Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 120(43): e2219801120, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37862381

ABSTRACT

Senescent cells are beneficial for repairing acute tissue damage, but they are harmful when they accumulate in tissues, as occurs with advancing age. Senescence-associated extracellular vesicles (S-EVs) can mediate cell-to-cell communication and export intracellular content to the microenvironment of aging tissues. Here, we studied the uptake of EVs from senescent cells (S-EVs) and proliferating cells (P-EVs) and found that P-EVs were readily taken up by proliferating cells (fibroblasts and cervical cancer cells) while S-EVs were not. We thus investigated the surface proteome (surfaceome) of P-EVs relative to S-EVs derived from cells that had reached senescence via replicative exhaustion, exposure to ionizing radiation, or treatment with etoposide. We found that relative to P-EVs, S-EVs from all senescence models were enriched in proteins DPP4, ANXA1, ANXA6, S10AB, AT1A1, and EPHB2. Among them, DPP4 was found to selectively prevent uptake by proliferating cells, as ectopic overexpression of DPP4 in HeLa cells rendered DPP4-expressing EVs that were no longer taken up by other proliferating cells. We propose that DPP4 on the surface of S-EVs makes these EVs refractory to internalization by proliferating cells, advancing our knowledge of the impact of senescent cells in aging-associated processes.


Subject(s)
Cellular Senescence , Extracellular Vesicles , Humans , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , HeLa Cells , Extracellular Vesicles/metabolism , Aging
2.
Genes Dev ; 32(13-14): 909-914, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29967290

ABSTRACT

The senescence-associated secretory phenotype (SASP) is a major trait of senescent cells, but the molecular regulators of SASP factor secretion are poorly understood. Mass spectrometry analysis revealed that secretory carrier membrane protein 4 (SCAMP4) levels were strikingly elevated on the surface of senescent cells compared with proliferating cells. Interestingly, silencing SCAMP4 in senescent fibroblasts reduced the secretion of SASP factors, including interleukin 6 (IL6), IL8, growth differentiation factor 15 (GDF-15), C-X-C motif chemokine ligand 1 (CXCL1), and IL7, while, conversely, SCAMP4 overexpression in proliferating fibroblasts increased SASP factor secretion. Our results indicate that SCAMP4 accumulates on the surface of senescent cells, promotes SASP factor secretion, and critically enhances the SASP phenotype.


Subject(s)
Carrier Proteins/metabolism , Cellular Senescence/genetics , Fibroblasts/metabolism , Membrane Proteins/metabolism , Carrier Proteins/genetics , Cell Line , Cell Proliferation/physiology , Fibroblasts/cytology , Gene Silencing , Humans , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
3.
Genes Dev ; 31(15): 1529-1534, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28877934

ABSTRACT

Senescent cell accumulation in aging tissues is linked to age-associated diseases and declining function, prompting efforts to eliminate them. Mass spectrometry analysis revealed that DPP4 (dipeptidyl peptidase 4) was selectively expressed on the surface of senescent, but not proliferating, human diploid fibroblasts. Importantly, the differential presence of DPP4 allowed flow cytometry-mediated isolation of senescent cells using anti-DPP4 antibodies. Moreover, antibody-dependent cell-mediated cytotoxicity (ADCC) assays revealed that the cell surface DPP4 preferentially sensitized senescent, but not dividing, fibroblasts to cytotoxicity by natural killer cells. In sum, the selective expression of DPP4 on the surface of senescent cells enables their preferential elimination.


Subject(s)
Cellular Senescence/physiology , Dipeptidyl Peptidase 4/metabolism , Membrane Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antibody-Dependent Cell Cytotoxicity , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Diploidy , Fibroblasts/metabolism , Flow Cytometry , Humans , Killer Cells, Natural/metabolism , Lymphocyte Subsets/enzymology , Mass Spectrometry , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism
4.
Immun Ageing ; 18(1): 2, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33397404

