Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 167: 115592, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37778272

ABSTRACT

INTRODUCTION: Glycogen synthase kinase 3 (GSK-3) has been proposed as a novel cancer target due to its regulating role in both tumor and immune cells. However, the connection between GSK-3 and immunoevasive contexture, including tumor budding (TB) has not been previously examined. METHODS: we investigated the expression levels of total GSK-3 as well as its isoforms (GSK-3ß and GSK-3α) and examined their potential correlation with TB grade and the programmed cell death-ligand 1 (PD-L1) in colorectal cancer (CRC) tumor samples. Additionally, we compared the efficacy of GSK-3-inhibition with PD-1/PD-L1 blockade in humanized patient-derived (PDXs) xenografts models of high-grade TB CRC. RESULTS: we show that high-grade (BD3) TB CRC is associated with elevated expression levels of total GSK-3, specifically the GSK-3ß isoform, along with increased expression of PD-L1 in tumor cells. Moreover, we define an improved risk stratification of CRC patients based on the presence of GSK-3+/PD-L1+/BD3 tumors, which are associated with a worse prognosis. Significantly, in contrast to the PD-L1/PD-1 blockade approach, the inhibition GSK-3 demonstrated a remarkable enhancement in the antitumor response. This was achieved through the reduction of tumor buds via necrosis and apoptosis pathways, along with a notable increase of activated tumor-infiltrating CD8+ T cells, NK cells, and CD4- CD8- T cells. CONCLUSIONS: our study provides compelling evidence for the clinical significance of GSK-3 expression and TB grade in risk stratification of CRC patients. Moreover, our findings strongly support GSK-3 inhibition as an effective therapy specifically targeting high-grade TB in CRC.


Subject(s)
CD8-Positive T-Lymphocytes , Colorectal Neoplasms , Humans , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinase 3 beta , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Clinical Relevance , Colorectal Neoplasms/pathology
2.
PLoS One ; 16(6): e0251731, 2021.
Article in English | MEDLINE | ID: mdl-34181666

ABSTRACT

Immunotherapy using checkpoint blockade (ICB) with antibodies such as anti-PD-1 has revolutionised the treatment of many cancers. Despite its use to treat COVID-19 patients and autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis, the effect of hydroxychloroquine (HCQ) on cancer immunotherapy has not been examined. In this study, remarkably, we find that HCQ alone, or in combination with azithromycin (AZ), at doses used to treat patients, decreased the therapeutic benefit of anti-PD-1 in cancer immunotherapy. No deleterious effect was seen on untreated tumors. Mechanistically, HCQ and HCQ/AZ inhibited PD-L1 expression on tumor cells, while specifically targeting the anti-PD-1 induced increase in progenitor CD8+CD44+PD-1+TCF1+ tumor infiltrating T cells (TILs) and the generation of CD8+CD44+PD-1+ effectors. Surprisingly, it also impaired the appearance of a subset of terminally exhausted CD8+ TILs. No effect was seen on the presence of CD4+ T cells, FoxP3+ regulatory T cells (Tregs), thymic subsets, B cells, antibody production, myeloid cells, or the vasculature of mice. This study indicates for the first time that HCQ and HCQ/AZ negatively impact the ability of anti-PD-1 checkpoint blockade to promote tumor rejection.


Subject(s)
Hydroxychloroquine/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Programmed Cell Death 1 Receptor/immunology , Animals , Azithromycin/pharmacology , Cell Line, Tumor , Drug Antagonism , Immune Checkpoint Inhibitors/immunology , Melanoma/pathology , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Semin Immunol ; 42: 101295, 2019 04.
Article in English | MEDLINE | ID: mdl-31604533

ABSTRACT

The past few years have witnessed exciting progress in the application of immune check-point blockade (ICB) for the treatment of various human cancers. ICB was first used against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to demonstrate durable anti-tumor responses followed by ICB against programmed cell death-1 (PD-1) or its ligand, PD-L1. Present approaches involve the use of combinations of blocking antibodies against CTLA-4, PD-1 and other inhibitory receptors (IRs) such as TIM3, TIGIT and LAG3. Despite this success, most patients are not cured by ICB therapy and there are limitations to the use of antibodies including cost, tumor penetration, the accessibility of receptors, and clearance from the cell surface as well as inflammatory and autoimmune complications. Recently, we demonstrated that the down-regulation or inhibition of glycogen synthase kinase 3 (GSK-3) down-regulates PD-1 expression in infectious diseases and cancer (Taylor et al., 2016 Immunity 44, 274-86; 2018 Cancer Research 78, 706-717; Krueger and Rudd 2018 Immunity 46, 529-531). In this Review, we outline the use of small molecule inhibitors (SMIs) that target intracellular pathways for co-receptor blockade in cancer immunotherapy.


