FGFR3 protein levels. In vivo, MMP-3 ablated tumors grew at a slower rate and were significantly less vascularized. Quantitative PCR analyses of wild type and MMP-3 silenced prostate cancer cells also demonstrate downregulation of a wide array of angiogenic factors. The extrinsic role for MMP-3 in angiogenesis was supported by in vitro endothelial tube formation assays where the lack of MMP-3 in prostate cancer conditioned media resulted in slower rates of tube formation. Taken together, our results suggest that tumor-derived MMP-3 contributes to prostate cancer growth in bone. These data indicate that selective inhibition of MMP-3 and/or targeting MMP generated products could be efficacious for the treatment of prostate to bone metastases.


Subject(s)
Bone Neoplasms/blood supply , Bone Neoplasms/secondary , Matrix Metalloproteinase 3/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Animals , Apoptosis , Bone Neoplasms/enzymology , Cell Proliferation , Disease Models, Animal , Humans , Male , Matrix Metalloproteinase 3/genetics , Mice , Neovascularization, Pathologic/enzymology , Prostatic Neoplasms/enzymology , Tumor Cells, Cultured
7.
Cancer Immunol Immunother ; 69(6): 1113-1130, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32114681

ABSTRACT

Bone metastatic prostate cancer (BM-PCa) significantly reduces overall patient survival and is currently incurable. Current standard immunotherapy showed promising results for PCa patients with metastatic, but less advanced, disease (i.e., fewer than 20 bone lesions) suggesting that PCa growth in bone contributes to response to immunotherapy. We found that: (1) PCa stimulates recruitment of neutrophils, the most abundant immune cell in bone, and (2) that neutrophils heavily infiltrate regions of prostate tumor in bone of BM-PCa patients. Based on these findings, we examined the impact of direct neutrophil-prostate cancer interactions on prostate cancer growth. Bone marrow neutrophils directly induced apoptosis of PCa in vitro and in vivo, such that neutrophil depletion in bone metastasis models enhanced BM-PCa growth. Neutrophil-mediated PCa killing was found to be mediated by suppression of STAT5, a transcription factor shown to promote PCa progression. However, as the tumor progressed in bone over time, neutrophils from late-stage bone tumors failed to elicit cytotoxic effector responses to PCa. These findings are the first to demonstrate that bone-resident neutrophils inhibit PCa and that BM-PCa are able to progress via evasion of neutrophil-mediated killing. Enhancing neutrophil cytotoxicity in bone may present a novel therapeutic option for bone metastatic prostate cancer.


Subject(s)
Bone Neoplasms/secondary , Neutrophils/metabolism , Prostatic Neoplasms/blood , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Neutrophils/cytology , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology
8.
BioDrugs ; 33(6): 647-659, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31552606

ABSTRACT

The adoptive transfer of genetically engineered T cells expressing a chimeric antigen receptor (CAR) has shown remarkable results against B cell malignancies. This immunotherapeutic approach has advanced and expanded rapidly from preclinical models to the recent approval of CAR-T cells to treat lymphomas and leukemia by the Food and Drug Administration (FDA). Ongoing research efforts are focused on employing CAR-T cells as a therapy for other cancers, and enhancing their efficacy and safety by optimizing their design. Here we summarize modifications in the intracellular domain of the CAR that gave rise to first-, second-, third- and next-generation CAR-T cells, together with the impact that these different designs have on CAR-T cell biology and function. Further, we describe how the structure of the antigen-sensing ectodomain can be enhanced, leading to superior CAR-T cell signaling and/or function. Finally we discuss how tissue-specific factors may impact the clinical efficacy of CAR-T cells for bone and the central nervous system, as examples of specific indications that may require further CAR signaling optimization to perform in such inhospitable microenvironments.


Subject(s)
Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunotherapy, Adoptive/methods
9.
Oncogene ; 38(44): 6959-6969, 2019 10.
Article in English | MEDLINE | ID: mdl-31409900