ABSTRACT

Extrinsic factors, such as lifestyle and diet, are shown to be essential in the control of human healthy aging, and thus, longevity. They do so by targeting at least in part the gut microbiome, a collection of commensal microorganisms (microbiota), which colonize the intestinal tract starting after birth, and is established by the age of three. The composition and abundance of individual microbiota appears to continue to change until adulthood, presumably reflecting lifestyle and geographic, racial, and individual differences. Although most of these changes appear to be harmless, a major shift in their composition in the gut (dysbiosis) can trigger harmful local and systemic inflammation. Recent reports indicate that dysbiosis is increased in aging and that the gut microbiota of elderly people is enriched in pro-inflammatory commensals at the expense of beneficial microbes. The clinical consequence of this change remains confusing due to contradictory reports and a high degree of variability of human microbiota and methodologies used. Here, we present the authors' thoughts that underscore dysbiosis as a primary cause of aging-associated morbidities, and thus, premature death of elderly people. We provide evidence that the dysbiosis triggers a chain of pathological and inflammatory events. Examples include alteration of levels of microbiota-affected metabolites, impaired function and integrity of the gastrointestinal tract, and increased gut leakiness. All of these enhance systemic inflammation, which when associated with aging is termed inflammaging, and result in consequent aging-associated pathologies.

5.
Brain Behav Immun ; 75: 163-180, 2019 01.
Article in English | MEDLINE | ID: mdl-30389461

ABSTRACT

Down Syndrome (DS), the most common cause of genetic intellectual disability, is characterized by over-expression of the APP and DYRK1A genes, located on the triplicated chromosome 21. This chromosomal abnormality leads to a cognitive decline mediated by Amyloid-ß (Aß) overproduction and tau hyper-phosphorylation as early as the age of 40. In this study, we used the Ts65Dn mouse model of DS to evaluate the beneficial effect of a DNA vaccination against the Aß1-11 fragment, in ameliorating Aß-related neuropathology and rescue of cognitive and behavioral abilities. Anti-Aß1-11 vaccination induced antibody production and facilitated clearance of soluble oligomers and small extracellular inclusions of Aß from the hippocampus and cortex of Ts65Dn mice. This was correlated with reduced neurodegeneration and restoration of the homeostatic phenotype of microglial and astroglial cells. Vaccinated Ts65Dn mice performed better in spatial-learning tasks, exhibited reduced motor hyperactivity typical for this strain, and restored short-term memory abilities. Our findings support the hypothesis that DS individuals may benefit from active immunotherapy against Aß from a young age by slowing the progression of dementia.


Subject(s)
Amyloid beta-Peptides/immunology , Down Syndrome/immunology , Down Syndrome/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Brain/metabolism , DNA/immunology , Disease Models, Animal , Hippocampus/metabolism , Immunization/methods , Male , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , tau Proteins , Dyrk Kinases
6.
Lancet Oncol ; 19(6): e295-e304, 2018 06.
Article in English | MEDLINE | ID: mdl-29893261

ABSTRACT

Cancer incidence substantially increases with ageing in both men and women, although the reason for this increase is unknown. In this Series paper, we propose that age-associated changes in gut commensal microbes, otherwise known as the microbiota, facilitate cancer development and growth by compromising immune fitness. Ageing is associated with a reduction in the beneficial commensal microbes, which control the expansion of pathogenic commensals and maintain the integrity of the intestinal barrier through the production of mucus and lipid metabolites, such as short-chain fatty acids. Expansion of gut dysbiosis and leakage of microbial products contributes to the chronic proinflammatory state (inflammaging), which negatively affects the immune system and impairs the removal of mutant and senescent cells, thereby enabling tumour outgrowth. Studies in animal models and the importance of commensals in cancer immunotherapy suggest that this status can be reversible. Thus, interventions that alter the composition of the gut microbiota might reduce inflammaging and rejuvenate immune functions to provide anticancer benefits in frail elderly people.


Subject(s)
Aging , Gastrointestinal Microbiome , Inflammation/microbiology , Intestines/microbiology , Neoplasms/microbiology , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Animals , Dysbiosis , Female , Frail Elderly , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions , Humans , Immunocompromised Host , Incidence , Inflammation/epidemiology , Inflammation/immunology , Inflammation Mediators/immunology , Intestines/immunology , Male , Neoplasms/epidemiology , Neoplasms/immunology , Prognosis , Risk Assessment , Risk Factors
7.
J Immunol ; 196(8): 3385-97, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26983789

