Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Trends Cancer ; 9(12): 1085-1096, 2023 12.
Article in English | MEDLINE | ID: mdl-37673713

ABSTRACT

In recent years technologies that can achieve readouts at cellular resolution such as single-cell RNA sequencing (scRNA-seq) have provided a comprehensive characterization of the cellular proportions and phenotypes that populate the tumor microenvironment (TME). However, because of the sample dissociation steps required by these protocols, they fail to capture information related to the intricate spatial context in which cells operate as well as their dense networks of interactions. Spatial profiling technologies have recently emerged as a valuable way to investigate the physical organization of cells crowding the TME in intact tissues. In this review we first discuss how spatial profiling technologies have propelled TME characterization, and then explore their potential to improve both diagnosis and prognosis for cancer patients in the clinic.


Subject(s)
Tumor Microenvironment , Humans , Tumor Microenvironment/genetics , Phenotype
2.
J Crohns Colitis ; 17(12): 1988-2001, 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-37462681

ABSTRACT

IFNγ-producing ex-Th17 cells ['Th1/17'] were shown to play a key pathogenic role in experimental colitis and are abundant in the intestine. Here, we identified and characterised a novel, potentially colitogenic subset of Th17 cells in the intestine of patients with Crohn's disease [CD]. Human Th17 cells expressing CCR5 ['pTh17'] co-expressed T-bet and RORC/γt and produced very high levels of IL-17, together with IFN-γ. They had a gene signature of Th17 effector cells and were distinct from established Th1/17 cells. pTh17 cells, but not Th1/17 cells, were associated with intestinal inflammation in CD, and decreased upon successful anti-TNF therapy with infliximab. Conventional CCR5[-]Th17 cells differentiated to pTh17 cells with IL-23 in vitro. Moreover, anti-IL-23 therapy with risankizumab strongly reduced pTh17 cells in the intestine. Importantly, intestinal pTh17 cells were selectively activated by adherent-invasive Escherichia coli [AIEC], but not by a commensal/probiotic E. coli strain. AIEC induced high levels of IL-23 and RANTES from dendritic cells [DC]. Intestinal CCR5+Th1/17 cells responded instead to cytomegalovirus and were reduced in ulcerative colitis [UC], suggesting an unexpected protective role. In conclusion, we identified an IL-23-inducible subset of human intestinal Th17 cells. pTh17 cells produced high levels of pro-inflammatory cytokines, were selectively associated with intestinal inflammation in CD, and responded to CD-associated AIEC, suggesting a key colitogenic role.


Subject(s)
Crohn Disease , Escherichia coli Infections , Humans , Crohn Disease/pathology , Escherichia coli , Th17 Cells/pathology , Tumor Necrosis Factor Inhibitors , Intestines/pathology , Inflammation/pathology , Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Interleukin-23 , Intestinal Mucosa/pathology , Bacterial Adhesion
3.
Methods Mol Biol ; 2655: 231-244, 2023.
Article in English | MEDLINE | ID: mdl-37213001

ABSTRACT

In the recent years, the establishment of self-organizing 3D cultures (organoids) generated from human primary tissues added a novel and physiological viewpoint to interrogate basic and pathological matters. Indeed, these 3D mini-organs, contrary to cell lines, faithfully reproduce the architecture and the molecular features of their original tissues. In cancer studies, the use of tumor patient-derived organoids (PDOs), capturing the histological and molecular heterogeneity of "pure" cancer cells, offered the opportunity to deeply explore tumor-specific regulatory networks. Accordingly, the study of polycomb group proteins (PcGs) can take advantage from this versatile technology to thoroughly investigate the molecular activity of these master regulators. In particular, the application of chromatin immunoprecipitations (ChIP)-sequencing (ChIP-seq) analyses to organoid models provides a powerful tool toward an accurate inquiry of PcG role in tumor development and maintenance.


