Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 559(7714): 363-369, 2018 07.
Article in English | MEDLINE | ID: mdl-29950727

ABSTRACT

Patients with prostate cancer frequently show resistance to androgen-deprivation therapy, a condition known as castration-resistant prostate cancer (CRPC). Acquiring a better understanding of the mechanisms that control the development of CRPC remains an unmet clinical need. The well-established dependency of cancer cells on the tumour microenvironment indicates that the microenvironment might control the emergence of CRPC. Here we identify IL-23 produced by myeloid-derived suppressor cells (MDSCs) as a driver of CRPC in mice and patients with CRPC. Mechanistically, IL-23 secreted by MDSCs can activate the androgen receptor pathway in prostate tumour cells, promoting cell survival and proliferation in androgen-deprived conditions. Intra-tumour MDSC infiltration and IL-23 concentration are increased in blood and tumour samples from patients with CRPC. Antibody-mediated inactivation of IL-23 restored sensitivity to androgen-deprivation therapy in mice. Taken together, these results reveal that MDSCs promote CRPC by acting in a non-cell autonomous manner. Treatments that block IL-23 can oppose MDSC-mediated resistance to castration in prostate cancer and synergize with standard therapies.


Subject(s)
Interleukin-23/antagonists & inhibitors , Interleukin-23/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Androgens/deficiency , Animals , Benzamides , Cell Proliferation , Cell Survival , Humans , Interleukin-23/blood , Interleukin-23/immunology , Male , Mice , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/immunology , Nitriles , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Receptors, Interleukin/metabolism , Signal Transduction
2.
Int J Mol Sci ; 24(11)2023 May 30.
Article in English | MEDLINE | ID: mdl-37298470

ABSTRACT

Natural killer cells (NK) are innate lymphocytes endowed with the ability to recognize and kill cancer cells. Consequently, adoptive transfer of autologous or allogeneic NK cells represents a novel opportunity in cancer treatment that is currently under clinical investigation. However, cancer renders NK cells dysfunctional, thus restraining the efficacy of cell therapies. Importantly, extensive effort has been employed to investigate the mechanisms that restrain NK cell anti-tumor function, and the results have offered forthcoming solutions to improve the efficiency of NK cell-based therapies. The present review will introduce the origin and features of NK cells, summarize the mechanisms of action and causes of dysfunction of NK cells in cancer, and frame NK cells in the tumoral microenvironment and in the context of immunotherapies. Finally, we will discuss therapeutic potential and current limitations of NK cell adoptive transfer in tumors.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Neoplasms/pathology , Killer Cells, Natural/pathology , Immunotherapy, Adoptive/methods , Immunotherapy/methods , Adoptive Transfer , Tumor Microenvironment
3.
J Hepatol ; 77(5): 1359-1372, 2022 11.
Article in English | MEDLINE | ID: mdl-35738508

ABSTRACT

BACKGROUND & AIMS: The landscape and function of the immune infiltrate of intrahepatic cholangiocarcinoma (iCCA), a rare, yet aggressive tumor of the biliary tract, remains poorly characterized, limiting development of successful immunotherapies. Herein, we aimed to define the molecular characteristics of tumor-infiltrating leukocytes with a special focus on CD4+ regulatory T cells (Tregs). METHODS: We used high-dimensional single-cell technologies to characterize the T-cell and myeloid compartments of iCCA tissues, comparing these with their tumor-free peritumoral and circulating counterparts. We further used genomics and cellular assays to define the iCCA-specific role of a novel transcription factor, mesenchyme homeobox 1 (MEOX1), in Treg biology. RESULTS: We found poor infiltration of putative tumor-specific CD39+ CD8+ T cells accompanied by abundant infiltration of hyperactivated CD4+ Tregs. Single-cell RNA-sequencing identified an altered network of transcription factors in iCCA-infiltrating compared to peritumoral T cells, suggesting reduced effector functions by tumor-infiltrating CD8+ T cells and enhanced immunosuppression by CD4+ Tregs. Specifically, we found that expression of MEOX1 was highly enriched in tumor-infiltrating Tregs, and demonstrated that MEOX1 overexpression is sufficient to reprogram circulating Tregs to acquire the transcriptional and epigenetic landscape of tumor-infiltrating Tregs. Accordingly, enrichment of the MEOX1-dependent gene program in Tregs was strongly associated with poor prognosis in a large cohort of patients with iCCA. CONCLUSIONS: We observed abundant infiltration of hyperactivated CD4+ Tregs in iCCA tumors along with reduced CD8+ T-cell effector functions. Interfering with hyperactivated Tregs should be explored as an approach to enhance antitumor immunity in iCCA. LAY SUMMARY: Immune cells have the potential to slow or halt the progression of tumors. However, some tumors, such as intrahepatic cholangiocarcinoma, are associated with very limited immune responses (and infiltration of cancer-targeting immune cells). Herein, we show that a specific population of regulatory T cells (a type of immune cell that actually suppresses the immune response) are hyperactivated in intrahepatic cholangiocarcinoma. Targeting these cells could enable cancer-targeting immune cells to act more effectively and should be looked at as a potential therapeutic approach to this aggressive cancer type.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , RNA/metabolism , T-Lymphocytes, Regulatory , Transcription Factors/metabolism , Tumor Microenvironment , Single-Cell Analysis
4.
Nature ; 515(7525): 134-7, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25156255