Subject(s)
Glycogen Synthase Kinase 3/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors
4.
Mol Cancer Ther ; 17(4): 869-882, 2018 04.
Article in English | MEDLINE | ID: mdl-29483207

ABSTRACT

A recurring historic finding in cancer drug development is encouraging antitumor effects observed in tumor-bearing mice that fail to translate into the clinic. An intriguing exception to this pattern is immune checkpoint therapy, as the sustained tumor regressions observed in subsets of cancer patients are rare in mice. Reasoning that this may be due in part to relatively low mutational loads of mouse tumors, we mutagenized transplantable mouse tumor cell lines EMT-6/P, B16F1, RENCA, CT26, and MC38 in vitro with methylnitro-nitrosoguanidine (MNNG) or ethylmethane sulfonate (EMS) and tested their responsiveness to PD-L1 blockade. Exome sequencing confirmed an increase in somatic mutations by mutagen treatment, an effect mimicked in EMT-6 variants chronically exposed in vivo to cisplatin or cyclophosphamide. Certain mutagenized variants of B16F1, EMT-6/P, CT26, and MC38 (but not RENCA) were more immunogenic than their parents, yet anti-PD-L1 sensitization developed only in some EMT-6/P and B16F1 variants. Treatment response patterns corresponded with changes in immune cell infiltration and especially increases in CD8+ T cells. Chronically cisplatin-exposed EMT-6 variants were also more responsive to anti-PD-L1 therapy. Although tumor PD-L1 expression was upregulated in in vivo chemotherapy-exposed variants, PD-L1 expression levels were not consistently associated with anti-PD-L1 treatment activity across mutagenized or chemotherapy-exposed variants. In summary, mutagenized and more immunogenic mouse tumors were not universally sensitized to PD-L1 blockade. Chemically mutagenized variants may be useful to evaluate the impact of immunologically "hot" or "cold" tumors with a high mutational load, to which certain chemotherapy agents may contribute, on immunotherapy outcomes. Mol Cancer Ther; 17(4); 869-82. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Ethyl Methanesulfonate/toxicity , Mammary Neoplasms, Experimental/genetics , Melanoma, Experimental/genetics , Methylnitronitrosoguanidine/toxicity , Mutation , Animals , Apoptosis , Cell Proliferation , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/chemically induced , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured
5.
Immunity ; 46(4): 529-531, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423334

ABSTRACT

The identity of PD-1 dependency on other receptors and signaling has been unclear. In a recent issue of Science, Hui et al. (2017) and Kamphorst et al. (2017) now show that CD28 expression is a target of PD-1-associated phosphatases and is needed for T cell expansion in anti-PD-1 immunotherapy.


Subject(s)
CD28 Antigens , T-Lymphocytes , Cell Cycle , Humans , Immunotherapy , Signal Transduction
6.
Nat Commun ; 8: 13991, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071661

ABSTRACT

Formation of organ-specific vasculatures requires cross-talk between developing tissue and specialized endothelial cells. Here we show how developing zebrafish spinal cord neurons coordinate vessel growth through balancing of neuron-derived Vegfaa, with neuronal sFlt1 restricting Vegfaa-Kdrl mediated angiogenesis at the neurovascular interface. Neuron-specific loss of flt1 or increased neuronal vegfaa expression promotes angiogenesis and peri-neural tube vascular network formation. Combining loss of neuronal flt1 with gain of vegfaa promotes sprout invasion into the neural tube. On loss of neuronal flt1, ectopic sprouts emanate from veins involving special angiogenic cell behaviours including nuclear positioning and a molecular signature distinct from primary arterial or secondary venous sprouting. Manipulation of arteriovenous identity or Notch signalling established that ectopic sprouting in flt1 mutants requires venous endothelium. Conceptually, our data suggest that spinal cord vascularization proceeds from veins involving two-tiered regulation of neuronal sFlt1 and Vegfaa via a novel sprouting mode.