Subject(s)
Neoplasms , Humans , Neoplasms/pathology , Organoids/metabolism , Polycomb-Group Proteins
4.
Cell Death Dis ; 14(2): 129, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792589

ABSTRACT

Lipid and cholesterol metabolism play a crucial role in tumor cell behavior and in shaping the tumor microenvironment. In particular, enzymatic and non-enzymatic cholesterol metabolism, and derived metabolites control dendritic cell (DC) functions, ultimately impacting tumor antigen presentation within and outside the tumor mass, dampening tumor immunity and immunotherapeutic attempts. The mechanisms accounting for such events remain largely to be defined. Here we perturbed (oxy)sterol metabolism genetically and pharmacologically and analyzed the tumor lipidome landscape in relation to the tumor-infiltrating immune cells. We report that perturbing the lipidome of tumor microenvironment by the expression of sulfotransferase 2B1b crucial in cholesterol and oxysterol sulfate synthesis, favored intratumoral representation of monocyte-derived antigen-presenting cells, including monocyte-DCs. We also found that treating mice with a newly developed antagonist of the oxysterol receptors Liver X Receptors (LXRs), promoted intratumoral monocyte-DC differentiation, delayed tumor growth and synergized with anti-PD-1 immunotherapy and adoptive T cell therapy. Of note, looking at LXR/cholesterol gene signature in melanoma patients treated with anti-PD-1-based immunotherapy predicted diverse clinical outcomes. Indeed, patients whose tumors were poorly infiltrated by monocytes/macrophages expressing LXR target genes showed improved survival over the course of therapy. Thus, our data support a role for (oxy)sterol metabolism in shaping monocyte-to-DC differentiation, and in tumor antigen presentation critical for responsiveness to immunotherapy. The identification of a new LXR antagonist opens new treatment avenues for cancer patients.


Subject(s)
Melanoma , Monocytes , Mice , Animals , Monocytes/metabolism , Cell Differentiation , Cholesterol/metabolism , Antigen Presentation , Dendritic Cells/metabolism , Tumor Microenvironment
5.
Eur J Immunol ; 53(5): e2149775, 2023 05.
Article in English | MEDLINE | ID: mdl-36653901

ABSTRACT

Type 1 regulatory (Tr1) T cells are currently defined all T cells with regulatory functions that lack FOXP3 expression and produce IL-10. Tr1 cells are heterogeneous, and the different reported properties of Tr1-cell populations have caused some confusion in the field. Moreover, understanding the role of Tr1 cells in immune-mediated diseases has been hampered by the lack of a lineage-defining transcription factor. Several independent studies indicated recently that the transcription factor Eomesodermin (EOMES) could act as a lineage-defining transcription factor in a population of IL-10 and IFN-γ co-producing Tr1-like cells, since EOMES directly induces IFN-γ and cytotoxicity, enhances IL-10, and antagonizes alternative T-cell fates. Here, we review the known properties of EOMES+ Tr1-like cells. They share several key characteristics with other Tr1 cells (i.e., "Tr1-like"), namely high IL-10 production, cytotoxicity, and suppressive capabilities. Notably, they also share some features with FOXP3+ Tregs, like downregulation of IL-7R and CD40L. In addition, they possess several unique, EOMES-dependent features, that is, expression of GzmK and IFN-γ, and downregulation of type-17 cytokines. Published evidence indicates that EOMES+ Tr1-like cells play key roles in graft-versus-host disease, colitis, systemic autoimmunity and in tumors. Thus, EOMES+ Tr1-like cells are key players of the adaptive immune system that are involved in several different immune-mediated diseases.


Subject(s)
Interleukin-10 , T-Lymphocytes, Regulatory , Interleukin-10/metabolism , Cell Differentiation , Forkhead Transcription Factors/metabolism , Biology
7.
Nat Mater ; 22(5): 644-655, 2023 05.
Article in English | MEDLINE | ID: mdl-36581770

ABSTRACT

The process in which locally confined epithelial malignancies progressively evolve into invasive cancers is often promoted by unjamming, a phase transition from a solid-like to a liquid-like state, which occurs in various tissues. Whether this tissue-level mechanical transition impacts phenotypes during carcinoma progression remains unclear. Here we report that the large fluctuations in cell density that accompany unjamming result in repeated mechanical deformations of cells and nuclei. This triggers a cellular mechano-protective mechanism involving an increase in nuclear size and rigidity, heterochromatin redistribution and remodelling of the perinuclear actin architecture into actin rings. The chronic strains and stresses associated with unjamming together with the reduction of Lamin B1 levels eventually result in DNA damage and nuclear envelope ruptures, with the release of cytosolic DNA that activates a cGAS-STING (cyclic GMP-AMP synthase-signalling adaptor stimulator of interferon genes)-dependent cytosolic DNA response gene program. This mechanically driven transcriptional rewiring ultimately alters the cell state, with the emergence of malignant traits, including epithelial-to-mesenchymal plasticity phenotypes and chemoresistance in invasive breast carcinoma.