ABSTRACT

Aberrant activation of oncogenes or loss of tumour suppressor genes opposes malignant transformation by triggering a stable arrest in cell growth, which is termed cellular senescence. This process is finely tuned by both cell-autonomous and non-cell-autonomous mechanisms that regulate the entry of tumour cells to senescence. Whether tumour-infiltrating immune cells can oppose senescence is unknown. Here we show that at the onset of senescence, PTEN null prostate tumours in mice are massively infiltrated by a population of CD11b(+)Gr-1(+) myeloid cells that protect a fraction of proliferating tumour cells from senescence, thus sustaining tumour growth. Mechanistically, we found that Gr-1(+) cells antagonize senescence in a paracrine manner by interfering with the senescence-associated secretory phenotype of the tumour through the secretion of interleukin-1 receptor antagonist (IL-1RA). Strikingly, Pten-loss-induced cellular senescence was enhanced in vivo when Il1ra knockout myeloid cells were adoptively transferred to PTEN null mice. Therapeutically, docetaxel-induced senescence and efficacy were higher in PTEN null tumours when the percentage of tumour-infiltrating CD11b(+)Gr-1(+) myeloid cells was reduced using an antagonist of CXC chemokine receptor 2 (CXCR2). Taken together, our findings identify a novel non-cell-autonomous network, established by innate immunity, that controls senescence evasion and chemoresistance. Targeting this network provides novel opportunities for cancer therapy.


Subject(s)
Cell Movement , Cellular Senescence , Myeloid Cells/cytology , Myeloid Cells/metabolism , Prostatic Neoplasms/pathology , Receptors, Chemokine/metabolism , Animals , Cellular Senescence/drug effects , Disease Progression , Docetaxel , Drug Resistance, Neoplasm , Humans , Immunity, Innate , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Male , Mice , Myeloid Cells/transplantation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Taxoids/pharmacology , Tumor Escape , Tumor Microenvironment
5.
Immunology ; 146(2): 251-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26095162

ABSTRACT

The p38 mitogen-activated protein kinase cascade is required for the induction of a T helper type 17 (Th17) -mediated autoimmune response, which underlies the development and progression of several autoimmune diseases, such as experimental autoimmune encephalomyelitis, the animal model of multiple sclerosis (MS). However, the contribution of p38 phosphorylation to human Th cell differentiation has not been clarified. Here we demonstrate that the p38 signalling pathway is implicated in the generation of Th17 lymphocytes from human CD4(+)  CD27(+)  CD45RA(+) naive T cells, both in healthy donors and in patients affected by the relapsing-remitting form of MS. Our data also indicate that p38 activation is essential for interleukin-17 release from central memory lymphocytes and committed Th17 cell clones. Furthermore, CD4(+) T cells isolated from individuals with relapsing-remitting MS display an altered responsiveness of the p38 cascade, resulting in increased p38 phosphorylation upon stimulation. These findings suggest that the p38 signalling pathway, by modulating the Th17 differentiation and response, is involved in the pathogenesis of MS, and open new perspectives for the use of p38 inhibitors in the treatment of Th17-mediated autoimmune diseases.


