Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Clin Transl Med ; 14(5): e1655, 2024 May.
Article in English | MEDLINE | ID: mdl-38711203

ABSTRACT

BACKGROUND: Uterine leiomyosarcomas (uLMS) are aggressive tumours with poor prognosis and limited treatment options. Although immune checkpoint blockade (ICB) has proven effective in some 'challenging-to-treat' cancers, clinical trials showed that uLMS do not respond to ICB. Emerging evidence suggests that aberrant PI3K/mTOR signalling can drive resistance to ICB. We therefore explored the relevance of the PI3K/mTOR pathway for ICB treatment in uLMS and explored pharmacological inhibition of this pathway to sensitise these tumours to ICB. METHODS: We performed an integrated multiomics analysis based on TCGA data to explore the correlation between PI3K/mTOR dysregulation and immune infiltration in 101 LMS. We assessed response to PI3K/mTOR inhibitors in immunodeficient and humanized uLMS patient-derived xenografts (PDXs) by evaluating tumour microenvironment modulation using multiplex immunofluorescence. We explored response to single-agent and a combination of PI3K/mTOR inhibitors with PD-1 blockade in humanized uLMS PDXs. We mapped intratumoural dynamics using single-cell RNA/TCR sequencing of serially collected biopsies. RESULTS: PI3K/mTOR over-activation (pS6high) associated with lymphocyte depletion and wound healing immune landscapes in (u)LMS, suggesting it contributes to immune evasion. In contrast, PI3K/mTOR inhibition induced profound tumour microenvironment remodelling in an ICB-resistant humanized uLMS PDX model, fostering adaptive anti-tumour immune responses. Indeed, PI3K/mTOR inhibition induced macrophage repolarisation towards an anti-tumourigenic phenotype and increased antigen presentation on dendritic and tumour cells, but also promoted infiltration of PD-1+ T cells displaying an exhausted phenotype. When combined with anti-PD-1, PI3K/mTOR inhibition led to partial or complete tumour responses, whereas no response to single-agent anti-PD-1 was observed. Combination therapy reinvigorated exhausted T cells and induced clonal hyper-expansion of a cytotoxic CD8+ T-cell population supported by a CD4+ Th1 niche. CONCLUSIONS: Our findings indicate that aberrant PI3K/mTOR pathway activation contributes to immune escape in uLMS and provides a rationale for combining PI3K/mTOR inhibition with ICB for the treatment of this patient population.


Subject(s)
Leiomyosarcoma , Tumor Microenvironment , Uterine Neoplasms , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Leiomyosarcoma/drug therapy , Humans , Female , Uterine Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Animals , Mice , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use
2.
Int J Mol Sci ; 24(7)2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37047398

ABSTRACT

Inflammatory bowel disease (IBD) refers to chronic intestinal immune-mediated diseases including two main disease manifestations: ulcerative colitis (UC) and Crohn's disease (CD). Epidemiological, clinical, and preclinical evidence has highlighted the potential anti-inflammatory properties of naturally occurring alkaloids. In the present study, we investigated the potential anti-inflammatory activities of the tobacco alkaloids nicotine and anatabine in a dextran sulfate sodium (DSS)-induced UC mouse model with a fully humanized immune system. Our results show that nicotine significantly reduced all acute colitis symptoms and improved colitis-specific endpoints, including histopathologically assessed colon inflammation, tissue damage, and mononuclear cell infiltration. The tobacco alkaloid anatabine showed similar effectiveness trends, although they were generally weaker or not significant. Gene expression analysis in the context of biological network models of IBD further pinpointed a possible mechanism by which nicotine attenuated DSS-induced colitis in humanized mice. The current study enables further investigation of possible molecular mechanisms by which tobacco alkaloids attenuate UC symptoms.