Subject(s)
Actins , Neoplasms , DNA , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Cytosol/metabolism , Signal Transduction
8.
Cell Death Differ ; 29(3): 614-626, 2022 03.
Article in English | MEDLINE | ID: mdl-34845371

ABSTRACT

High Grade Serous Ovarian cancer (HGSOC) is a major unmet need in oncology, due to its precocious dissemination and the lack of meaningful human models for the investigation of disease pathogenesis in a patient-specific manner. To overcome this roadblock, we present a new method to isolate and grow single cells directly from patients' metastatic ascites, establishing the conditions for propagating them as 3D cultures that we refer to as single cell-derived metastatic ovarian cancer spheroids (sMOCS). By single cell RNA sequencing (scRNAseq) we define the cellular composition of metastatic ascites and trace its propagation in 2D and 3D culture paradigms, finding that sMOCS retain and amplify key subpopulations from the original patients' samples and recapitulate features of the original metastasis that do not emerge from classical 2D culture, including retention of individual patients' specificities. By enabling the enrichment of uniquely informative cell subpopulations from HGSOC metastasis and the clonal interrogation of their diversity at the functional and molecular level, this method provides a powerful instrument for precision oncology in ovarian cancer.


Subject(s)
Ascites , Ovarian Neoplasms , Ascites/genetics , Ascites/pathology , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/pathology , Precision Medicine , Spheroids, Cellular/pathology
9.
Eur J Immunol ; 51(12): 3243-3246, 2021 12.
Article in English | MEDLINE | ID: mdl-34528258

ABSTRACT

Ex vivo gene expression and miRNA profiling of Eomes+ Tr1-like cells suggested that they represent a differentiation stage that is intermediate between Th1-cells and cytotoxic CD4+ T-cells. Several microRNAs were downregulated in Eomes+ Tr1-like cells that might inhibit Tr1-cell differentiation. In particular, miR-92a targeted Eomes, while miR-125a inhibited IFN-g and IL-10R expression.


Subject(s)
Gene Expression Profiling , MicroRNAs/immunology , Receptors, Interleukin-10/immunology , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Humans
10.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34312224

ABSTRACT

Regulatory T cells (Tregs) play fundamental roles in maintaining peripheral tolerance to prevent autoimmunity and limit legitimate immune responses, a feature hijacked in tumor microenvironments in which the recruitment of Tregs often extinguishes immune surveillance through suppression of T-effector cell signaling and tumor cell killing. The pharmacological tuning of Treg activity without impacting on T conventional (Tconv) cell activity would likely be beneficial in the treatment of various human pathologies. PIP4K2A, 2B, and 2C constitute a family of lipid kinases that phosphorylate PtdIns5P to PtdIns(4,5)P2 They are involved in stress signaling, act as synthetic lethal targets in p53-null tumors, and in mice, the loss of PIP4K2C leads to late onset hyperinflammation. Accordingly, a human single nucleotide polymorphism (SNP) near the PIP4K2C gene is linked with susceptibility to autoimmune diseases. How PIP4Ks impact on human T cell signaling is not known. Using ex vivo human primary T cells, we found that PIP4K activity is required for Treg cell signaling and immunosuppressive activity. Genetic and pharmacological inhibition of PIP4K in Tregs reduces signaling through the PI3K, mTORC1/S6, and MAPK pathways, impairs cell proliferation, and increases activation-induced cell death while sparing Tconv. PIP4K and PI3K signaling regulate the expression of the Treg master transcriptional activator FOXP3 and the epigenetic signaling protein Ubiquitin-like containing PHD and RING finger domains 1 (UHRF1). Our studies suggest that the pharmacological inhibition of PIP4K can reprogram human Treg identity while leaving Tconv cell signaling and T-helper differentiation to largely intact potentially enhancing overall immunological activity.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , T-Lymphocytes, Regulatory/physiology , Ubiquitin-Protein Ligases/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Proliferation , Cell Survival , Cloning, Molecular , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Enzymologic/physiology , Humans , Immunosuppression Therapy , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Quinazolines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Ubiquitin-Protein Ligases/genetics
11.
Science ; 372(6542)2021 05 07.
Article in English | MEDLINE | ID: mdl-33958447