ABSTRACT

B cell dysregulation in aging is thought to mostly occur in conventional B2 cells without affecting innate B1 cells. Elderly humans and mice also accumulate 4-1BBL(+)MHC class-I(Hi)CD86(Hi)B cells of unknown origin. In this article, we report that these cells, termed 4BL cells, are activated murine and possibly human B1a cells. The activation is mediated by aging human monocytes and murine peritoneal macrophages. They induce expression and activation of 4-1BBL and IFN-γR1 on B1a cells to subsequently upregulate membrane TNF-α and CD86. As a result, activated B1a/4BL cells induce expression of granzyme B in CD8(+)T cells by targeting TNFR2 via membrane TNF-α and providing costimulation with CD86. Thus, for the first time, to our knowledge, these results indicate that aging affects the function of B1a cells. Upon aging, these cells lose their tumor-supporting activity and become inducers of potentially antitumor and autoimmune CD8(+)T cells.


Subject(s)
Aging/immunology , B-Lymphocyte Subsets/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , 4-1BB Ligand/metabolism , Adult , Aged , Animals , Autoimmunity/immunology , B-Lymphocyte Subsets/immunology , B7-2 Antigen/biosynthesis , B7-2 Antigen/metabolism , Cells, Cultured , Cellular Senescence , Enzyme Activation/immunology , Female , Granzymes/biosynthesis , Histocompatibility Antigens Class I/metabolism , Humans , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Neoplasms/immunology , Neoplasms/pathology , Receptors, Interferon/biosynthesis , Receptors, Interferon/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Interferon gamma Receptor
8.
Blood ; 124(9): 1450-9, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25037628

ABSTRACT

Although the accumulation of highly-differentiated and granzyme B (GrB)-expressing CD8(+)CD28(-) T cells has been associated with aging, the mechanism for their enrichment and contribution to immune function remains poorly understood. Here we report a novel B-cell subset expressing 4-1BBL, which increases with age in humans, rhesus macaques, and mice, and with immune reconstitution after chemotherapy and autologous progenitor cell transplantation. These cells (termed 4BL cells) induce GrB(+)CD8(+) T cells by presenting endogenous antigens and using the 4-1BBL/4-1BB axis. We found that the 4BL cells increase antitumor responses in old mice, which may explain in part the paradox of retarded tumor growth in the elderly. 4BL cell accumulation and its capacity to evoke the generation of GrB(+)CD8(+) T cells can be eliminated by inducing reconstitution of B cells in old mice, suggesting that the age-associated skewed cellular immune responses are reversible. We propose that 4BL cells and the 4-1BBL signaling pathway are useful targets for improved effectiveness of natural antitumor defenses and therapeutic immune manipulations in the elderly.


Subject(s)
4-1BB Ligand/metabolism , Aging/immunology , B-Lymphocyte Subsets/immunology , CD8-Positive T-Lymphocytes/immunology , Granzymes/metabolism , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/enzymology , Female , Humans , Immunity, Cellular , Immunity, Innate , Macaca mulatta , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
9.
FASEB J ; 29(2): 589-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25342129

ABSTRACT

Insulin resistance causes diminished glucose uptake in similar regions of the brain in Alzheimer's disease (AD) and type 2 diabetes mellitus (DM2). Brain tissue studies suggested that insulin resistance is caused by low insulin receptor signaling attributable to its abnormal association with more phospho (P)-serine-type 1 insulin receptor substrate (IRS-1) and less P-tyrosine-IRS-1. Plasma exosomes enriched for neural sources by immunoabsorption were obtained once from 26 patients with AD, 20 patients with DM2, 16 patients with frontotemporal dementia (FTD), and matched case control subjects. At 2 time points, they were obtained from 22 others when cognitively normal and 1 to 10 yr later when diagnosed with AD. Mean exosomal levels of extracted P-serine 312-IRS-1 and P-pan-tyrosine-IRS-1 by ELISA and the ratio of P-serine 312-IRS-1 to P-pan-tyrosine-IRS-1 (insulin resistance factor, R) for AD and DM2 and P-serine 312-IRS-1 and R for FTD were significantly different from those for case control subjects. The levels of R for AD were significantly higher than those for DM2 or FTD. Stepwise discriminant modeling showed correct classification of 100% of patients with AD, 97.5% of patients with DM2, and 84% of patients with FTD. In longitudinal studies of 22 patients with AD, exosomal levels of P-serine 312-IRS-1, P-pan-tyrosine-IRS-1, and R were significantly different 1 to 10 yr before and at the time of diagnosis compared with control subjects. Insulin resistance reflected in R values from this blood test is higher for patients with AD, DM2, and FTD than case control subjects; higher for patients with AD than patients with DM2 or FTD; and accurately predicts development of AD up to 10 yr prior to clinical onset.