Subject(s)
Neurons/physiology , Spinal Cord/embryology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Veins/embryology , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Embryo, Nonmammalian/cytology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , Mutation , Neovascularization, Physiologic , Receptors, Notch/genetics , Receptors, Notch/metabolism , Spinal Cord/blood supply , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Veins/metabolism , Zebrafish Proteins/genetics
7.
Article in English | MEDLINE | ID: mdl-31544130

ABSTRACT

T-cell activation is mediated by a combination of signals from the antigen receptor (TCR) and co-receptors such as CD28, cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed cell death antigen 1 (PD-1), CD28H and others. Each is a member of the CD28 receptor gene family. CD28 sends positive signals that promote T-cell responses, while CTLA-4 and PD-1 limit responses. It is the balance between these positive and negative signals that determines the amplitude and level of T-cell responses. The regulatory role of other family members is also becoming the focus of increasing interest. The function of certain CD28 family members such as CTLA-4 and PD-1 is dependent the expression of CD28. Together, these findings have important implications in generation of immune responses and the application of anti-receptor blocking reagents in immunotherapy.

8.
Nature ; 495(7441): 333-8, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23446348

ABSTRACT

Circular RNAs (circRNAs) in animals are an enigmatic class of RNA with unknown function. To explore circRNAs systematically, we sequenced and computationally analysed human, mouse and nematode RNA. We detected thousands of well-expressed, stable circRNAs, often showing tissue/developmental-stage-specific expression. Sequence analysis indicated important regulatory functions for circRNAs. We found that a human circRNA, antisense to the cerebellar degeneration-related protein 1 transcript (CDR1as), is densely bound by microRNA (miRNA) effector complexes and harbours 63 conserved binding sites for the ancient miRNA miR-7. Further analyses indicated that CDR1as functions to bind miR-7 in neuronal tissues. Human CDR1as expression in zebrafish impaired midbrain development, similar to knocking down miR-7, suggesting that CDR1as is a miRNA antagonist with a miRNA-binding capacity ten times higher than any other known transcript. Together, our data provide evidence that circRNAs form a large class of post-transcriptional regulators. Numerous circRNAs form by head-to-tail splicing of exons, suggesting previously unrecognized regulatory potential of coding sequences.


Subject(s)
Gene Expression Regulation , RNA/metabolism , Animals , Autoantigens/genetics , Autoantigens/metabolism , Binding Sites , Brain/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line , Conserved Sequence , Female , HEK293 Cells , Humans , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA/genetics , RNA, Circular , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
9.
Int J Dev Biol ; 55(4-5): 419-29, 2011.
Article in English | MEDLINE | ID: mdl-21858767

ABSTRACT

The circulatory system is the first organ system that develops during embryogenesis, and is essential for embryo viability and survival. Crucial for developing a functional vasculature are the specification of arterial-venous identity in vessels and the formation of a hierarchical branched vascular network. Sprouting angiogenesis, intussusception, and flow driven remodeling events collectively contribute to establishing the vascular architecture. At the molecular level, arterial-venous identity and branching are regulated by genetically hardwired mechanisms involving Notch, vascular endothelial growth factor and neural guidance molecule signaling pathways, modulated by hemodynamic factors. MicroRNAs are small, non-coding RNAs that act as silencers to fine-tune the gene expression profile. MicroRNAs are known to influence cell fate decisions, and microRNA expression can be controlled by blood flow, thus placing microRNAs potentially at the center of the genetic cascades regulating vascular differentiation. In the present review, we summarize current progress regarding microRNA functions in blood vessel development with an emphasis on studies performed in zebrafish and mouse models.


Subject(s)
Blood Vessels/growth & development , MicroRNAs/physiology , Neovascularization, Physiologic , Regional Blood Flow/physiology , Animals , Endothelial Cells/physiology , Gene Expression Profiling , Mice , MicroRNAs/genetics , Myocytes, Smooth Muscle/physiology , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Zebrafish
10.
Development ; 138(10): 1935-45, 2011 May.
Article in English | MEDLINE | ID: mdl-21471154

ABSTRACT

Endodermal organogenesis requires a precise orchestration of cell fate specification and cell movements, collectively coordinating organ size and shape. In Caenorhabditis elegans, uncoordinated-53 (unc-53) encodes a neural guidance molecule that directs axonal growth. One of the vertebrate homologs of unc-53 is neuron navigator 3 (Nav3). Here, we identified a novel vertebrate neuron navigator 3 isoform in zebrafish, nav3a, and we provide genetic evidence in loss- and gain-of-function experiments showing its functional role in endodermal organogenesis during zebrafish embryogenesis. In zebrafish embryos, nav3a expression was initiated at 22 hpf in the gut endoderm and at 40 hpf expanded to the newly formed liver bud. Endodermal nav3a expression was controlled by Wnt2bb signaling and was independent of FGF and BMP signaling. Morpholino-mediated knockdown of nav3a resulted in a significantly reduced liver size, and impaired development of pancreas and swim bladder. In vivo time-lapse imaging of liver development in nav3a morphants revealed a failure of hepatoblast movement out from the gut endoderm during the liver budding stage, with hepatoblasts being retained in the intestinal endoderm. In hepatocytes in vitro, nav3a acts as a positive modulator of actin assembly in lamellipodia and filipodia extensions, allowing cellular movement. Knockdown of nav3a in vitro impeded hepatocyte movement. Endodermal-specific overexpression of nav3a in vivo resulted in additional ectopic endodermal budding beyond the normal liver and pancreatic budding sites. We conclude that nav3a is required for directing endodermal organogenesis involving coordination of endodermal cell behavior.


Subject(s)
Liver/embryology , Liver/metabolism , Nerve Tissue Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , DNA Primers/genetics , Endoderm/embryology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , In Situ Hybridization , Intestinal Mucosa/metabolism , Intestines/embryology , Liver/abnormalities , Liver/innervation , Models, Biological , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Organogenesis/physiology , RNA, Antisense/genetics , Signal Transduction , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
11.
Development ; 138(10): 2111-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21521739

ABSTRACT

Endothelial tip cells guide angiogenic sprouts by exploring the local environment for guidance cues such as vascular endothelial growth factor (VegfA). Here we present Flt1 (Vegf receptor 1) loss- and gain-of-function data in zebrafish showing that Flt1 regulates tip cell formation and arterial branching morphogenesis. Zebrafish embryos expressed soluble Flt1 (sFlt1) and membrane-bound Flt1 (mFlt1). In Tg(flt1(BAC):yfp) × Tg(kdrl:ras-cherry)(s916) embryos, flt1:yfp was expressed in tip, stalk and base cells of segmental artery sprouts and overlapped with kdrl:cherry expression in these domains. flt1 morphants showed increased tip cell numbers, enhanced angiogenic behavior and hyperbranching of segmental artery sprouts. The additional arterial branches developed into functional vessels carrying blood flow. In support of a functional role for the extracellular VEGF-binding domain of Flt1, overexpression of sflt1 or mflt1 rescued aberrant branching in flt1 morphants, and overexpression of sflt1 or mflt1 in controls resulted in short arterial sprouts with reduced numbers of filopodia. flt1 morphants showed reduced expression of Notch receptors and of the Notch downstream target efnb2a, and ectopic expression of flt4 in arteries, consistent with loss of Notch signaling. Conditional overexpression of the notch1a intracellular cleaved domain in flt1 morphants restored segmental artery patterning. The developing nervous system of the trunk contributed to the distribution of Flt1, and the loss of flt1 affected neurons. Thus, Flt1 acts in a Notch-dependent manner as a negative regulator of tip cell differentiation and branching. Flt1 distribution may be fine-tuned, involving interactions with the developing nervous system.


Subject(s)
Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-1/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Blood Vessels/embryology , Blood Vessels/metabolism , Cell Differentiation , Gene Expression Regulation, Developmental , Macrophages/metabolism , Nervous System/embryology , Nervous System/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Receptors, Notch/metabolism , Signal Transduction , Solubility , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...