ABSTRACT

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Subject(s)
Atlases as Topic , Corpus Striatum/cytology , Corpus Striatum/embryology , Neurogenesis/genetics , RNA, Long Noncoding/genetics , Single-Cell Analysis , Transcription Factors/genetics , Fetus , GABAergic Neurons/metabolism , Humans , RNA-Seq , Transcription, Genetic
12.
Nat Immunol ; 22(6): 735-745, 2021 06.
Article in English | MEDLINE | ID: mdl-34017124

ABSTRACT

Regulatory T (Treg) cells are a barrier for tumor immunity and a target for immunotherapy. Using single-cell transcriptomics, we found that CD4+ T cells infiltrating primary and metastatic colorectal cancer and non-small-cell lung cancer are highly enriched for two subsets of comparable size and suppressor function comprising forkhead box protein P3+ Treg and eomesodermin homolog (EOMES)+ type 1 regulatory T (Tr1)-like cells also expressing granzyme K and chitinase-3-like protein 2. EOMES+ Tr1-like cells, but not Treg cells, were clonally related to effector T cells and were clonally expanded in primary and metastatic tumors, which is consistent with their proliferation and differentiation in situ. Using chitinase-3-like protein 2 as a subset signature, we found that the EOMES+ Tr1-like subset correlates with disease progression but is also associated with response to programmed cell death protein 1-targeted immunotherapy. Collectively, these findings highlight the heterogeneity of Treg cells that accumulate in primary tumors and metastases and identify a new prospective target for cancer immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Clonal Hematopoiesis/immunology , Colorectal Neoplasms/immunology , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/therapy , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation/genetics , Chemotherapy, Adjuvant/methods , Chitinases/metabolism , Colectomy , Colon/pathology , Colon/surgery , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Datasets as Topic , Disease Progression , Drug Resistance, Neoplasm/immunology , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/immunology , Granzymes/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Primary Cell Culture , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA-Seq , Single-Cell Analysis , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism
13.
Nat Commun ; 12(1): 2340, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879786

ABSTRACT

Cancer is characterized by pervasive epigenetic alterations with enhancer dysfunction orchestrating the aberrant cancer transcriptional programs and transcriptional dependencies. Here, we epigenetically characterize human colorectal cancer (CRC) using de novo chromatin state discovery on a library of different patient-derived organoids. By exploring this resource, we unveil a tumor-specific deregulated enhancerome that is cancer cell-intrinsic and independent of interpatient heterogeneity. We show that the transcriptional coactivators YAP/TAZ act as key regulators of the conserved CRC gained enhancers. The same YAP/TAZ-bound enhancers display active chromatin profiles across diverse human tumors, highlighting a pan-cancer epigenetic rewiring which at single-cell level distinguishes malignant from normal cell populations. YAP/TAZ inhibition in established tumor organoids causes extensive cell death unveiling their essential role in tumor maintenance. This work indicates a common layer of YAP/TAZ-fueled enhancer reprogramming that is key for the cancer cell state and can be exploited for the development of improved therapeutic avenues.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms/genetics , Enhancer Elements, Genetic , Epigenesis, Genetic , Trans-Activators/genetics , Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Histone Code , Humans , Models, Genetic , Organoids/metabolism , RNA-Seq , Single-Cell Analysis , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Cells, Cultured , YAP-Signaling Proteins
14.
Front Chem ; 9: 598802, 2021.
Article in English | MEDLINE | ID: mdl-33718327

ABSTRACT

The exposure to pathogens triggers the activation of adaptive immune responses through antigens bound to surface receptors of antigen presenting cells (APCs). T cell receptors (TCR) are responsible for initiating the immune response through their physical direct interaction with antigen-bound receptors on the APCs surface. The study of T cell interactions with antigens is considered of crucial importance for the comprehension of the role of immune responses in cancer growth and for the subsequent design of immunomodulating anticancer drugs. RNA sequencing experiments performed on T cells represented a major breakthrough for this branch of experimental molecular biology. Apart from the gene expression levels, the hypervariable CDR3α/ß sequences of the TCR loops can now be easily determined and modelled in the three dimensions, being the portions of TCR mainly responsible for the interaction with APC receptors. The most direct experimental method for the investigation of antigens would be based on peptide libraries, but their huge combinatorial nature, size, cost, and the difficulty of experimental fine tuning makes this approach complicated time consuming, and costly. We have implemented in silico methodology with the aim of moving from CDR3α/ß sequences to a library of potentially antigenic peptides that can be used in immunologically oriented experiments to study T cells' reactivity. To reduce the size of the library, we have verified the reproducibility of experimental benchmarks using the permutation of only six residues that can be considered representative of all ensembles of 20 natural amino acids. Such a simplified alphabet is able to correctly find the poses and chemical nature of original antigens within a small subset of ligands of potential interest. The newly generated library would have the advantage of leading to potentially antigenic ligands that would contribute to a better understanding of the chemical nature of TCR-antigen interactions. This step is crucial in the design of immunomodulators targeted towards T-cells response as well as in understanding the first principles of an immune response in several diseases, from cancer to autoimmune disorders.