Subject(s)
Alzheimer Disease/metabolism , Exosomes/metabolism , Gene Expression Regulation , Insulin Receptor Substrate Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Case-Control Studies , Cognition Disorders/blood , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Frontotemporal Dementia/blood , Humans , Insulin Resistance , Male , Middle Aged , Phosphorylation , Retrospective Studies , Time Factors , Treatment Outcome
10.
Retrovirology ; 12: 73, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289727

ABSTRACT

BACKGROUND: Adult T-cell leukemia (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1) infection. However, there are no therapies to prevent ATL development in high-risk asymptomatic carriers. To develop a therapy targeting HTLV-1-infected cells that are known to express CCR4 frequently, we tested whether truncated Pseudomonas exotoxin (PE38) fused to a CCR4 ligand, CCL17/thymus and activation-regulated chemokine (TARC), selectively eliminates such cells. RESULTS: Our data show that TARC-PE38 efficiently killed HTLV-1-infected cell lines. It also shrank HTLV-1-associated solid tumors in an infected-cell-engrafted mouse model. In HTLV-1-positive humanized mice, TARC-PE38 markedly inhibited the proliferation of HTLV-1-infected human CD4(+)CD25(+) or CD4(+)CD25(+)CCR4(+) cells and reduced the proviral loads (PVLs) in peripheral blood mononuclear cells (PBMCs). Importantly, TARC-PE38 significantly reduced the PVLs in PBMCs obtained from asymptomatic carriers. We show that the cytotoxicity of TARC-PE38 is mediated by the expression of the proprotein convertase, furin. The expression of furin was enhanced in HTLV-1-infected cells and correlated positively with PVLs in HTLV-1-infected individuals, suggesting that infected cells are more susceptible to TARC-PE38 than normal cells. CONCLUSIONS: TARC-PE38 robustly controls HTLV-1 infection by eliminating infected cells in both a CCR4- and furin-dependent manner, indicating the excellent therapeutic potential of TARC-PE38.


Subject(s)
Chemokine CCL17/pharmacology , Exotoxins/pharmacology , Furin/genetics , Furin/pharmacology , Human T-lymphotropic virus 1/drug effects , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/virology , Receptors, CCR4/metabolism , Recombinant Fusion Proteins/pharmacology , Adult , Animals , Asymptomatic Infections/therapy , Cell Line, Tumor , Chemokines/genetics , Disease Models, Animal , Human T-lymphotropic virus 1/growth & development , Humans , Jurkat Cells , Leukocytes, Mononuclear/virology , Mice , Proviruses/drug effects , Proviruses/physiology , Receptors, CCR4/genetics , U937 Cells
11.
Antimicrob Agents Chemother ; 59(1): 527-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385102

ABSTRACT

Despite years of research dedicated to preventing the sexual transmission of herpes simplex virus 2 (HSV-2), there is still no protective vaccine or microbicide against one of the most common sexually transmitted infections in the world. Using a phage display library constructed from a llama immunized with recombinant HSV-2 glycoprotein D, we identified a single-domain antibody VHH, R33, which binds to the viral surface glycoprotein D. Although R33 does not demonstrate any HSV-2 neutralization activity in vitro, when expressed with the cytotoxic domain of exotoxin A, the resulting immunotoxin (R33ExoA) specifically and potently kills HSV-2-infected cells, with a 50% neutralizing dilution (IC50) of 6.7 nM. We propose that R33ExoA could be used clinically to prevent transmission of HSV-2 through killing of virus-producing epithelial cells during virus reactivation. R33 could also potentially be used to deliver other cytotoxic effectors to HSV-2-infected cells.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 2, Human/drug effects , Single-Domain Antibodies/pharmacology , Viral Envelope Proteins/metabolism , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Camelids, New World , Chlorocebus aethiops , Exotoxins/genetics , Exotoxins/immunology , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/pharmacology , Neutralization Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Toxicity Tests/methods , Vero Cells/drug effects , Vero Cells/virology , Virulence Factors/genetics , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
12.
J Immunol ; 190(6): 2575-84, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23408836