Subject(s)
Cell Differentiation , Lymphocyte Activation , MAP Kinase Signaling System , Multiple Sclerosis, Relapsing-Remitting/enzymology , Th17 Cells/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphatases/metabolism , Adult , Case-Control Studies , Cation Transport Proteins/metabolism , Cells, Cultured , Copper-Transporting ATPases , Enzyme Activation , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Interleukin-17/metabolism , Interleukins/metabolism , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/immunology , Phenotype , Phosphorylation , Th17 Cells/immunology
6.
J Immunol ; 187(5): 2093-100, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21788446

ABSTRACT

Persistent viral infections and inflammatory syndromes induce the accumulation of T cells with characteristics of terminal differentiation or senescence. However, the mechanism that regulates the end-stage differentiation of these cells is unclear. Human CD4(+) effector memory (EM) T cells (CD27(-)CD45RA(-)) and also EM T cells that re-express CD45RA (CD27(-)CD45RA(+); EMRA) have many characteristics of end-stage differentiation. These include the expression of surface KLRG1 and CD57, reduced replicative capacity, decreased survival, and high expression of nuclear γH2AX after TCR activation. A paradoxical observation was that although CD4(+) EMRA T cells exhibit defective telomerase activity after activation, they have significantly longer telomeres than central memory (CM)-like (CD27(+)CD45RA(-)) and EM (CD27(-)CD45RA(-)) CD4(+) T cells. This suggested that telomerase activity was actively inhibited in this population. Because proinflammatory cytokines such as TNF-α inhibited telomerase activity in T cells via a p38 MAPK pathway, we investigated the involvement of p38 signaling in CD4(+) EMRA T cells. We found that the expression of both total and phosphorylated p38 was highest in the EM and EMRA compared with that of other CD4(+) T cell subsets. Furthermore, the inhibition of p38 signaling, especially in CD4(+) EMRA T cells, significantly enhanced their telomerase activity and survival after TCR activation. Thus, activation of the p38 MAPK pathway is directly involved in certain senescence characteristics of highly differentiated CD4(+) T cells. In particular, CD4(+) EMRA T cells have features of telomere-independent senescence that are regulated by active cell signaling pathways that are reversible.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Telomerase/immunology , Adult , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Humans , Immunologic Memory/immunology , In Situ Hybridization, Fluorescence , Leukocyte Common Antigens/biosynthesis , Leukocyte Common Antigens/immunology , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , Telomerase/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Exp Med ; 220(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36828392

ABSTRACT

In the prostate bone metastasis microenvironment, macrophages activate a cascade that involves Activin A, the extracellular matrix, and SRC kinase and drives resistance to anti-androgen therapy. These findings (Li et al., 2023. J. Exp. Med.https://doi.org/10.1084/jem.20221007) have broad implications, including metastasis diversity in different tissue milieus and the interplay between hormones and immunity.


Subject(s)
Bone Neoplasms , Macrophages , Male , Humans , Tumor Microenvironment
8.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35798535

ABSTRACT

Macrophages are main players of the innate immune system. They show great heterogeneity and play diverse functions that include support to development, sustenance of tissue homeostasis and defense against infections. Dysfunctional macrophages have been described in multiple pathologies including cancer. Indeed tumor-associated macrophages (TAMs) are abundant in most tumors and sustain cancer growth, promote invasion and mediate immune evasion. Importantly, lipid metabolism influences macrophage activation and lipid accumulation confers pathogenic features on macrophages. Notably, a subset of lipid-loaded macrophages has been recently identified in many tumor types. Lipid-loaded TAMs support tumor growth and progression and exert immune-suppressive activities. In this review, we describe the role of lipid metabolism in macrophage activation in physiology and pathology and we discuss the impact of lipid accumulation in macrophages in the context of cancer.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Lipids , Macrophages
9.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-34919143

ABSTRACT

Tumor-associated macrophages (TAMs) are correlated with the progression of prostatic adenocarcinoma (PCa). The mechanistic basis of this correlation and therapeutic strategies to target TAMs in PCa remain poorly defined. Here, single-cell RNA sequencing was used to profile the transcriptional landscape of TAMs in human PCa, leading to identification of a subset of macrophages characterized by dysregulation in transcriptional pathways associated with lipid metabolism. This subset of TAMs correlates positively with PCa progression and shorter disease-free survival and is characterized by an accumulation of lipids that is dependent on Marco. Mechanistically, cancer cell-derived IL-1ß enhances Marco expression on macrophages, and reciprocally, cancer cell migration is promoted by CCL6 released by lipid-loaded TAMs. Moreover, administration of a high-fat diet to tumor-bearing mice raises the abundance of lipid-loaded TAMs. Finally, targeting lipid accumulation by Marco blockade hinders tumor growth and invasiveness and improves the efficacy of chemotherapy in models of PCa, pointing to combinatorial strategies that may influence patient outcomes.


Subject(s)
Lipids , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Animals , Cell Plasticity/genetics , Cell Plasticity/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Knockdown Techniques , Heterografts , Humans , Lipid Metabolism , Lipids/chemistry , Male , Metabolic Networks and Pathways , Mice , Prostatic Neoplasms/pathology , Single-Cell Analysis
10.
Immunology ; 132(3): 326-39, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21214539

ABSTRACT

The relative roles that ageing and lifelong cytomegalovirus (CMV) infection have in shaping naive and memory CD4+ T-cell repertoires in healthy older people is unclear. Using multiple linear regression analysis we found that age itself is a stronger predictor than CMV seropositivity for the decrease in CD45RA+ CD27+ CD4+ T cells over time. In contrast, the increase in CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells is almost exclusively the result of CMV seropositivity, with age alone having no significant effect. Furthermore, the majority of the CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells in CMV-seropositive donors are specific for this virus. CD45RA+ CD27⁻ CD4+ T cells have significantly reduced CD28, interleukin-7 receptor α (IL-7Rα) and Bcl-2 expression, Akt (ser473) phosphorylation and reduced ability to survive after T-cell receptor activation compared with the other T-cell subsets in the same donors. Despite this, the CD45RA+ CD27⁻ subset is as multifunctional as the CD45RA⁻ D27+ and CD45RA⁻ CD27⁻ CD4+ T-cell subsets, indicating that they are not an exhausted population. In addition, CD45RA+ CD27⁻ CD4+ T cells have cytotoxic potential as they express high levels of granzyme B and perforin. CD4+ memory T cells re-expressing CD45RA can be generated from the CD45RA⁻ CD27+ population by the addition of IL-7 and during this process these cells down-regulated expression of IL-7R and Bcl-2 and so resemble their counterparts in vivo. Finally we showed that the proportion of CD45RA+ CD27⁻ CD4+ T cells of multiple specificities was significantly higher in the bone marrow than the blood of the same individuals, suggesting that this may be a site where these cells are generated.


Subject(s)
Aging/immunology , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Interleukin-7/physiology , T-Lymphocyte Subsets/immunology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Cell Separation , Cell Survival , Cytomegalovirus Infections/pathology , Flow Cytometry , Humans , Leukocyte Common Antigens/biosynthesis , Middle Aged , Signal Transduction/immunology , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Young Adult
11.
Blood ; 113(4): 827-36, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-18941119

ABSTRACT

The adoptive transfer of regulatory Foxp3(+) T (Treg) cells has been shown in various animal models to prevent inflammatory immune and autoimmune diseases. Translation into therapeutic applications, however, is hindered by the lack of suitable techniques and markers. CD25, commonly used to isolate Treg cells from mice, has only limited value in humans as it is also present on proinflammatory CD4(+) effector cells. Here we show that clean populations of human Foxp3(+) Treg cells can be obtained with antibodies directed against CD49d. The marker is present on proinflammatory peripheral blood mononuclear cells but is absent on immune-suppressive Treg cells. Depletion with alpha-CD49d removes contaminating interferon-gamma (IFN-gamma)- and interleukin-17 (IL-17)-secreting cells from Treg preparations of CD4(+)CD25(high) cells. More importantly, in combination with alpha-CD127 it allows the isolation of "untouched" Foxp3(+) Treg (ie, cells that have not been targeted by an antibody during purification). The removal of CD49d(+)/CD127(+) cells leaves a population of Foxp3(+) Treg virtually free of contaminating CD25(+) effector cells. The cells can be expanded in vitro and are effective suppressors both in vitro and in vivo. Thus, CD49d provides access to highly pure populations of untouched Foxp3(+) Treg cells conferring maximal safety for future clinical applications.


Subject(s)
Cell Separation/methods , Forkhead Transcription Factors/immunology , Integrin alpha1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Mice , T-Lymphocytes, Helper-Inducer/immunology , Time Factors
12.
Nat Rev Cancer ; 20(9): 485-503, 2020 09.
Article in English | MEDLINE | ID: mdl-32694624

ABSTRACT

Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Neutrophils/immunology , Animals , Humans , Immunotherapy , Neoplastic Processes , Tumor Microenvironment/immunology
13.
Cell Rep ; 28(8): 2156-2168.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433989

ABSTRACT

Tumor-associated macrophages (TAMs) represent a major component of the tumor microenvironment supporting tumorigenesis. TAMs re-education has been proposed as a strategy to promote tumor inhibition. However, whether this approach may work in prostate cancer is unknown. Here we find that Pten-null prostate tumors are strongly infiltrated by TAMs expressing C-X-C chemokine receptor type 2 (CXCR2), and activation of this receptor through CXCL2 polarizes macrophages toward an anti-inflammatory phenotype. Notably, pharmacological blockade of CXCR2 receptor by a selective antagonist promoted the re-education of TAMs toward a pro-inflammatory phenotype. Strikingly, CXCR2 knockout monocytes infused in Ptenpc-/-; Trp53pc-/- mice differentiated in tumor necrosis factor alpha (TNF-α)-releasing pro-inflammatory macrophages, leading to senescence and tumor inhibition. Mechanistically, PTEN-deficient tumor cells are vulnerable to TNF-α-induced senescence, because of an increase of TNFR1. Our results identify TAMs as targets in prostate cancer and describe a therapeutic strategy based on CXCR2 blockade to harness anti-tumorigenic potential of macrophages against this disease.


Subject(s)
Cellular Senescence , Macrophages/pathology , Prostatic Neoplasms/pathology , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Polarity , Chemokine CXCL2/administration & dosage , Chemokine CXCL2/pharmacology , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Neutralization Tests , PTEN Phosphohydrolase/metabolism , Receptors, Interleukin-8B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Transplantation ; 86(1): 171-5, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18622296

ABSTRACT

We have previously demonstrated that the compound 3-(2-ethylphenyl)-5-(3-methoxyphenyl)-1H-1,2,4-triazole exerts immunosuppressive effects in several experimental models of autoimmunity. These results were achieved by subcutaneously administering ST1959 after dissolution in an oily vehicle, because of its poor water solubility. To circumvent this problem, we sought to determine whether nanocochleate technology could be successfully exploited to deliver ST1959 and protect mice undergoing lethal acute graft-versus-host disease (GVHD). Orally-administered encochleated ST1959 significantly protected animals from lethality, resulting in survival rates of 57% and 100% at doses of 2 and 10 mg/kg, respectively, whereas oral administration of 2 mg/kg ST1959, mixed with empty nanocochleates, was completely inactive. Increased survival was associated with diminished serum chemokine levels and donor CD8+ T cells in the spleen of ST1959-treated mice. Moreover, ST1959 treatment significantly counteracted GVHD-induced normocitic anemia by increasing hemoglobin, hematocrit, platelet, and red and white blood cell counts. Overall, these data show that orally-administered encochleated ST1959 significantly protects mice from GVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Immunosuppressive Agents/pharmacology , Triazoles/pharmacology , Administration, Oral , Anemia/etiology , Anemia/prevention & control , Animals , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Chemokines/blood , Disease Models, Animal , Drug Compounding , Female , Graft vs Host Disease/blood , Graft vs Host Disease/complications , Graft vs Host Disease/immunology , Hematologic Tests , Immunosuppressive Agents/administration & dosage , Mice , Mice, Inbred C57BL , Nanocapsules , Time Factors , Triazoles/administration & dosage
17.
Nat Genet ; 50(2): 219-228, 2018 02.
Article in English | MEDLINE | ID: mdl-29335542

ABSTRACT

The mechanisms by which mitochondrial metabolism supports cancer anabolism remain unclear. Here, we found that genetic and pharmacological inactivation of pyruvate dehydrogenase A1 (PDHA1), a subunit of the pyruvate dehydrogenase complex (PDC), inhibits prostate cancer development in mouse and human xenograft tumor models by affecting lipid biosynthesis. Mechanistically, we show that in prostate cancer, PDC localizes in both the mitochondria and the nucleus. Whereas nuclear PDC controls the expression of sterol regulatory element-binding transcription factor (SREBF)-target genes by mediating histone acetylation, mitochondrial PDC provides cytosolic citrate for lipid synthesis in a coordinated manner, thereby sustaining anabolism. Additionally, we found that PDHA1 and the PDC activator pyruvate dehydrogenase phosphatase 1 (PDP1) are frequently amplified and overexpressed at both the gene and protein levels in prostate tumors. Together, these findings demonstrate that both mitochondrial and nuclear PDC sustain prostate tumorigenesis by controlling lipid biosynthesis, thus suggesting this complex as a potential target for cancer therapy.


Subject(s)
Cell Compartmentation/physiology , Lipogenesis , Prostatic Neoplasms/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex/physiology , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , Humans , Lipogenesis/genetics , Male , Mice , Mice, Knockout , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/metabolism
18.
Trends Cell Biol ; 26(3): 215-226, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26564316

ABSTRACT

Cellular senescence is a permanent growth arrest that is broadly recognized to act as a barrier against tumorigenesis. Senescence is predominant in premalignant tumors, and senescence escape is thought to be required for tumor progression. Importantly, evidences indicate that cell-autonomous mechanisms, such as genetic alterations or therapeutic interventions targeting specific genetic pathways, can affect the senescence response in cancer. Nevertheless, new findings have emerged in the last few years that indicate a fundamental role for the tumor microenvironment in the regulation of cellular senescence. Indeed, cytokines belonging to the senescent secretome, as well as tumor-infiltrating immune subsets, have been described to modulate the senescence response in tumors. Such evidence demonstrates that senescence initiation also relies on non-cell-autonomous mechanisms, which are discussed in the present review.


Subject(s)
Cellular Senescence , Neoplasms/pathology , Animals , Cell Transformation, Neoplastic/immunology , Cytokines/physiology , Humans , Inflammation/pathology , Neoplasms/genetics , Neoplasms/immunology , Oncogenes , Tumor Microenvironment/immunology
19.
Clin Cancer Res ; 21(14): 3108-12, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25967145

ABSTRACT

Tumor-infiltrating myeloid-derived suppressor cells (MDSC) are a heterogeneous and immunosuppressive cell subset that blocks the proliferation and the activity of both T and natural killer (NK) cells and promotes tumor vasculogenesis and progression. Recent evidences demonstrate that the recruitment of MDSCs in tumors also blocks senescence induced by chemotherapy promoting chemoresistance. Hence, the need of novel therapeutic approaches that can efficiently target MDSC recruitment and function in cancer. Among them, novel combinatorial treatments of chemotherapy and immunotherapy or treatments that induce depletion of MDSCs in peripheral sites should be taken in consideration.


Subject(s)
Immune Tolerance/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Humans
20.
Oncoimmunology ; 4(9): e988473, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26405613

ABSTRACT

The present study supports a model in which Pten loss-induced senescence is hindered in prostate tumor cells by non cell-autonomous mechanisms. Indeed, paracrine signaling by tumor-infiltrating CD11b+Gr-1+ myeloid cells triggers senescence evasion in prostate lesions of Pten-null mice, eventually promoting tumor progression.

SELECTION OF CITATIONS
SEARCH DETAIL