15.
EMBO Mol Med ; 13(4): e12433, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33665914

ABSTRACT

MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.


Subject(s)
Methyl-CpG-Binding Protein 2 , Rett Syndrome , Animals , Brain/metabolism , Female , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/metabolism , Phenotype , Rett Syndrome/genetics
16.
Nucleic Acids Res ; 48(W1): W332-W339, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32313927

ABSTRACT

Fluorescence in situ hybridization (FISH) is a powerful single-cell technique that harnesses nucleic acid base pairing to detect the abundance and positioning of cellular RNA and DNA molecules in fixed samples. Recent technology development has paved the way to the construction of FISH probes entirely from synthetic oligonucleotides (oligos), allowing the optimization of thermodynamic properties together with the opportunity to design probes against any sequenced genome. However, comparatively little progress has been made in the development of computational tools to facilitate the oligos design, and even less has been done to extend their accessibility. OligoMiner is an open-source and modular pipeline written in Python that introduces a novel method of assessing probe specificity that employs supervised machine learning to predict probe binding specificity from genome-scale sequence alignment information. However, its use is restricted to only those people who are confident with command line interfaces because it lacks a Graphical User Interface (GUI), potentially cutting out many researchers from this technology. Here, we present OligoMinerApp (http://oligominerapp.org), a web-based application that aims to extend the OligoMiner framework through the implementation of a smart and easy-to-use GUI and the introduction of new functionalities specially designed to make effective probe mining available to everyone.


Subject(s)
In Situ Hybridization, Fluorescence/methods , Oligonucleotide Probes , Software , Genome , Internet
17.
J Clin Invest ; 130(6): 3137-3150, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32125291

ABSTRACT

The molecular mechanisms responsible for the high immunosuppressive capacity of CD4+ Tregs in tumors are not well known. High-dimensional single-cell profiling of T cells from chemotherapy-naive individuals with non-small-cell lung cancer identified the transcription factor IRF4 as specifically expressed by a subset of intratumoral CD4+ effector Tregs with superior suppressive activity. In contrast to the IRF4- counterparts, IRF4+ Tregs expressed a vast array of suppressive molecules, and their presence correlated with multiple exhausted subpopulations of T cells. Integration of transcriptomic and epigenomic data revealed that IRF4, either alone or in combination with its partner BATF, directly controlled a molecular program responsible for immunosuppression in tumors. Accordingly, deletion of Irf4 exclusively in Tregs resulted in delayed tumor growth in mice while the abundance of IRF4+ Tregs correlated with poor prognosis in patients with multiple human cancers. Thus, a common mechanism underlies immunosuppression in the tumor microenvironment irrespective of the tumor type.


Subject(s)
Cell Differentiation/immunology , Interferon Regulatory Factors/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Aged , Aged, 80 and over , Animals , Humans , Male , Mice , Middle Aged , Neoplasms/pathology , T-Lymphocytes, Regulatory/pathology
18.
Cardiovasc Res ; 116(3): 698-707, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31135876

ABSTRACT

AIMS: The pathogenetic mechanisms underlying unprovoked venous thromboembolism (uVTE) are largely unknown. In this study, we investigated the molecular mechanisms involved in uVTE pathogenesis by using ex vivo expanded endothelial colony-forming cells (ECFCs), which represent a valuable non-invasive tool for the assessment of endothelial function. METHODS AND RESULTS: We isolated and expanded ECFCs from the peripheral blood of uVTE patients and observed that these cells underwent earlier senescence and showed lower growth rate compared with ECFCs obtained from healthy donors. Through microarray expression profiling, we demonstrated that 2905 genes were differentially expressed between patients and controls. Among them, the anti-angiogenic cytokine TNF superfamily member 15 (TNFSF15) and its death-receptor TNFRSF25 were up-regulated in uVTE ECFCs, and this finding was validated by RT-qPCR. TNFSF15 up-regulation was confirmed at the protein level in ECFC supernatants, and the in vivo relevance of these findings was further corroborated by demonstrating that also the plasmatic levels of TNFSF15 are increased in uVTE patients. After proving that exogenous TNFSF15 exerts pro-apoptotic and anti-proliferative activity on control ECFCs, we demonstrated through blocking experiments that TNFSF15 up-regulation contributes to impaired survival and proliferation of uVTE ECFCs. CONCLUSION: By providing evidence that TNFSF15 impairs ECFC functions crucial to endothelial repair, and that uVTE patients have increased TNFSF15 levels both ex vivo and in vivo, the results of this study suggest that pathologic up-regulation of TNFSF15-TNFRSF25 axis may contribute to uVTE pathogenesis, and may represent the target for novel therapeutic strategies aimed at preventing recurrences in uVTE patients.


Subject(s)
Endothelial Progenitor Cells/metabolism , Endothelium, Vascular/metabolism , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Venous Thromboembolism/metabolism , Adult , Apoptosis , Case-Control Studies , Cell Proliferation , Cell Survival , Cells, Cultured , Cellular Senescence , Endothelial Progenitor Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Male , Middle Aged , Phenotype , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Venous Thromboembolism/pathology , Venous Thromboembolism/physiopathology
19.
Clin Cancer Res ; 26(6): 1372-1384, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31831554

ABSTRACT

PURPOSE: Defects in the homologous recombination (HR) repair pathway are of clinical interest due to sensitivity of HR-deficient cells to PARP inhibitors. We were interested in defining PARP vulnerability in patients with metastatic colorectal cancer (mCRC) carrying KRAS and BRAF mutations who display poor prognosis, have limited therapeutic options, and represent an unmet clinical need. EXPERIMENTAL DESIGN: We tested colorectal cancer cell lines, patient-derived organoids (PDO), and patient-derived xenografts (PDX) enriched for KRAS and BRAF mutations for sensitivity to the PARP inhibitor olaparib, and the chemotherapeutic agents oxaliplatin and 5-fluorouracil (5-FU). Genomic profiles and DNA repair proficiency of colorectal cancer models were compared with pharmacologic response. RESULTS: Thirteen of 99 (around 13%) colorectal cancer cell lines were highly sensitive to clinically active concentrations of olaparib and displayed functional deficiency in HR. Response to PARP blockade was positively correlated with sensitivity to oxaliplatin in colorectal cancer cell lines as well as patient-derived organoids. Treatment of PDXs with olaparib impaired tumor growth and maintenance therapy with PARP blockade after initial oxaliplatin response delayed disease progression in mice. CONCLUSIONS: These results indicate that a colorectal cancer subset characterized by poor prognosis and limited therapeutic options is vulnerable to PARP inhibition and suggest that PDO-based drug-screening assays can be used to identify patients with colorectal cancer likely to benefit from olaparib. As patients with mCRC almost invariably receive therapies based on oxaliplatin, "maintenance" treatment with PARP inhibitors warrants further clinical investigation.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Oxaliplatin/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Recombinational DNA Repair , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
20.
Stem Cell Reports ; 13(5): 847-861, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31607568

ABSTRACT

The regulation of the proliferation and polarity of neural progenitors is crucial for the development of the brain cortex. Animal studies have implicated glycogen synthase kinase 3 (GSK3) as a pivotal regulator of both proliferation and polarity, yet the functional relevance of its signaling for the unique features of human corticogenesis remains to be elucidated. We harnessed human cortical brain organoids to probe the longitudinal impact of GSK3 inhibition through multiple developmental stages. Chronic GSK3 inhibition increased the proliferation of neural progenitors and caused massive derangement of cortical tissue architecture. Single-cell transcriptome profiling revealed a direct impact on early neurogenesis and uncovered a selective role of GSK3 in the regulation of glutamatergic lineages and outer radial glia output. Our dissection of the GSK3-dependent transcriptional network in human corticogenesis underscores the robustness of the programs determining neuronal identity independent of tissue architecture.


Subject(s)
Cerebral Cortex/cytology , Glycogen Synthase Kinase 3/metabolism , Neurogenesis , Neurons/cytology , Organoids/cytology , Cell Line , Cell Proliferation , Cerebral Cortex/metabolism , Gene Deletion , Glycogen Synthase Kinase 3/genetics , Humans , Neurons/metabolism , Organoids/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...