ABSTRACT

Breast cancer cells facilitate distant metastasis through the induction of immunosuppressive regulatory B cells, designated tBregs. We report in this study that, to do this, breast cancer cells produce metabolites of the 5-lipoxygenase pathway such as leukotriene B4 to activate the peroxisome proliferator-activated receptor α (PPARα) in B cells. Inactivation of leukotriene B4 signaling or genetic deficiency of PPARα in B cells blocks the generation of tBregs and thereby abrogates lung metastasis in mice with established breast cancer. Thus, in addition to eliciting fatty acid oxidation and metabolic signals, PPARα initiates programs required for differentiation of tBregs. We propose that PPARα in B cells and/or tumor 5-lipoxygenase pathways represents new targets for pharmacological control of tBreg-mediated cancer escape.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Lipoxygenase/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , PPAR alpha/physiology , Animals , B-Lymphocyte Subsets/enzymology , Cell Line, Tumor , Cells, Cultured , Female , Lipoxygenase/genetics , Lipoxygenase/metabolism , Melanoma, Experimental/genetics , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , PPAR alpha/deficiency , PPAR alpha/genetics , Tumor Escape/genetics , Tumor Escape/immunology
13.
J Immunol ; 191(8): 4141-51, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043896

ABSTRACT

We reported previously that tumor-evoked regulatory B cells (tBregs) play an essential role in breast cancer lung metastasis by inducing TGF-ß-dependent conversion of metastasis-promoting Foxp3(+) regulatory T cells (Tregs). In this article, we show that resveratrol (RSV), a plant-derived polyphenol, at low and noncytotoxic doses for immune cells, can efficiently inhibit lung metastasis in mice. The mechanism of this process is that RSV inactivates Stat3, preventing the generation and function of tBregs, including expression of TGF-ß. As a result, it frees antitumor effector immune responses by disabling tBreg-induced conversion of Foxp3(+) Tregs. We propose that low doses of RSV may also benefit humans by controlling cancer escape-promoting tBregs/Tregs without nonspecific inactivation of effector immune cells.


Subject(s)
B-Lymphocytes, Regulatory/drug effects , Lung Neoplasms/prevention & control , Mammary Neoplasms, Animal/drug therapy , Stilbenes/therapeutic use , T-Lymphocytes, Regulatory/immunology , Animals , Female , Forkhead Transcription Factors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Resveratrol , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/biosynthesis
14.
Semin Cancer Biol ; 22(1): 50-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22257681

ABSTRACT

Metastatic cancer is usually an incurable disease. Cancers have a broad repertoire of subversive tactics to defeat the immune system. They mimic self, they down-regulate MHC molecules so that T cells are blind to their presence, they interfere with antigen presentation, and they produce factors that can kill T cells or paralyze their response to antigens. Furthermore, the same powerful machinery designed to prevent harmful autoimmune responses is also acting to protect cancers. In particular, cancer is protected with the help of so-called regulatory immune cells. These unique subsets of cells, represented by almost every immune cell type, function to control responses of effector immune cells. In this review, we will discuss the evidence that cancer actively promotes cross-talk of regulatory immune cells to evade immunosurveillance. We will also discuss the role of a newly described cell type, regulatory B cells, by emphasizing their importance in suppression of antitumor immune responses. Thus, cancer not only directly suppresses immune function, but also recruits components of the immune system to become traitors and protect the tumor from immune attack.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Chemokines/physiology , Immunologic Surveillance/physiology , Neoplasm Metastasis/physiopathology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape/physiology , Disease Progression , Humans , Immunotherapy , Neoplasms/therapy
15.
FASEB J ; 26(3): 1301-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22121051

