Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(11)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652549

ABSTRACT

CD8+ T cell dysfunction impedes antitumor immunity in solid cancers, but the underlying mechanisms are diverse and poorly understood. Extracellular matrix (ECM) composition has been linked to impaired T cell migration and enhanced tumor progression; however, impacts of individual ECM molecules on T cell function in the tumor microenvironment (TME) are only beginning to be elucidated. Upstream regulators of aberrant ECM deposition and organization in solid tumors are equally ill-defined. Therefore, we investigated how ECM composition modulates CD8+ T cell function in undifferentiated pleomorphic sarcoma (UPS), an immunologically active desmoplastic tumor. Using an autochthonous murine model of UPS and data from multiple human patient cohorts, we discovered a multifaceted mechanism wherein the transcriptional coactivator YAP1 promotes collagen VI (COLVI) deposition in the UPS TME. In turn, COLVI induces CD8+ T cell dysfunction and immune evasion by remodeling fibrillar collagen and inhibiting T cell autophagic flux. Unexpectedly, collagen I (COLI) opposed COLVI in this setting, promoting CD8+ T cell function and acting as a tumor suppressor. Thus, CD8+ T cell responses in sarcoma depend on oncogene-mediated ECM composition and remodeling.


Subject(s)
CD8-Positive T-Lymphocytes , Extracellular Matrix , Sarcoma , Tumor Microenvironment , YAP-Signaling Proteins , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Animals , Tumor Microenvironment/immunology , Mice , YAP-Signaling Proteins/immunology , YAP-Signaling Proteins/genetics , Humans , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Sarcoma/immunology , Sarcoma/pathology , Sarcoma/genetics , Sarcoma/metabolism , Collagen Type VI/genetics , Collagen Type VI/immunology , Collagen Type VI/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/immunology , Oncogenes , Neoplasm Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type I/immunology
3.
Cell Rep ; 34(13): 108917, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789113

ABSTRACT

Tumor-associated macrophages (TAMs) play an important role in tumor immunity and comprise of subsets that have distinct phenotype, function, and ontology. Transcriptomic analyses of human medulloblastoma, the most common malignant pediatric brain cancer, showed that medulloblastomas (MBs) with activated sonic hedgehog signaling (SHH-MB) have significantly more TAMs than other MB subtypes. Therefore, we examined MB-associated TAMs by single-cell RNA sequencing of autochthonous murine SHH-MB at steady state and under two distinct treatment modalities: molecular-targeted inhibitor and radiation. Our analyses reveal significant TAM heterogeneity, identify markers of ontologically distinct TAM subsets, and show the impact of brain microenvironment on the differentiation of tumor-infiltrating monocytes. TAM composition undergoes dramatic changes with treatment and differs significantly between molecular-targeted and radiation therapy. We identify an immunosuppressive monocyte-derived TAM subset that emerges with radiation therapy and demonstrate its role in regulating T cell and neutrophil infiltration in MB.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Hedgehog Proteins/metabolism , Macrophages/metabolism , Macrophages/pathology , Medulloblastoma/pathology , Medulloblastoma/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/immunology , Genetic Markers , Humans , Medulloblastoma/genetics , Medulloblastoma/immunology , Mice , Microglia/pathology , Monocytes/pathology , Single-Cell Analysis , Transcription, Genetic , Tumor Microenvironment
4.
STAR Protoc ; 1(3): 100188, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377082

ABSTRACT

The regulation of monocyte differentiation in the tumor microenvironment is of significant interest to tumor immunologists. Monocytes injected into the circulation may not track into tumors in sufficient numbers, making intratumoral injections a preferred experimental approach. Monocyte enrichment with antibody-based positive selection may activate downstream signaling, while cell sorters expose monocytes to mechanical stress. Here, we describe an approach of intratumoral monocyte transfer that circumvents these limitations by using negative selection and fluorescent reporter mice. For complete details on the use and execution of this protocol, please refer to Devalaraja et al. (2020).


Subject(s)
Cell Differentiation , Monocytes/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Female , Male , Mice, Inbred C57BL , Microspheres
5.
Cell Rep ; 31(13): 107825, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610126

ABSTRACT

Activated macrophages must carefully calibrate their inflammatory responses to balance efficient pathogen control with inflammation-mediated tissue damage, but the molecular underpinnings of this "balancing act" remain unclear. Using genetically engineered mouse models and primary macrophage cultures, we show that Toll-like receptor (TLR) signaling induces the expression of the transcription factor Spic selectively in patrolling monocytes and tissue macrophages by a nuclear factor κB (NF-κB)-dependent mechanism. Functionally, Spic downregulates pro-inflammatory cytokines and promotes iron efflux by regulating ferroportin expression in activated macrophages. Notably, interferon-gamma blocks Spic expression in a STAT1-dependent manner. High levels of interferon-gamma are indicative of ongoing infection, and in its absence, activated macrophages appear to engage a "default" Spic-dependent anti-inflammatory pathway. We also provide evidence for the engagement of this pathway in sterile inflammation. Taken together, our findings uncover a pathway wherein counter-regulation of Spic by NF-κB and STATs attune inflammatory responses and iron metabolism in macrophages.


Subject(s)
DNA-Binding Proteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Iron/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Biological Transport , Down-Regulation/genetics , Female , Heme/metabolism , Interferon-gamma/metabolism , Ligands , Macrophage Activation , Male , Mice, Inbred C57BL , Monocytes/metabolism , Toll-Like Receptors/metabolism
6.
Cell ; 180(6): 1098-1114.e16, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169218

ABSTRACT

The immunosuppressive tumor microenvironment (TME) is a major barrier to immunotherapy. Within solid tumors, why monocytes preferentially differentiate into immunosuppressive tumor-associated macrophages (TAMs) rather than immunostimulatory dendritic cells (DCs) remains unclear. Using multiple murine sarcoma models, we find that the TME induces tumor cells to produce retinoic acid (RA), which polarizes intratumoral monocyte differentiation toward TAMs and away from DCs via suppression of DC-promoting transcription factor Irf4. Genetic inhibition of RA production in tumor cells or pharmacologic inhibition of RA signaling within TME increases stimulatory monocyte-derived cells, enhances T cell-dependent anti-tumor immunity, and synergizes with immune checkpoint blockade. Furthermore, an RA-responsive gene signature in human monocytes correlates with an immunosuppressive TME in multiple human tumors. RA has been considered as an anti-cancer agent, whereas our work demonstrates its tumorigenic capability via myeloid-mediated immune suppression and provides proof of concept for targeting this pathway for tumor immunotherapy.


Subject(s)
Monocytes/immunology , Tretinoin/metabolism , Tumor Microenvironment/immunology , Animals , Carcinogenesis/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Immunosuppression Therapy/methods , Immunotherapy/methods , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism
7.
Mol Cancer Res ; 18(4): 560-573, 2020 04.
Article in English | MEDLINE | ID: mdl-31988250

ABSTRACT

High-grade sarcomas are metastatic and pose a serious threat to patient survival. Undifferentiated pleomorphic sarcoma (UPS) is a particularly dangerous and relatively common sarcoma subtype diagnosed in adults. UPS contains large quantities of extracellular matrix (ECM) including hyaluronic acid (HA), which is linked to metastatic potential. Consistent with these observations, expression of the HA receptor, hyaluronan-mediated motility receptor (HMMR/RHAMM), is tightly controlled in normal tissues and upregulated in UPS. Moreover, HMMR expression correlates with poor clinical outcome in these patients. Deregulation of the tumor-suppressive Hippo pathway is also linked to poor outcome in these patients. YAP1, the transcriptional regulator and central effector of Hippo pathway, is aberrantly stabilized in UPS and was recently shown to control RHAMM expression in breast cancer cells. Interestingly, both YAP1 and RHAMM are linked to TGFß signaling. Therefore, we investigated crosstalk between YAP1 and TGFß resulting in enhanced RHAMM-mediated cell migration and invasion. We observed that HMMR expression is under the control of both YAP1 and TGFß and can be effectively targeted with small-molecule approaches that inhibit these pathways. Furthermore, we found that RHAMM expression promotes tumor cell proliferation and migration/invasion. To test these observations in a robust and quantifiable in vivo system, we developed a zebrafish xenograft assay of metastasis, which is complimentary to our murine studies. Importantly, pharmacologic inhibition of the TGFß-YAP1-RHAMM axis prevents vascular migration of tumor cells to distant sites. IMPLICATIONS: These studies reveal key metastatic signaling mechanisms and highlight potential approaches to prevent metastatic dissemination in UPS.YAP1 and TGFß cooperatively enhance proliferation and migration/invasion of UPS and fibrosarcomas.


Subject(s)
Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/metabolism , Protein Serine-Threonine Kinases/metabolism , Sarcoma/metabolism , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Fibrosarcoma , HCT116 Cells , HEK293 Cells , Hippo Signaling Pathway , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Sarcoma/pathology , Transcription Factors/metabolism , YAP-Signaling Proteins , Zebrafish
8.
J Bone Miner Res ; 34(10): 1780-1788, 2019 10.
Article in English | MEDLINE | ID: mdl-31441962

ABSTRACT

Primary malignant bone tumors are rare, occur in all age groups, and include distinct entities such as osteosarcoma, Ewing sarcoma, and chondrosarcoma. Traditional treatment with some combination of chemotherapy, surgery, and radiation has reached the limit of efficacy, with substantial room for improvement in patient outcome. Furthermore, genomic characterization of these tumors reveals a paucity of actionable molecular targets. Against this backdrop, recent advances in cancer immunotherapy represent a silver lining in the treatment of primary bone cancer. Major strategies in cancer immunotherapy include stimulating naturally occurring anti-tumor T cells and adoptive transfer of tumor-specific cytotoxic T cells. Chimeric antigen receptor T cells (CAR-T cells) belong to the latter strategy and are an impressive application of both insights into T cell biology and advances in genetic engineering. In this review, we briefly describe the CAR-T approach and discuss its applications in primary bone tumors. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/therapeutic use , Bone Neoplasms/immunology , Humans , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism
9.
Front Immunol ; 8: 33, 2017.
Article in English | MEDLINE | ID: mdl-28167947

ABSTRACT

Erythroid function and development is intimately linked to macrophages. The primary function of erythrocytes is oxygen delivery, which is mediated by iron-containing hemoglobin. The major source of this iron is a recycling pathway where macrophages scavenge old and damaged erythrocytes to release iron contained within the heme moiety. Macrophages also promote erythropoiesis by providing a supportive niche in the bone marrow as an integral component of "erythorblastic islands." Importantly, inflammation leads to alterations in iron handling by macrophages with significant impact on iron homeostasis and erythropoiesis. The importance of macrophages in erythropoiesis and iron homeostasis is well established and has been extensively reviewed. However, this developmental relationship is not one way, and erythrocytes can also regulate macrophage development and function. Erythrocyte-derived heme can induce the development of iron-recycling macrophages from monocytes, engage pattern recognition receptors to activate macrophages, and act as ligand for specific nuclear receptors to modulate macrophage function. Here, we discuss the role of heme as a signaling molecule impacting macrophage homeostasis. We will review these actions of heme within the framework of our current understanding of the role of micro-environmental factors in macrophage development and function.

10.
J Biol Chem ; 292(8): 3420-3432, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28069811

ABSTRACT

Maintenance of glucose homeostasis is essential for normal physiology. Deviation from normal glucose levels, in either direction, increases susceptibility to serious medical complications such as hypoglycemia and diabetes. Maintenance of glucose homeostasis is achieved via functional interactions among various organs: liver, skeletal muscle, adipose tissue, brain, and the endocrine pancreas. The liver is the primary site of endogenous glucose production, especially during states of prolonged fasting. However, enhanced gluconeogenesis is also a signature feature of type 2 diabetes (T2D). Thus, elucidating the signaling pathways that regulate hepatic gluconeogenesis would allow better insight into the process of normal endogenous glucose production as well as how this process is impaired in T2D. Here we demonstrate that the TGF-ß1/Smad3 signaling pathway promotes hepatic gluconeogenesis, both upon prolonged fasting and during T2D. In contrast, genetic and pharmacological inhibition of TGF-ß1/Smad3 signals suppressed endogenous glucose production. TGF-ß1 and Smad3 signals achieved this effect via the targeting of key regulators of hepatic gluconeogenesis, protein phosphatase 2A (PP2A), AMP-activated protein kinase (AMPK), and FoxO1 proteins. Specifically, TGF-ß1 signaling suppressed the LKB1-AMPK axis, thereby facilitating the nuclear translocation of FoxO1 and activation of key gluconeogenic genes, glucose-6-phosphatase and phosphoenolpyruvate carboxykinase. These findings underscore an important role of TGF-ß1/Smad3 signaling in hepatic gluconeogenesis, both in normal physiology and in the pathophysiology of metabolic diseases such as diabetes, and are thus of significant medical relevance.


Subject(s)
Gluconeogenesis , Liver/metabolism , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Female , Forkhead Box Protein O1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Protein Phosphatase 2/metabolism
11.
Front Cell Dev Biol ; 3: 71, 2015.
Article in English | MEDLINE | ID: mdl-26618158

ABSTRACT

The patent eligibility of stem cells-particularly those derived from human embryos-has long been under debate in both the scientific and legal communities. On the basis of moral grounds, the European Patent Office (EPO) has refrained from granting patents for stem cells obtained through the destruction of human embryos. On the contrary, the United States Patent and Trademark Office (USPTO) has historically granted patents regarding the isolation and use of human embryonic and other stem cells. To date, these US patents remain valid despite an increasing onslaught of challenges in court. However, recent precedents established in US courts significantly narrow the scope of patent eligibility within biotechnology. This article compares the implications of recent legal changes on stem cell patent eligibility between the EU and US.

12.
Food Res Int ; 44(7): 1856-1865, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21857774

ABSTRACT

The prevalence and severity of obesity, type 2-diabetes, and the resultant metabolic syndrome are rapidly increasing. As successful preventive and therapeutic strategies for these life-threatening health ailments often come with adverse side effects, nutritional elements are widely used in many countries as preventive therapies to prevent or manage metabolic syndrome. Fruits are important dietary components, and contain various bioactive constituents. Many of these constituents have been proven to be useful to manage and treat various chronic diseases such as diabetes, obesity, cancer and cardiovascular diseases. Although exotic fruits are understudied throughout the world due to their limited regional presence, many studies reveal their potent ability to ameliorate metabolic derangements and the resultant conditions i.e. diabetes and obesity. The aim of this article is to review the role of exotic fruits and their constituents in the regulation of metabolic functions, which can beneficially alter diabetes and obesity pathophysiology.

SELECTION OF CITATIONS
SEARCH DETAIL
...