ABSTRACT

Bone metastatic prostate cancer provokes extensive osteogenesis by driving the recruitment and osteoblastic differentiation of mesenchymal stromal cells (MSCs). The resulting lesions greatly contribute to patient morbidity and mortality, underscoring the need for defining how prostate metastases subvert the MSC-osteoblast differentiation program. To gain insights into this process we profiled the effects of co-culture of primary MSCs with validated bone metastatic prostate cancer cell line models. These analyses revealed a cast of shared differentially induced genes in MSC, including betaglycan, a co-receptor for TGFß. Betaglycan has not been studied in the context of bone metastatic disease previously. Here we report that loss of betaglycan in MSC is sufficient to augment TGFß signaling, proliferation and migration, and completely blocks the MSC-osteoblast differentiation program. Further, betaglycan was revealed as necessary for prostate cancer-induced osteogenesis in vivo. Mechanistically, gene expression analysis revealed betaglycan controls the expression of a large repertoire of genes in MSCs, and that betaglycan loss provokes >60-fold increase in the expression of Wnt5a that plays important roles in stemness. In accord with the increased Wnt5a levels, there was a marked induction of canonical Wnt signaling in betaglycan ablated MSCs, and the addition of recombinant Wnt5a to MSCs was sufficient to impair osteogenic differentiation. Finally, the addition of Wnt5a neutralizing antibody was sufficient to induce the expression of osteogenic genes in betaglycan-ablated MSCs. Collectively, these findings suggest a betaglycan-Wnt5a circuit represents an attractive vulnerability to ameliorate prostate cancer-induced osteogenesis.


Subject(s)
Mesenchymal Stem Cells/pathology , Osteoblasts/pathology , Osteogenesis , Prostatic Neoplasms/pathology , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Humans , Male , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Wnt-5a Protein/metabolism
10.
Int J Mol Sci ; 20(11)2019 Jun 08.
Article in English | MEDLINE | ID: mdl-31181800

ABSTRACT

Parathyroid hormone-related protein (PTHrP), with isoforms ranging from 139 to 173 amino acids, has long been implicated in the development and regulation of multiple tissues, including that of the skeleton, via paracrine and autocrine signaling. PTHrP is also known as a potent mediator of cancer-induced bone disease, contributing to a vicious cycle between tumor cells and the bone microenvironment that drives the formation and progression of metastatic lesions. The abundance of roles ascribed to PTHrP have largely been attributed to the N-terminal 1-36 amino acid region, however, activities for mid-region and C-terminal products as well as additional shorter N-terminal species have also been described. Studies of the protein sequence have indicated that PTHrP is susceptible to post-translational proteolytic cleavage by multiple classes of proteases with emerging evidence pointing to novel functional roles for these PTHrP products in regulating cell behavior in homeostatic and pathological contexts. As a consequence, PTHrP products are also being explored as potential biomarkers of disease. Taken together, our enhanced understanding of the post-translational regulation of PTHrP bioactivity could assist in developing new therapeutic approaches that can effectively treat skeletal malignancies.


Subject(s)
Bone Neoplasms/metabolism , Parathyroid Hormone-Related Protein/metabolism , Proteolysis , Animals , Bone Neoplasms/secondary , Humans , Metalloproteases/metabolism , Protein Processing, Post-Translational
11.
Front Immunol ; 9: 2887, 2018.
Article in English | MEDLINE | ID: mdl-30574153

ABSTRACT

Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1ß, TGFß, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppression Therapy/methods , Leukemia/therapy , X-Box Binding Protein 1/antagonists & inhibitors , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Isoantibodies/immunology , Isoantibodies/metabolism , Isoantigens/immunology , Leukemia/immunology , Lymphocyte Activation/drug effects , Male , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Skin Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Chimera , Transplantation, Homologous/adverse effects , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
12.
Cancer Control ; 22(1): 109-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25504285

ABSTRACT

BACKGROUND: A paucity of therapeutic options is available to treat men with metastatic castration-resistant prostate cancer (mCRPC). However, recent developments in our understanding of the disease have resulted in several new therapies that show promise in improving overall survival rates in this patient population. METHODS: Agents approved for use in the United States and those undergoing clinical trials for the treatment of mCRPC are reviewed. Recent contributions to the understanding of prostate biology and bone metastasis are discussed as well as how the underlying mechanisms may represent opportunities for therapeutic intervention. New challenges to delivering effective mCRPC treatment will also be examined. RESULTS: New and emerging treatments that target androgen synthesis and utilization or the microenvironment may improve overall survival rates for men diagnosed with mCRPC. Determining how factors derived from the primary tumor can promote the development of premetastatic niches and how prostate cancer cells parasitize niches in the bone microenvironment, thus remaining dormant and protected from systemic therapy, could yield new therapies to treat mCRPC. Challenges such as intratumoral heterogeneity and patient selection can potentially be circumvented via computational biology approaches. CONCLUSIONS: The emergence of novel treatments for mCRPC, combined with improved patient stratification and optimized therapy sequencing, suggests that significant gains may be made in terms of overall survival rates for men diagnosed with this form of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Tumor Microenvironment/drug effects , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Humans , Male , Prostatic Neoplasms, Castration-Resistant/mortality
13.
Exp Cell Res ; 315(13): 2265-74, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19422822

ABSTRACT

CD40, a member of tumor necrosis factor receptor superfamily, and its cognate ligand CD40L are both elevated in the brain of Alzheimer's disease (AD) patients compared to controls. We have shown that pharmacological or genetic interruption of CD40/CD40L interaction results in mitigation of AD-like pathology in vivo in transgenic AD mouse models, and in vitro. Recently, we showed that CD40L stimulation could increase Abeta levels via NFkappaB signaling, presumably through TRAFs. In the present work, using CD40 mutants, we show that CD40L can increase levels of Abeta(1-40), Abeta(1-42), sAPPbeta, sAPPalpha and CTFbeta independently of TRAF signaling. We report an increase in mature/immature APP ratio after CD40L treatment of CD40wt and CD40-mutant cells, reflecting alterations in APP trafficking. In addition, results from CD40L treatment of a neuroblastoma cell line over-expressing the C-99 APP fragment suggest that CD40L has an effect on gamma-secretase. Furthermore, inhibition of gamma-secretase activity significantly reduces sAPPbeta levels in the CD40L treated HEK/APPsw CD40wt and the CD40-mutant cells. The latter suggests CD40/CD40L interaction primarily acts on gamma-secretase and affects beta-secretase via a positive feedback mechanism. Taken together, our data suggest that CD40/CD40L interaction modulates APP processing independently of TRAF signaling.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Peptide Fragments/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Amyloid beta-Peptides/genetics , Animals , Cell Line , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Peptide Fragments/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Cytokine ; 42(3): 336-44, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18434187

ABSTRACT

One of the hallmarks of Alzheimer's disease (AD) is the accumulation of amyloid beta (Abeta) plaques in the brain parenchyma. An inflammatory component to AD has been suggested in association with increased cytokine release. We have previously shown that CD40L stimulation of microglia induces increases in pro-inflammatory cytokines such as interleukin-1beta (IL-1beta), IL-6, IL-8 and GM-CSF. We have also shown that CD40L stimulation increases Abeta levels in HEK-293 cells over-expressing both the amyloid precursor protein (APP) and CD40 (HEK/APPsw/CD40). In this study, we show that GM-CSF neutralizing antibodies mitigate the CD40L-induced production of Abeta in HEK/APPsw/CD40 cells. In addition, we demonstrate that treatment of these cells with recombinant GM-CSF significantly increases Abeta levels. Furthermore, we show that shRNA silencing of the GM-CSF receptor gene significantly reduces Abeta levels to below base line in non-stimulated HEK/APPsw/CD40 cells. Analysis of cell surface proteins revealed that silencing of the GM-CSF receptor also decreases APP endocytosis (therefore reducing the availability of APP to be cleaved in the endosomes). Taken together, our results suggest that GM-CSF operates downstream of CD40/CD40L interaction and that GM-CSF modulates Abeta production by influencing APP trafficking. GM-CSF signaling may be a suitable therapeutic target against Abeta production in AD.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Animals , CD40 Ligand/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Endocytosis , Humans , Membrane Proteins/metabolism , RNA Interference , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
15.
Eur J Neurosci ; 25(6): 1685-95, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17432959

ABSTRACT

The CD40 receptor is a member of the tumor necrosis factor (TNF) super-family of trans-membrane receptors. Interaction of CD40 with its ligand CD40L mediates a broad range of immune and inflammatory responses in the periphery and in the central nervous system. Recently it has been suggested that CD40/CD40L interaction is involved in amyloid precursor protein (APP) processing and Alzheimer's disease (AD)-like pathology in transgenic mouse models of AD. We have previously shown that pharmacologically inhibiting CD40/CD40L interaction improves memory deficits in the PSAPP AD mouse model. We have also recently shown that CD40 deficiency mitigates amyloid deposition in APPsw and PSAPP mouse models. In the present report, using human embryonic kidney cells (HEK293) over-expressing both the APPsw mutation and CD40, we demonstrate that CD40/CD40L interaction directly increases the production of APP metabolites (Abeta 1-40, Abeta 1-42, CTFs, sAPPbeta and sAPPalpha). The results also show that CD40/CD40L interaction affects APP processing via the NF-kappaB pathway. Using NFkappaB inhibitors and SiRNAs to silence diverse elements of the NFkappaB pathway, we observe a reduction in levels of both Abeta 1-40 and Abeta 1-42. Taken together, our results further suggest that CD40L stimulation may be a key component in AD pathology and that elements of the NF-kappaB pathway may be suitable targets for therapeutic approaches against AD.


Subject(s)
Amyloid beta-Peptides/metabolism , CD40 Antigens/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Amyloid beta-Peptides/classification , Amyloid beta-Peptides/genetics , CD40 Antigens/genetics , Cell Line, Transformed , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Humans , Mutation/physiology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...