ABSTRACT

A role for adenosine in immunosenescence was investigated in T cells from older (≥65 yr) and younger (24-45 yr) healthy humans. Adenosine concentrations in cultures of activated T cells were significantly higher (P<0.0001) for older (145±47 nM, mean±sd) than younger (58±5.5 nM) subjects. Expression of the activation coreceptor CD28 was suppressed significantly by 0.1 to 1 µM exogenous adenosine, with greater effects of 1 µM (P<0.01) on T cells of younger (mean suppression of 67 and 65% for CD4 and CD8 T cells, respectively) than older (means of 42 and 46%) subjects. T-cell chemotaxis to CCL21 was suppressed significantly by 0.3 and 1 µM exogenous adenosine, with mean maximum decreases of 39 and 49%, respectively, for younger subjects and 28 and 31% for older subjects. Generation of IL-2 and IFN-γ by T cells of younger and older subjects was suppressed substantially only at adenosine levels of 3 µM or higher. Lower baseline expression of CD28 and chemotaxis to CCL21 and S1P for T cells from older subjects attributable to endogenous adenosine were reversed completely by two different A(2A) adenosine receptor antagonists without affecting T cells of younger subjects. Adenosine is an endogenous T-cell immunosuppressor in older humans, and A(2A) antagonists reverse adenosine-induced T-cell deficiencies of aging.


Subject(s)
Adenosine/immunology , Adenosine/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adult , Age Factors , Aged , Aged, 80 and over , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cells, Cultured , Chemotaxis/drug effects , Cytokines/immunology , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Male , Middle Aged , Phenethylamines/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes/metabolism , Triazoles/pharmacology , Young Adult
16.
J Immunol ; 186(10): 5656-62, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21490155

ABSTRACT

Inflammation is a double-edged sword that can promote or suppress cancer progression. In this study, we report that thymic stromal lymphopoietin (TSLP), an IL-7-like type 1 inflammatory cytokine that is often associated with the induction of Th2-type allergic responses in the lungs, is also expressed in human and murine cancers. Our studies with murine cancer cells indicate that TSLP plays an essential role in cancer escape, as its inactivation in cancer cells alone was sufficient to almost completely abrogate cancer progression and lung metastasis. The cancer-promoting activity of TSLP primarily required signaling through the TSLP receptor on CD4(+) T cells, promoting Th2-skewed immune responses and production of immunosuppressive factors such as IL-10 and IL-13. Expression of TSLP therefore may be a useful prognostic marker, and its targeting could have therapeutic potential.


Subject(s)
Breast Neoplasms/immunology , Cytokines/metabolism , Mammary Neoplasms, Animal/immunology , Animals , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Disease Progression , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , RNA Interference , RNA, Small Interfering , Th2 Cells/immunology , Tumor Escape , Thymic Stromal Lymphopoietin
17.
Nat Commun ; 13(1): 5376, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36104343

ABSTRACT

We have recently reported that some cancers induce accumulation of bone marrow (BM) B-cell precursors in the spleen to convert them into metastasis-promoting, immunosuppressive B cells. Here, using various murine tumor models and samples from humans with breast and ovarian cancers, we provide evidence that cancers also co-opt differentiation of these B-cell precursors to generate macrophage-like cells (termed B-MF). We link the transdifferentiation to a small subset of CSF1R+ Pax5Low cells within BM pre-B and immature B cells responding to cancer-secreted M-CSF with downregulation of the transcription factor Pax5 via CSF1R signaling. Although the primary source of tumor-associated macrophages is monocytes, B-MFs are phenotypically and functionally distinguishable. Compared to monocyte-derived macrophages, B-MFs more efficiently phagocytize apoptotic cells, suppress proliferation of T cells and induce FoxP3+ regulatory T cells. In mouse tumor models, B-MFs promote shrinkage of the tumor-infiltrating IFNγ+ CD4 T cell pool and increase cancer progression and metastasis, suggesting that this cancer-induced transdifferentiation pathway is functionally relevant and hence could serve as an immunotherapeutic target.


Subject(s)
Macrophages , Neoplasms , Animals , B-Lymphocytes , Cell Differentiation , Humans , Mice , Monocytes
18.
Geroscience ; 44(5): 2471-2490, 2022 10.
Article in English | MEDLINE | ID: mdl-35996062

ABSTRACT

Caloric restriction (CR) is the leading non-pharmacological intervention to delay induced and spontaneous tumors in pre-clinical models. These effects of CR are largely attributed to canonical inhibition of pro-growth pathways. However, our recent data suggest that CR impairs primary tumor growth and cancer progression in the murine 4T1 model of triple negative breast cancer (TNBC), at least in part, through reduced frequency of the myeloid-derived suppressor cells (MDSC). In the present study, we sought to determine whether injection of excess MDSCs could block regression in 4T1 tumor growth and metastatic spread in BALB/cJ female mice undergoing daily CR. Our findings show that MDSC injection impeded CR-mediated protection against tumor growth without increasing lung metastatic burden. Overall, these results reveal that CR can slow cancer progression by affecting immune suppressive cells.Impact statement: Inoculation of MDSCs from donor mice effectively impedes the ability of calorie restriction to protect against primary tumor growth without impacting lung metastatic burden in recipient animals.