Subject(s)
Alkaloids , Antineoplastic Agents , Colitis, Ulcerative , Colitis , Inflammatory Bowel Diseases , Animals , Mice , Nicotiana/adverse effects , Nicotine/adverse effects , Colitis/chemically induced , Colitis/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Inflammatory Bowel Diseases/metabolism , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Alkaloids/pharmacology , Alkaloids/metabolism , Immune System/metabolism , Dextran Sulfate/toxicity , Mice, Inbred C57BL , Colon/metabolism
3.
Blood Adv ; 5(21): 4338-4351, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34516642

ABSTRACT

Tissue invasion by tumor cells induces a host inflammatory response that variably impacts tumorigenesis. This has been well documented for tumor-associated macrophages (TAMs) that could play a pro/M2- or an anti/M1-tumoral function. TAMs frequently infiltrate diffuse large B-cell lymphoma (DLBCL), an aggressive neoplasm arising from germinal center-experienced B cells. However, the pathway leading to the presence of TAMs in DLBCL remains unknown, and their impact is unclear. Here, we show that some DLBCL tumor cells expressed the chemokine CCL5, enabling the differential recruitment of blood monocytes through their expression of CCR1 and CCR5. CCL5 expression by DLBCL was not related to molecular subtypes, and healthy tonsillar B cells did not produce this chemokine, implying a posttransformation event. A single-cell analysis revealed that most DLBCL TAMs had a noncanonical gene signature with the concomitant expression of M1 and M2 genes. The presence of noncanonical TAMs may explain the lack of impact of macrophages on DLBCL development reported in some survival studies.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Chemokine CCL5/genetics , Germinal Center , Humans , Leukocyte Count , Macrophages , Monocytes , Tumor Microenvironment
4.
Int J Mol Sci ; 21(23)2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33297592

ABSTRACT

Genomic instability is generally considered as a hallmark of tumorigenesis and a prerequisite condition for malignant transformation. Aluminium salts are suspected environmental carcinogens that transform mammary epithelial cells in vitro through unknown mechanisms. We report here that long-term culture in the presence of aluminium chloride (AlCl3) enables HC11 normal mouse mammary epithelial cells to form tumours and metastases when injected into the syngeneic and immunocompetent BALB/cByJ strain. We demonstrate that AlCl3 rapidly increases chromosomal structural abnormalities in mammary epithelial cells, while we failed to detect direct modulation of specific mRNA pathways. Our observations provide evidence that clastogenic activity-a well-recognized inducer of genomic instability-might account in part for the transforming abilities of aluminium in mammary epithelial cells.


Subject(s)
Aluminum/toxicity , Carcinogenesis/genetics , Carcinogens, Environmental/toxicity , Genomic Instability , Animals , Carcinogenesis/chemically induced , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C
5.
J Virol ; 93(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31375580

ABSTRACT

Long-acting antiretrovirals could provide a useful alternative to daily oral therapy for HIV-1-infected individuals. Building on a bi-specific molecule with adnectins targeting CD4 and gp41, a potential long-acting biologic, GSK3732394, was developed with three independent and synergistic modes of HIV entry inhibition that potentially could be self-administered as a long-acting subcutaneous injection. Starting with the bi-specific inhibitor, an α-helical peptide inhibitor was optimized as a linked molecule to the anti-gp41 adnectin, with each separate inhibitor exhibiting at least single-digit nanomolar (or lower) potency and a broad spectrum. Combination of the two adnectins and peptide activities into a single molecule was shown to have synergistic advantages in potency, the resistance barrier, and the ability to inhibit HIV-1 infections at low levels of CD4 receptor occupancy, showing that GSK3732394 can work in trans on a CD4+ T cell. Addition of a human serum albumin molecule prolongs the half-life in a human CD4 transgenic mouse, suggesting that it may have potential as a long-acting agent. GSK3732394 was shown to be highly effective in a humanized mouse model of infection. GSK3732394 is currently in clinical trials.IMPORTANCE There continue to be significant unmet medical needs for patients with HIV-1 infection. One way to improve adherence and decrease the likelihood of drug-drug interactions in HIV-1-infected patients is through the development of long-acting biologic inhibitors. Building on a bi-specific inhibitor approach targeting CD4 and gp41, a tri-specific molecule was generated with three distinct antiviral activities. The linkage of these three biologic inhibitors creates synergy that offers a series of advantages to the molecule. The addition of human serum albumin to the tri-specific inhibitor could allow it to function as a long-acting self-administered treatment for patients with HIV infection. This molecule is currently in early clinical trials.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Amino Acid Sequence , Animals , Disease Models, Animal , Drug Resistance, Viral , HIV Fusion Inhibitors/chemistry , HIV Infections/drug therapy , HIV Infections/virology , Humans , Mice , Mice, Transgenic , Models, Molecular , Peptides/chemistry , Peptides/pharmacology , Protein Conformation
6.
Cancer Res ; 79(3): 663-675, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30514753

ABSTRACT

Chronic myeloid leukemia (CML) is a chronic disease resulting in myeloid cell expansion through expression of the BCR-ABL1 fusion transcript. Tyrosine kinase inhibitors (TKI) have significantly increased survival of patients with CML, and deep responders may consider stopping the treatment. However, more than 50% of patients relapse and restart TKI, subsequently suffering unknown toxicity. Because CML is a model immune system-sensitive disease, we hypothesize that chimeric antigen receptor (CAR) T cells targeting IL1 receptor-associated protein (IL1RAP) in quiescent CML stem cells may offer an opportunity for a permanent cure. In this study, we produced and molecularly characterized a specific monoclonal anti-IL1RAP antibody from which fragment antigen-binding nucleotide coding sequences were cloned as a single chain into a lentiviral backbone and secured with the suicide gene iCASP9/rimiducid system. Our CAR T-cell therapy exhibited cytotoxicity against both leukemic stem cells and, to a lesser extent, monocytes expressing IL1RAP, with no apparent effect on the hematopoietic system, including CD34+ stem cells. This suggests IL1RAP as a tumor-associated antigen for immunotherapy cell targeting. IL1RAP CAR T cells were activated in the presence of IL1RAP+ cell lines or primary CML cells, resulting in secretion of proinflammatory cytokines and specifically killing in vitro and in a xenograft murine model. Overall, we demonstrate the proof of concept of a CAR T-cell immunotherapy approach in the context of CML that is applicable for young patients and primary TKI-resistant, intolerant, or allograft candidate patients. SIGNIFICANCE: These findings present the first characterization and proof of concept of a chimeric antigen receptor directed against IL1RAP expressed by leukemic stem cells in the context of CML.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Interleukin-1 Receptor Accessory Protein/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Animals , Antibodies, Monoclonal/immunology , Cell Engineering/methods , Cytotoxicity, Immunologic , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
7.
Cancer Res ; 77(5): 1097-1107, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27923834

ABSTRACT

Tumor-infiltrating neutrophils have been implicated in malignant development and progression, but mechanisms are ill defined. Neutrophils produce a proliferation-inducing ligand APRIL/TNFSF13, a factor that promotes development of tumors from diverse origins, including diffuse large B-cell lymphoma (DLBCL). High APRIL expression in DLBCL correlates with reduced patient survival, but the pathway(s) dictating APRIL expression are not known. Here, we show that all blood neutrophils constitutively secrete APRIL, and inflammation-associated stimuli, such as TNF, further upregulate APRIL. In a significant fraction of DLBCL patients, tumor cells constitutively produced the ELC-CXC chemokine CXCL-8 (IL8), enabling them to recruit APRIL-producing blood neutrophils. CXCL-8 production in DLBCL was unrelated to the cell of origin, as APRIL-producing neutrophils infiltrated CXCL-8+ DLBCL from both germinal center (GC) and non-GC subtypes. Rather, CXCL-8 production implied events affecting DNA methylation and acetylation. Overall, our results showed that chemokine-mediated recruitment of neutrophils secreting the tumor-promoting factor APRIL mediates DLBCL progression. Cancer Res; 77(5); 1097-107. ©2016 AACR.


Subject(s)
Interleukin-8/biosynthesis , Lymphoma, Large B-Cell, Diffuse/immunology , Neutrophils/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/biosynthesis , Animals , Humans , Interleukin-8/immunology , Ligands , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Neutrophils/metabolism , Neutrophils/pathology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology
8.
Proc Natl Acad Sci U S A ; 113(44): 12532-12537, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791128

ABSTRACT

Therapeutic targeting of the VEGF signaling axis by the VEGF-neutralizing monoclonal antibody bevacizumab has clearly demonstrated clinical benefit in cancer patients. To improve this strategy using a polyclonal approach, we developed a vaccine targeting VEGF using 3D-structured peptides that mimic the bevacizumab binding site. An in-depth study on peptide optimization showed that the antigen's 3D structure is essential to achieve neutralizing antibody responses. Peptide 1 adopts a clear secondary, native-like structure, including the typical cysteine-knot fold, as evidenced by CD spectroscopy. Binding and competition studies with bevacizumab in ELISA and surface plasmon resonance analysis revealed that peptide 1 represents the complete bevacizumab binding site, including the hairpin loop (ß5-turn-ß6) and the structure-supporting ß2-α2-ß3 loop. Vaccination with peptide 1 elicited high titers of cross-reactive antibodies to VEGF, with potent neutralizing activity. Moreover, vaccination-induced antisera displayed strong angiostatic and tumor-growth-inhibiting properties in a preclinical mouse model for colorectal carcinoma, whereas antibodies raised with peptides exclusively encompassing the ß5-turn-ß6 loop (peptides 15 and 20) did not. Immunization with peptide 1 or 7 (murine analog of 1) in combination with the potent adjuvant raffinose fatty acid sulfate ester (RFASE) showed significant inhibition of tumor growth in the B16F10 murine melanoma model. Based on these data, we conclude that this vaccination technology, which is currently being investigated in a phase I clinical trial (NCT02237638), can potentially outperform currently applied anti-VEGF therapeutics.


Subject(s)
Bevacizumab/therapeutic use , Colonic Neoplasms/drug therapy , Peptides/therapeutic use , Vaccination/methods , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Bevacizumab/immunology , Binding Sites/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cross Reactions/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Targeted Therapy/methods , Peptides/chemistry , Peptides/immunology , Rats, Wistar , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
9.
J Clin Invest ; 125(8): 2979-94, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26214525

ABSTRACT

Heterozygous germline mutations in the zinc finger transcription factor GATA2 have recently been shown to underlie a range of clinical phenotypes, including Emberger syndrome, a disorder characterized by lymphedema and predisposition to myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). Despite well-defined roles in hematopoiesis, the functions of GATA2 in the lymphatic vasculature and the mechanisms by which GATA2 mutations result in lymphedema have not been characterized. Here, we have provided a molecular explanation for lymphedema predisposition in a subset of patients with germline GATA2 mutations. Specifically, we demonstrated that Emberger-associated GATA2 missense mutations result in complete loss of GATA2 function, with respect to the capacity to regulate the transcription of genes that are important for lymphatic vessel valve development. We identified a putative enhancer element upstream of the key lymphatic transcriptional regulator PROX1 that is bound by GATA2, and the transcription factors FOXC2 and NFATC1. Emberger GATA2 missense mutants had a profoundly reduced capacity to bind this element. Conditional Gata2 deletion in mice revealed that GATA2 is required for both development and maintenance of lymphovenous and lymphatic vessel valves. Together, our data unveil essential roles for GATA2 in the lymphatic vasculature and explain why a select catalogue of human GATA2 mutations results in lymphedema.


Subject(s)
GATA2 Transcription Factor/metabolism , Lymphatic Vessels/embryology , Lymphedema/embryology , Mutation , Animals , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/genetics , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/pathology , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Oncotarget ; 6(12): 10253-66, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25860935

ABSTRACT

The interaction between tumor cells and their microenvironment is an essential aspect of tumor development. Therefore, understanding how this microenvironment communicates with tumor cells is crucial for the development of new anti-cancer therapies. MicroRNAs (miRNAs) are small non-coding RNAs that inhibit gene expression. They are secreted into the extracellular medium in vesicles called exosomes, which allow communication between cells via the transfer of their cargo. Consequently, we hypothesized that circulating endothelial miRNAs could be transferred to tumor cells and modify their phenotype. Using exogenous miRNA, we demonstrated that endothelial cells can transfer miRNA to tumor cells via exosomes. Using miRNA profiling, we identified miR-503, which exhibited downregulated levels in exosomes released from endothelial cells cultured under tumoral conditions. The modulation of miR-503 in breast cancer cells altered their proliferative and invasive capacities. We then identified two targets of miR-503, CCND2 and CCND3. Moreover, we measured increased plasmatic miR-503 in breast cancer patients after neoadjuvant chemotherapy, which could be partly due to increased miRNA secretion by endothelial cells. Taken together, our data are the first to reveal the involvement of the endothelium in the modulation of tumor development via the secretion of circulating miR-503 in response to chemotherapy treatment.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Chemotherapy, Adjuvant , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Human Umbilical Vein Endothelial Cells , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , Neoadjuvant Therapy
12.
Nat Med ; 20(7): 741-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24929950

ABSTRACT

The N-terminal fragment of prolactin (16K PRL) inhibits tumor growth by impairing angiogenesis, but the underlying mechanisms are unknown. Here, we found that 16K PRL binds the fibrinolytic inhibitor plasminogen activator inhibitor-1 (PAI-1), which is known to contextually promote tumor angiogenesis and growth. Loss of PAI-1 abrogated the antitumoral and antiangiogenic effects of 16K PRL. PAI-1 bound the ternary complex PAI-1-urokinase-type plasminogen activator (uPA)-uPA receptor (uPAR), thereby exerting antiangiogenic effects. By inhibiting the antifibrinolytic activity of PAI-1, 16K PRL also protected mice against thromboembolism and promoted arterial clot lysis. Thus, by signaling through the PAI-1-uPA-uPAR complex, 16K PRL impairs tumor vascularization and growth and, by inhibiting the antifibrinolytic activity of PAI-1, promotes thrombolysis.


Subject(s)
Fibrinolysis , Neovascularization, Pathologic , Plasminogen Activator Inhibitor 1/physiology , Prolactin/physiology , Animals , Cell Division , Cells, Cultured , Humans , Mice , Mice, Knockout , Neoplasms/blood supply , Neoplasms/pathology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Prolactin/chemistry
13.
Mol Oncol ; 8(3): 689-703, 2014 May.
Article in English | MEDLINE | ID: mdl-24593875

ABSTRACT

Bone metastasis represents one of the most deleterious clinical consequences arising in the context of many solid tumors. Severe osteolysis results from tumor cell colonization of the bone compartment, a process which entails reciprocal exchange of soluble signals between tumor cells and their osseous microenvironment. Recent evidence indicates that tumor-intrinsic miRNAs are pleiotropic regulators of gene expression. But they are also frequently released in exosome-like vesicles (ELV). Yet the functional relevance of the transference of tumor-derived ELV and their miRNA cargo to the extracellular milieu during osseous colonization is unknown. Comparative transcriptomic profiling using an in vivo murine model of bone metastasis identified a repressed miRNA signature associated with high prometastatic activity. Forced expression of single miRNAs identified miR-192 that markedly appeased osseous metastasis in vivo, as shown by X-ray, bioluminescence imaging and microCT scans. Histological examination of metastatic lesions revealed impaired tumor-induced angiogenesis in vivo, an effect that was associated in vitro with decreased hallmarks of angiogenesis. Isolation and characterization of ELV by flow cytometry, Western blot analysis, transmission electron microscopy and nanoparticle tracking analysis revealed the ELV cargo enrichment in miR-192. Consistent with these findings, fluorescent labeled miR-192-enriched-ELV showed the in vitro transfer and release of miR-192 in target endothelial cells and abrogation of the angiogenic program by repression of proangiogenic IL-8, ICAM and CXCL1. Moreover, in vivo infusion of fluorescent labeled ELV efficiently targeted cells of the osseous compartment. Furthermore, treatment with miR-192 enriched ELV in a model of in vivo bone metastasis pre-conditioned osseous milieu and impaired tumor-induced angiogenesis, thereby reducing the metastatic burden and tumor colonization. Changes in the miRNA-cargo content within ELV represent a novel mechanism heavily influencing bone metastatic colonization, which is most likely relevant in other target organs. Mechanistic mimicry of this phenomenon by synthetic nanoparticles could eventually emerge as a novel therapeutic approach.


Subject(s)
Adenocarcinoma/pathology , Bone Neoplasms/secondary , Bone and Bones/pathology , Exosomes/pathology , Lung Neoplasms/pathology , MicroRNAs/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone and Bones/metabolism , Cell Line, Tumor , Exosomes/genetics , Exosomes/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
14.
Angiogenesis ; 16(4): 877-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23800974

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disorder characterized by arteriovenous malformations and hemorrhages. This vascular disease results mainly from mutations in 2 genes involved in the TGF-ß pathway (ENG and ALK1) that are exclusively expressed by endothelial cells. The present study identified miR-27a and miR-205 as two circulating miRNAs differentially expressed in HHT patients. The plasma levels of miR-27a are elevated while those of miR-205 are reduced in both HHT1 and HHT2 patients compared to healthy controls. The role of miR-205 in endothelial cells was further investigated. Our data indicates that miR-205 expression displaces the TGF-ß balance towards the anti-angiogenic side by targeting Smad1 and Smad4. In line, overexpression of miR-205 in endothelial cells reduces proliferation, migration and tube formation while its inhibition shows opposite effects. This study not only suggests that detection of circulating miRNA (miR-27a and miR-205) could help for the screening of HHT patients but also provides a functional link between the deregulated expression of miR-205 and the HHT phenotype.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/physiology , Neovascularization, Pathologic/genetics , Signal Transduction/physiology , Telangiectasia, Hereditary Hemorrhagic/genetics , Transcriptome , Transforming Growth Factor beta/physiology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Down-Regulation , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , MicroRNAs/blood , MicroRNAs/genetics , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/physiopathology , Oligonucleotides, Antisense/pharmacology , Phenotype , ROC Curve , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/genetics , Smad1 Protein/biosynthesis , Smad1 Protein/genetics , Smad4 Protein/biosynthesis , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/blood , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/physiopathology , Transforming Growth Factor beta/pharmacology
15.
J Clin Invest ; 123(5): 2143-54, 2013 May.
Article in English | MEDLINE | ID: mdl-23619365

ABSTRACT

Peripartum cardiomyopathy (PPCM) is a life-threatening pregnancy-associated cardiomyopathy in previously healthy women. Although PPCM is driven in part by the 16-kDa N-terminal prolactin fragment (16K PRL), the underlying molecular mechanisms are poorly understood. We found that 16K PRL induced microRNA-146a (miR-146a) expression in ECs, which attenuated angiogenesis through downregulation of NRAS. 16K PRL stimulated the release of miR-146a-loaded exosomes from ECs. The exosomes were absorbed by cardiomyocytes, increasing miR-146a levels, which resulted in a subsequent decrease in metabolic activity and decreased expression of Erbb4, Notch1, and Irak1. Mice with cardiomyocyte-restricted Stat3 knockout (CKO mice) exhibited a PPCM-like phenotype and displayed increased cardiac miR-146a expression with coincident downregulation of Erbb4, Nras, Notch1, and Irak1. Blocking miR-146a with locked nucleic acids or antago-miRs attenuated PPCM in CKO mice without interrupting full-length prolactin signaling, as indicated by normal nursing activities. Finally, miR-146a was elevated in the plasma and hearts of PPCM patients, but not in patients with dilated cardiomyopathy. These results demonstrate that miR-146a is a downstream-mediator of 16K PRL that could potentially serve as a biomarker and therapeutic target for PPCM.


Subject(s)
Cardiomyopathies/blood , Cardiomyopathies/genetics , MicroRNAs/blood , Pregnancy Complications, Cardiovascular/blood , Prolactin/metabolism , Animals , Biomarkers/blood , Endothelial Cells/cytology , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neovascularization, Pathologic , Peripartum Period , Pregnancy , Pregnancy Complications, Cardiovascular/metabolism , Rats , STAT3 Transcription Factor/metabolism , Signal Transduction
16.
Cancer Discov ; 2(3): 270-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22585997

ABSTRACT

UNLABELLED: Invasion and metastasis increase after the inhibition of VEGF signaling in some preclinical tumor models. In the present study we asked whether selective VEGF inhibition is sufficient to increase invasion and metastasis and whether selective c-Met inhibition is sufficient to block this effect. Treatment of pancreatic neuroendocrine tumors in RIP-Tag2 mice with a neutralizing anti-VEGF antibody reduced tumor burden but increased tumor hypoxia, hypoxia-inducible factor-1α, and c-Met activation and also increased invasion and metastasis. However, invasion and metastasis were reduced by concurrent inhibition of c-Met by PF-04217903 or PF-02341066 (crizotinib). A similar benefit was found in orthotopic Panc-1 pancreatic carcinomas treated with sunitinib plus PF-04217903 and in RIP-Tag2 tumors treated with XL184 (cabozantinib), which simultaneously blocks VEGF and c-Met signaling. These findings document that invasion and metastasis are promoted by selective inhibition of VEGF signaling and can be reduced by the concurrent inhibition of c-Met. SIGNIFICANCE: This report examines the mechanism of increased tumor aggressiveness after anti-VEGF therapy and presents evidence for roles of vascular pruning, hypoxia, and c-Met activation. The results show that simultaneous inhibition of c-Met and VEGF signaling not only slows tumor growth but also reduces invasion and metastasis.


Subject(s)
Anilides/pharmacology , Antibodies, Neutralizing/pharmacology , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Neutralizing/immunology , Cell Line, Tumor , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
17.
PLoS One ; 6(11): e27318, 2011.
Article in English | MEDLINE | ID: mdl-22087289

ABSTRACT

BACKGROUND: Angiogenesis, the formation of new blood vessels from existing vasculature, plays an essential role in tumor growth, invasion, and metastasis. 16K hPRL, the antiangiogenic 16-kDa N-terminal fragment of human prolactin was shown to prevent tumor growth and metastasis by modifying tumor vessel morphology. METHODOLOGY/PRINCIPAL FINDINGS: Here we investigated the effect of 16K hPRL on tumor vessel maturation and on the related signaling pathways. We show that 16K hPRL treatment leads, in a murine B16-F10 tumor model, to a dysfunctional tumor vasculature with reduced pericyte coverage, and disruption of the PDGF-B/PDGFR-B, Ang/Tie2, and Delta/Notch pathways. In an aortic ring assay, 16K hPRL impairs endothelial cell and pericyte outgrowth from the vascular ring. In addition, 16K hPRL prevents pericyte migration to endothelial cells. This event was independent of a direct inhibitory effect of 16K hPRL on pericyte viability, proliferation, or migration. In endothelial cell-pericyte cocultures, we found 16K hPRL to disturb Notch signaling. CONCLUSIONS/SIGNIFICANCE: Taken together, our data show that 16K hPRL impairs functional tumor neovascularization by inhibiting vessel maturation and for the first time that an endogenous antiangiogenic agent disturbs Notch signaling. These findings provide new insights into the mechanisms of 16K hPRL action and highlight its potential for use in anticancer therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Blood Vessels/growth & development , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Prolactin/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Blood Vessels/drug effects , Blood Vessels/pathology , Coculture Techniques , Endothelial Cells , Mice , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Pericytes , Prolactin/therapeutic use , Signal Transduction/drug effects
18.
Am J Pathol ; 178(6): 2897-909, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21550017

ABSTRACT

Blood vessel leakiness is an early, transient event in acute inflammation but can also persist as vessels undergo remodeling in sustained inflammation. Angiopoietin/Tie2 signaling can reduce the leakiness through changes in endothelial cells. The role of pericytes in this action has been unknown. We used the selective PDGF-B-blocking oligonucleotide aptamer AX102 to determine whether disruption of pericyte-endothelial crosstalk alters vascular leakiness or remodeling in the airways of mice under four different conditions: i) baseline, ii) acute inflammation induced by bradykinin, iii) sustained inflammation after 7-day infection by the respiratory pathogen Mycoplasma pulmonis, or iv) leakage after bradykinin challenge in the presence of vascular stabilization by the angiopoietin-1 (Ang1) mimic COMP-Ang1 for 7 days. AX102 reduced pericyte coverage but did not alter the leakage of microspheres from tracheal blood vessels at baseline or after bradykinin; however, AX102 exaggerated leakage at 7 days after M. pulmonis infection and increased vascular remodeling and disease severity at 14 days. AX102 also abolished the antileakage effect of COMP-Ang1 at 7 days. Together, these findings show that pericyte contributions to endothelial stability have greater dependence on PDGF-B during the development of sustained inflammation, when pericyte dynamics accompany vascular remodeling, than under baseline conditions or in acute inflammation. The findings also show that the antileakage action of Ang1 requires PDGF-dependent actions of pericytes in maintaining endothelial stability.


Subject(s)
Angiopoietin-1/metabolism , Inflammation/pathology , Pericytes/pathology , Trachea/blood supply , Trachea/pathology , Actins/metabolism , Animals , Aptamers, Nucleotide/pharmacology , Bradykinin/pharmacology , Cell Count , Cell Shape/drug effects , Desmin/metabolism , Inflammation/complications , Mice , Mice, Inbred C57BL , Microspheres , Mycoplasma Infections/complications , Mycoplasma Infections/pathology , Mycoplasma pulmonis/drug effects , Mycoplasma pulmonis/physiology , Pericytes/drug effects , Pericytes/microbiology , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/pharmacology , Trachea/microbiology
19.
Am J Pathol ; 177(6): 3233-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20952594

ABSTRACT

Vascular remodeling is a feature of chronic inflammation during which capillaries transform into venules that expand the region of the vasculature in which leakage and leukocyte emigration both occur. Recently, we found that angiopoietin/Tie2 receptor signaling drives the transformation of capillaries into venules at an early stage of the sustained inflammatory response in the airways of mice infected with Mycoplasma pulmonis. However, the precise contributions of both angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are not clear. In this study, we sought to determine the contribution of Ang2 to this vascular remodeling. Ang2 mRNA expression levels increased and phosphorylated Tie2 immunoreactivity in mucosal blood vessels decreased, indicative of diminished receptor signaling after infection. Selective inhibition of Ang2 throughout the infection by administration of either of two distinct function-blocking antibodies reduced the suppression of Tie2 phosphorylation and decreased the remodeling of mucosal capillaries into venules, the amount of leukocyte influx, and disease severity. These findings are consistent with Ang2 acting as an antagonist of Tie2 receptors and the reduction of Tie2 phosphorylation in endothelial cells rendering the vasculature more responsive to cytokines that promote both vascular remodeling and the consequences of inflammation after M. pulmonis infection. By blocking such changes, Ang2 inhibitors may prove beneficial in the treatment of sustained inflammation in which vascular remodeling, leakage, and leukocyte influx contribute to its pathophysiology.


Subject(s)
Angiopoietin-2/physiology , Blood Vessels/physiology , Neovascularization, Physiologic/genetics , Respiratory System/blood supply , Respiratory Tract Diseases/genetics , Angiopoietin-2/genetics , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Animals , Blood Vessels/metabolism , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mycoplasma Infections/complications , Mycoplasma Infections/genetics , Mycoplasma Infections/metabolism , Mycoplasma pulmonis/physiology , Neovascularization, Physiologic/physiology , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/metabolism , Pneumonia/pathology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-2 , Respiratory System/metabolism , Respiratory System/pathology , Respiratory Tract Diseases/metabolism , Respiratory Tract Diseases/pathology
20.
Mol Cancer ; 9: 231, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20813052

ABSTRACT

BACKGROUND: Disorganized angiogenesis is associated with several pathologies, including cancer. The identification of new genes that control tumor neovascularization can provide novel insights for future anti-cancer therapies. Sprouty1 (SPRY1), an inhibitor of the MAPK pathway, might be one of these new genes. We identified SPRY1 by comparing the transcriptomes of untreated endothelial cells with those of endothelial cells treated by the angiostatic agent 16 K prolactin (16 K hPRL). In the present study, we aimed to explore the potential function of SPRY1 in angiogenesis. RESULTS: We confirmed 16 K hPRL induced up-regulation of SPRY1 in primary endothelial cells. In addition, we demonstrated the positive SPRY1 regulation in a chimeric mouse model of human colon carcinoma in which 16 K hPRL treatment was shown to delay tumor growth. Expression profiling by qRT-PCR with species-specific primers revealed that induction of SPRY1 expression by 16 K hPRL occurs only in the (murine) endothelial compartment and not in the (human) tumor compartment. The regulation of SPRY1 expression was NF-κB dependent. Partial SPRY1 knockdown by RNA interference protected endothelial cells from apoptosis as well as increased endothelial cell proliferation, migration, capillary network formation, and adhesion to extracellular matrix proteins. SPRY1 knockdown was also shown to affect the expression of cyclinD1 and p21 both involved in cell-cycle regulation. These findings are discussed in relation to the role of SPRY1 as an inhibitor of ERK/MAPK signaling and to a possible explanation of its effect on cell proliferation. CONCLUSIONS: Taken together, these results suggest that SPRY1 is an endogenous angiogenesis inhibitor.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neovascularization, Pathologic/metabolism , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Prolactin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , Cattle , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Female , HCT116 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Peptide Fragments/pharmacology , Phosphoproteins/genetics , Prolactin/pharmacology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...