Subject(s)
Myeloid-Derived Suppressor Cells , Triple Negative Breast Neoplasms , Humans , Female , Mice , Animals , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Caloric Restriction , Disease Models, Animal , Cell Line, Tumor
19.
Aging Cancer ; 3(2): 116-129, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36188490

ABSTRACT

Background: Age is the most significant risk factor for ovarian cancer (OvCa), the deadliest gynecologic malignancy. Metastasizing OvCa cells adhere to the omentum, a peritoneal structure rich in collagen, adipocytes, and immune cells. Ultrastructural changes in the omentum and the omental collagen matrix with aging have not been evaluated. Aim: The aim of this study was to test the hypothesis that age-related changes in collagen in the ovarian tumor microenvironment promote OvCa metastatic success in the aged host. Methods/Results: Young (3-6 months) and aged mice (20-23 months) were used to study the role of aging in metastatic success. Intra-peritoneal (IP) injection of ID8Trp53 -/- ovarian cancer cells showed enhanced IP dissemination in aged vs young mice. In vitro assays using purified collagen demonstrated reduced collagenolysis of aged fibers, as visualized using scanning electron microscopy (SEM) and quantified with a hydroxyproline release assay. Omental tumors in young and aged mice showed similar collagen deposition; however enhanced intra-tumoral collagen remodeling was seen in aged mice probed with a biotinylated collagen hybridizing peptide (CHP). In contrast, second harmonic generation (SHG) microscopy showed significant differences in collagen fiber structure and organization in omental tissue and SEM demonstrated enhanced omental fenestration in aged omenta. Combined SHG and Alexa Fluor-CHP microscopy in vivo demonstrated that peri-tumoral collagen was remodeled more extensively in young mice. This collagen population represents truly aged host collagen, in contrast to intra-tumoral collagen that is newly synthesized, likely by cancer associated fibroblasts (CAFs). Conclusions: Our results demonstrate that tumors in an aged host can grow with minimal collagen remodeling, while tumors in the young host must remodel peri-tumoral collagen to enable effective proliferation, providing a mechanism whereby age-induced ultrastructural changes in collagen and collagen-rich omenta establish a permissive pre-metastatic niche contributing to enhanced OvCa metastatic success in the aged host.

20.
Blood ; 113(3): 575-84, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18840710

ABSTRACT

Dexamethasone (DM) is a synthetic member of the glucocorticoid (GC) class of hormones that possesses anti-inflammatory and immunosuppressant activity and is commonly used to treat chronic inflammatory disorders, severe allergies, and other disease states. Although GCs are known to mediate well-defined transcriptional effects via GC receptors (GCR), there is increasing evidence that GCs also initiate rapid nongenomic signaling events in a variety of cell types. Here, we report that DM induces the phosphorylation of Lck and the activation of other downstream mediators, including p59Fyn, Zap70, Rac1, and Vav in resting but not activated human T cells. DM treatment also augments CXCL12-mediated signaling in resting T cells through its cell surface receptor, CXCR4 resulting in the enhanced actin polymerization, Rac activation, and cell migration on ligand exposure. Lck was found to be a critical intermediate in these DM-induced signaling activities. Moreover, DM-mediated Lck phosphorylation in T cells was dependent on the presence of both the GCR and the CD45 molecule. Overall, these results elucidate additional nongenomic effects of DM and the GCR on resting human T cells, inducing Lck and downstream kinase activation and augmenting chemokine signaling and function.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/drug effects , Receptors, CXCR4/drug effects , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Cell Line , Cell Movement/drug effects , Enzyme Activation/drug effects , Enzyme Activation/immunology , Flow Cytometry , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphorylation/drug effects , Receptors, CXCR4/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL