Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 41(3): 450-465, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36917950

ABSTRACT

Regulatory T cells (Tregs) are abundant in tumor tissues, raising a question of whether immunosuppressive tumor-infiltrating Tregs (TI-Tregs) can be selectively depleted or functionally attenuated to evoke effective anti-tumor immune responses by conventional T cells (Tconvs), without perturbing Treg-dependent immune homeostasis in healthy organs and causing autoimmunity. Here, we review current cancer immunotherapy strategies, including immune checkpoint blockade (ICB) antibodies against CTLA-4 and PD-1 and discuss their effects on TI-Tregs. We also discuss approaches that exploit differentially regulated molecules on the cell surface (e.g., CTLA-4) and intracellularly (e.g., T cell receptor signaling molecules) between TI-Tregs and Tconvs as well as their dependence on cytokines (e.g., IL-2) and metabolites (e.g., lactate). We envisage that targeting TI-Tregs could be effective as a monotherapy and/or when combined with ICB antibodies.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , CTLA-4 Antigen , Immunity , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment
2.
Cancers (Basel) ; 13(1)2020 Dec 26.
Article in English | MEDLINE | ID: mdl-33375291

ABSTRACT

Antibody-mediated disruption of the programmed cell death protein 1 (PD-1) pathway has brought much success to the fight against cancer. Nevertheless, a significant proportion of patients respond poorly to anti-PD-1 treatment. Cases of accelerated and more aggressive forms of cancer following therapy have also been reported. Termed hyper-progressive disease (HPD), this phenomenon often results in fatality, thus requires urgent attention. Among possible causes of HPD, regulatory T-cells (Tregs) are of suspect due to their high expression of PD-1, which modulates Treg activity. Tregs are a subset of CD4+ T-cells that play a non-redundant role in the prevention of autoimmunity and is functionally dependent on the X chromosome-linked transcription factor FoxP3. In cancer, CD4+FoxP3+ Tregs migrate to tumors to suppress anti-tumor immune responses, allowing cancer cells to persist. Hence, Treg accumulation in tumors is associated with poor prognosis. In mice, the anti-tumor efficacy of anti-PD-1 can be enhanced by depleting Tregs. This suggests Tregs pose resistance to anti-PD-1 therapy. In this article, we review the relevant Treg functions that suppress tumor immunity and the potential effects anti-PD-1 could have on Tregs which are counter-productive to the treatment of cancer, occasionally causing HPD.

3.
Adv Exp Med Biol ; 1189: 179-210, 2019.
Article in English | MEDLINE | ID: mdl-31758535

ABSTRACT

Foxp3-expressing regulatory T cells (Tregs) perform a vital function in the maintenance of immune homeostasis. A large part of Treg suppressive function is derived from their ability to control and restrict the availability of co-signal molecules to other T cells. However, Tregs themselves also depend on many of the same co-signals for their own homeostasis, making this a complex system of feedback. In this chapter, we discuss the critical role of co-signaling in Treg cell biology.


Subject(s)
Signal Transduction , T-Lymphocytes, Regulatory/cytology , Forkhead Transcription Factors/metabolism , Homeostasis , Humans
4.
Proc Natl Acad Sci U S A ; 116(20): 9999-10008, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028147

ABSTRACT

PD-1 blockade is a cancer immunotherapy effective in various types of cancer. In a fraction of treated patients, however, it causes rapid cancer progression called hyperprogressive disease (HPD). With our observation of HPD in ∼10% of anti-PD-1 monoclonal antibody (mAb)-treated advanced gastric cancer (GC) patients, we explored how anti-PD-1 mAb caused HPD in these patients and how HPD could be treated and prevented. In the majority of GC patients, tumor-infiltrating FoxP3highCD45RA-CD4+ T cells [effector Treg (eTreg) cells], which were abundant and highly suppressive in tumors, expressed PD-1 at equivalent levels as tumor-infiltrating CD4+ or CD8+ effector/memory T cells and at much higher levels than circulating eTreg cells. Comparison of GC tissue samples before and after anti-PD-1 mAb therapy revealed that the treatment markedly increased tumor-infiltrating proliferative (Ki67+) eTreg cells in HPD patients, contrasting with their reduction in non-HPD patients. Functionally, circulating and tumor-infiltrating PD-1+ eTreg cells were highly activated, showing higher expression of CTLA-4 than PD-1- eTreg cells. PD-1 blockade significantly enhanced in vitro Treg cell suppressive activity. Similarly, in mice, genetic ablation or antibody-mediated blockade of PD-1 in Treg cells increased their proliferation and suppression of antitumor immune responses. Taken together, PD-1 blockade may facilitate the proliferation of highly suppressive PD-1+ eTreg cells in HPDs, resulting in inhibition of antitumor immunity. The presence of actively proliferating PD-1+ eTreg cells in tumors is therefore a reliable marker for HPD. Depletion of eTreg cells in tumor tissues would be effective in treating and preventing HPD in PD-1 blockade cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Nivolumab/adverse effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Stomach Neoplasms/immunology , T-Lymphocytes, Regulatory/drug effects , Aged , Animals , CTLA-4 Antigen/metabolism , Disease Progression , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Male , Mice , Stomach Neoplasms/drug therapy , T-Lymphocytes, Regulatory/metabolism
5.
Front Immunol ; 10: 3046, 2019.
Article in English | MEDLINE | ID: mdl-31998318

ABSTRACT

Interaction between B and CD4 T cells is crucial for their optimal responses in adaptive immunity. Immune responses augmented by their partnership promote chronic inflammation. Here we report that interaction between B and CD4 T cells augments their atherogenicity to promote lipid-induced atherosclerosis. Genetic deletion of the gene encoding immunoglobulin mu (µ) heavy chain (µMT) in ApoE-/- mice resulted in global loss of B cells including those in atherosclerotic plaques, undetectable immunoglobulins and impaired germinal center formation. Despite unaffected numbers in the circulation and peripheral lymph nodes, CD4 T cells were also reduced in spleens as were activated and memory CD4 T cells. In hyperlipidemic µMT-/- ApoE-/- mice, B cell deficiency decreased atherosclerotic lesions, accompanied by absence of immunoglobulins and reduced CD4 T cell accumulation in lesions. Adoptive transfer of B cells deficient in either MHCII or co-stimulatory molecule CD40, molecules required for B and CD4 T cell interaction, into B cell-deficient µMT-/- ApoE-/- mice failed to increase atherosclerosis. In contrast, wildtype B cells transferred into µMT-/- ApoE-/- mice increased atherosclerosis and increased CD4 T cells in lesions including activated and memory CD4 T cells. Transferred B cells also increased their expression of atherogenic cytokines IL-1ß, TGF-ß, MCP-1, M-CSF, and MIF, with partial restoration of germinal centers and plasma immunoglobulins. Our study demonstrates that interaction between B and CD4 T cells utilizing MHCII and CD40 is essential to augment their function to increase atherosclerosis in hyperlipidemic mice. These findings suggest that targeting B cell and CD4 T cell interaction may be a therapeutic strategy to limit atherosclerosis progression.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Animals , Apolipoproteins E/immunology , Cytokines/immunology , Immunoglobulins/immunology , Male , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/immunology
6.
J Am Heart Assoc ; 7(13)2018 06 23.
Article in English | MEDLINE | ID: mdl-29936416

ABSTRACT

BACKGROUND: Peritoneal B1a cells attenuate atherosclerosis by secreting natural polyclonal immunoglobulin M (IgM). Regulatory B cells expressing T-cell immunoglobulin mucin domain-1 (TIM-1) expanded through TIM-1 ligation by anti-TIM-1 monoclonal antibody (RMT1-10) induces immune tolerance. METHODS AND RESULTS: We examined the capacity of RMT1-10 to expand peritoneal B1a cells to prevent atherosclerosis development and retard progression of established atherosclerosis. RMT1-10 treatment selectively doubled peritoneal B1a cells, tripled TIM-1+ B1a cells and increased TIM-1+IgM+interleukin (IL)-10+ by 3-fold and TIM-1+IgM+IL-10- B1a cells by 2.5-fold. Similar expansion of B1a B cells was observed in spleens. These effects reduced atherosclerotic lesion size, increased plasma IgM and lesion IgM deposits, and decreased oxidatively modified low-density lipoproteins in lesions. Lesion CD4+ and CD8+ T cells, macrophages and monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, expression of proinflammatory cytokines monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, IL1ß, apoptotic cell numbers and necrotic cores were also reduced. RMT1-10 treatment failed to expand peritoneal B1a cells and reduce atherosclerosis after splenectomy that reduces B1a cells, indicating that these effects are B1a cell-dependent. Apolipoprotein E-KO mice fed a high-fat diet for 6 weeks before treatment with RMT1-10 also increased TIM-1+IgM+IL-10+ and TIM-1+IgM+IL-10- B1a cells and IgM levels and attenuated progression of established atherosclerosis. CONCLUSIONS: RMT1-10 treatment attenuates atherosclerosis development and progression by selectively expanding IgM producing atheroprotective B1a cells. Antibody-based in vivo expansion of B1a cells could be an attractive approach for treating atherosclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , B-Lymphocytes, Regulatory/drug effects , Cell Proliferation/drug effects , Hepatitis A Virus Cellular Receptor 1/antagonists & inhibitors , Immunoglobulin M/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apoptosis/drug effects , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Disease Models, Animal , Disease Progression , Hepatitis A Virus Cellular Receptor 1/immunology , Hepatitis A Virus Cellular Receptor 1/metabolism , Immunoglobulin M/immunology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Plaque, Atherosclerotic , Signal Transduction/drug effects
7.
Arterioscler Thromb Vasc Biol ; 38(5): e71-e84, 2018 05.
Article in English | MEDLINE | ID: mdl-29599140

ABSTRACT

OBJECTIVE: B cells promote or protect development of atherosclerosis. In this study, we examined the role of MHCII (major histocompatibility II), CD40 (cluster of differentiation 40), and Blimp-1 (B-lymphocyte-induced maturation protein) expression by follicular B (FO B) cells in development of atherosclerosis together with the effects of IgG purified from atherosclerotic mice. APPROACH AND RESULTS: Using mixed chimeric Ldlr-/- mice whose B cells are deficient in MHCII or CD40, we demonstrate that these molecules are critical for the proatherogenic actions of FO B cells. During development of atherosclerosis, these deficiencies affected T-B cell interactions, germinal center B cells, plasma cells, and IgG. As FO B cells differentiating into plasma cells require Blimp-1, we also assessed its role in the development of atherosclerosis. Blimp-1-deficient B cells greatly attenuated atherosclerosis and immunoglobulin-including IgG production, preventing IgG accumulation in atherosclerotic lesions; Blimp-1 deletion also attenuated lesion proinflammatory cytokines, apoptotic cell numbers, and necrotic core. To determine the importance of IgG for atherosclerosis, we purified IgG from atherosclerotic mice. Their transfer but not IgG from nonatherosclerotic mice into Ldlr-/- mice whose B cells are Blimp-1-deficient increased atherosclerosis; transfer was associated with IgG accumulating in atherosclerotic lesions, increased lesion inflammatory cytokines, apoptotic cell numbers, and necrotic core size. CONCLUSIONS: The mechanism by which FO B cells promote atherosclerosis is highly dependent on their expression of MHCII, CD40, and Blimp-1. FO B cell differentiation into IgG-producing plasma cells also is critical for their proatherogenic actions. Targeting B-T cell interactions and pathogenic IgG may provide novel therapeutic strategies to prevent atherosclerosis and its adverse cardiovascular complications.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , Cell Differentiation , Germinal Center/immunology , Immunoglobulin G/immunology , Plasma Cells/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/metabolism , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , Disease Models, Animal , Disease Progression , Germinal Center/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunoglobulin G/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Phenotype , Plaque, Atherosclerotic , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Secretory Pathway , Signal Transduction , T-Lymphocytes/metabolism
8.
Proc Natl Acad Sci U S A ; 114(31): E6400-E6409, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28698369

ABSTRACT

T-follicular helper (Tfh) cells differentiate through a multistep process, culminating in germinal center (GC) localized GC-Tfh cells that provide support to GC-B cells. T-follicular regulatory (Tfr) cells have critical roles in the control of Tfh cells and GC formation. Although Tfh-cell differentiation is inhibited by IL-2, regulatory T (Treg) cell differentiation and survival depend on it. Here, we describe a CD25- subpopulation within both murine and human PD1+CXCR5+Foxp3+ Tfr cells. It is preferentially located in the GC and can be clearly differentiated from CD25+ non-GC-Tfr, Tfh, and effector Treg (eTreg) cells by the expression of a wide range of molecules. In comparison to CD25+ Tfr and eTreg cells, CD25- Tfr cells partially down-regulate IL-2-dependent canonical Treg features, but retain suppressive function, while simultaneously up-regulating genes associated with Tfh and GC-Tfh cells. We suggest that, similar to Tfh cells, Tfr cells follow a differentiation pathway generating a mature GC-localized subpopulation, CD25- Tfr cells.


Subject(s)
Germinal Center/cytology , Germinal Center/immunology , Interleukin-2 Receptor alpha Subunit/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Down-Regulation/immunology , Forkhead Transcription Factors/biosynthesis , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptors, CXCR5/biosynthesis , Repressor Proteins/biosynthesis
9.
J Am Heart Assoc ; 5(11)2016 11 14.
Article in English | MEDLINE | ID: mdl-27930350

ABSTRACT

BACKGROUND: We previously identified peritoneal B1a cells that secrete natural IgM as a key atheroprotective B cell subset. However, the molecules that activate atheroprotective B1a cells are unknown. Here, we investigated whether Toll-like receptors (TLRs) TLR2, TLR4, and TLR9 expressed by B1a cells are required for IgM-mediated atheroprotection. METHODS AND RESULTS: We adoptively transferred B1a cells from wild-type mice or from mice deficient in TLR2, TLR4, TLR9, or myeloid differentiation primary response 88 (MyD88) into ApoE-/- mice depleted of peritoneal B1a cells by splenectomy and fed a high-fat diet for 8 weeks. Elevations in plasma total, anti-oxLDL (oxidized low-density lipoprotein), anti-leukocyte, anti-CD3, anti-CD8, and anti-CD4 IgMs in atherosclerotic mice required B1a cells expressing TLR4 and MyD88, indicating a critical role for TLR4-MyD88 signaling for IgM secretion. Suppression of atherosclerosis was also critically dependent on B1a cells expressing TLR4-MyD88. Atherosclerosis suppression was associated not only with reductions in lesion apoptotic cells, necrotic cores, and oxLDL, but also with reduced lesion CD4+ and CD8+ T cells. Transforming growth factor beta 1 (TGF-ß1) expression, including macrophages expressing TGF-ß1, was increased, consistent with increased IgM-mediated phagocytosis of apoptotic cells by macrophages. Reductions in lesion inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin (IL) 1ß, and IL-18 were consistent with augmented TGF-ß1 expression. CONCLUSIONS: TLR4-MyD88 expression on B1a cells is critical for their IgM-dependent atheroprotection that not only reduced lesion apoptotic cells and necrotic cores, but also decreased CD4 and CD8 T-cell infiltrates and augmented TGF-ß1 expression accompanied by reduced lesion inflammatory cytokines TNF-α, IL-1ß, and IL-18.


Subject(s)
Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Immunoglobulin M/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 4/immunology , Animals , Atherosclerosis/genetics , B-Lymphocytes/immunology , Diet, High-Fat , Interleukin-18/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Knockout , Mice, Knockout, ApoE , Myeloid Differentiation Factor 88/genetics , Peritoneum/cytology , Peritoneum/immunology , Phagocytosis/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/immunology
10.
Cardiovasc Res ; 111(4): 385-97, 2016 09.
Article in English | MEDLINE | ID: mdl-27492217

ABSTRACT

AIMS: B2 lymphocytes promote atherosclerosis development but their mechanisms of action are unknown. Here, we investigated the role of tumour necrosis factor alpha (TNF-α) produced by B2 cells in atherogenesis. METHODS AND RESULTS: We found that 50% of TNF-α-producing spleen lymphocytes were B2 cells and ∼20% of spleen and aortic B cells produced TNF-α in hyperlipidemic ApoE(-/-) mice. We generated mixed bone marrow (80% µMT/20% TNF-α(-/-)) chimeric LDLR(-/-) mice where only B cells did not express TNF-α. Atherosclerosis was reduced in chimeric LDLR(-/-) mice with TNF-α-deficient B cells. TNF-α expression in atherosclerotic lesions and in macrophages were also reduced accompanied by fewer apoptotic cells, reduced necrotic cores, and reduced lesion Fas, interleukin-1ß and MCP-1 in mice with TNF-α-deficient B cells compared to mice with TNF-α-sufficient B cells. To confirm that the reduced atherosclerosis is attributable to B2 cells, we transferred wild-type and TNF-α-deficient B2 cells into ApoE(-/-) mice deficient in B cells or in lymphocytes. After 8 weeks of high fat diet, we found that atherosclerosis was increased by wild-type but not TNF-α-deficient B2 cells. Lesions of mice with wild-type B2 cells but not TNF-α-deficient B2 cells also had increased apoptotic cells and necrotic cores. Transferred B2 cells were found in lesions of recipient mice, suggesting that TNF-α-producing B2 cells promote atherosclerosis within lesions. CONCLUSION: We conclude that TNF-α produced by B2 cells is a key mechanism by which B2 cells promote atherogenesis through augmenting macrophage TNF-α production to induce cell death and inflammation that promote plaque vulnerability.


Subject(s)
B-Lymphocytes/metabolism , Inflammation/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Cell Death , Diet, High-Fat , Mice, Knockout , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
11.
Cardiovasc Res ; 106(3): 443-52, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25681396

ABSTRACT

AIMS: To investigate whether activation of atheroprotective peritoneal B1a cells by apoptotic cells or phosphatidylserine liposomes (PSLs) can enhance their protective actions during atherosclerosis development. METHODS AND RESULTS: Male apolipoprotein E-knockout (ApoE-/-) mice were treated with apoptotic cells or PSLs at the beginning of 8-week high-fat diet. Intraperitoneally administered apoptotic cells attenuated atherosclerosis in hypercholesterolemic ApoE-/- mice by 53% and macrophage accumulation by 52%, effects mimicked by administering PSLs and abolished by B1a cell depletion by splenectomy. These effects were associated with reduced lesion CD4+ and CD8+ T cells, mRNAs of MCP-1, VCAM-1, TNF-α, IL-1ß, IL-12, and IL-18 while anti-inflammatory TGF-ß mRNA levels doubled. Apoptotic cells or PSLs increased B1a lymphocytes including TIM-1+ B1a cells in vivo and in vitro while other lymphocyte populations were unaffected. Total plasma IgM, anti-leucocyte, anti-CD3, anti-CD4, and anti-oxLDL IgM were elevated. IgM in atherosclerotic lesions was also elevated and this was associated with reduced lesion MDA-LDL (oxLDL), apoptotic cells and necrotic core size. These effects of activating B1a cells could be attributed to B1a-derived polyreactive IgM deposited in lesions that reduce inflammatory cytokines by lowering lesion ox-LDL via anti-oxLDL IgM, T-cells via anti-leucocyte, anti-CD3, and anti-CD4 IgM, apoptotic cells and necrotic core size via IgM binding to apoptotic cells and enhancing phagocytosis, which also elevates anti-inflammatory cytokines. CONCLUSION: Targeting B1a cell activation by PSLs may be a potentially potent therapeutic strategy to attenuate atherosclerosis and reduce the incidence of atherosclerosis-dependent myocardial infarction and stroke.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apoptosis , Atherosclerosis/prevention & control , B-Lymphocyte Subsets/drug effects , Immunoglobulin M/biosynthesis , Lymphocyte Activation/drug effects , Phosphatidylserines/administration & dosage , Thymocytes/transplantation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Immunoglobulin M/immunology , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Liposomes , Male , Mice, Knockout , Necrosis , Phagocytosis , Phenotype , Splenectomy , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/pathology , Time Factors
12.
Arterioscler Thromb Vasc Biol ; 34(3): 516-25, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24436372

ABSTRACT

OBJECTIVE: Atherosclerosis is driven by inflammatory reactions that are shared with the innate immune system. Toll-like receptor-9 (TLR9) is an intracellular pattern recognition receptor of the innate immune system that is currently under clinical investigation as a therapeutic target in inflammatory diseases. Here, we investigated whether TLR9 has a role in the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. APPROACH AND RESULTS: Newly generated double-knockout ApoE(-/-):TLR9(-/-) mice and control ApoE(-/-) mice were fed a high-fat diet from 8 weeks and effects on lesion size, cellular composition, inflammatory status, and plasma lipids were assessed after 8, 12, 15, and 20 weeks. All 4 time points demonstrated exacerbated atherosclerotic lesion severity in ApoE(-/-):TLR9(-/-) mice, with a corresponding increase in lipid deposition and accumulation of macrophages, dendritic cells, and CD4(+) T cells. Although ApoE(-/-):TLR9(-/-) mice exhibited an increase in plasma very low-density lipoprotein/low-density-lipoprotein cholesterol, the very low-density lipoprotein/low-density lipoprotein:high-density lipoprotein ratio was unaltered because of a parallel increase in plasma high-density lipoprotein cholesterol. As a potential mechanism accounting for plaque progression in ApoE(-/-):TLR9(-/-) mice, CD4(+) T-cell accumulation was further investigated and depletion of these cells in ApoE(-/-):TLR9(-/-) mice significantly reduced lesion severity. As a final translational approach, administration of a TLR9 agonist (type B CpG oligodeoxynucleotide 1668) to ApoE(-/-) mice resulted in a reduction of lesion severity. CONCLUSIONS: Genetic deletion of the innate immune receptor TLR9 exacerbated atherosclerosis in ApoE(-/-) mice fed a high-fat diet. CD4(+) T cells were identified as potential mediators of this effect. A type B CpG oligodeoxynucleotide TLR9 agonist reduced lesion severity, thus identifying a novel therapeutic approach in atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Toll-Like Receptor 9/physiology , Animals , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Dietary Fats/toxicity , Disease Progression , Lipoproteins, HDL/blood , Lipoproteins, VLDL/blood , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/therapeutic use , Random Allocation , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
13.
PLoS One ; 8(4): e60430, 2013.
Article in English | MEDLINE | ID: mdl-23560095

ABSTRACT

AIMS: Option to attenuate atherosclerosis by depleting B2 cells is currently limited to anti-CD20 antibodies which deplete all B-cell subtypes. In the present study we evaluated the capacity of a monoclonal antibody to B cell activating factor-receptor (BAFFR) to selectively deplete atherogenic B2 cells to prevent both development and progression of atherosclerosis in the ApoE(-/-) mouse. METHODS AND RESULTS: To determine whether the BAFFR antibody prevents atherosclerosis development, we treated ApoE(-/-) mice with the antibody while feeding them a high fat diet (HFD) for 8 weeks. Mature CD93(-) CD19(+) B2 cells were reduced by treatment, spleen B-cell zones disrupted and spleen CD20 mRNA expression decreased while B1a cells and non-B cells were spared. Atherosclerosis was ameliorated in the hyperlipidemic mice and CD19(+) B cells, CD4(+) and CD8(+) T cells were reduced in atherosclerotic lesions. Expressions of proinflammatory cytokines, IL1ß, TNFα, and IFNγ in the lesions were also reduced, while MCP1, MIF and VCAM-1 expressions were unaffected. Plasma immunoglobulins were reduced, but MDA-oxLDL specific antibodies were unaffected. To determine whether anti-BAFFR antibody ameliorates progression of atherosclerosis, we first fed ApoE(-/-) mice a HFD for 6 weeks, and then instigated anti-BAFFR antibody treatment for a further 6 week-HFD. CD93(-) CD19(+) B2 cells were selectively decreased and atherosclerotic lesions were reduced by this treatment. CONCLUSION: Anti-BAFFR monoclonal antibody selectively depletes mature B2 cells while sparing B1a cells, disrupts spleen B-cell zones and ameliorates atherosclerosis development and progression in hyperlipidemic ApoE(-/-) mice. Our findings have potential for clinical translation to manage atherosclerosis-based cardiovascular diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , B-Lymphocyte Subsets/drug effects , Hyperlipidemias/drug therapy , Spleen/drug effects , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Atherosclerosis/complications , Atherosclerosis/immunology , Atherosclerosis/pathology , B-Cell Activation Factor Receptor/antagonists & inhibitors , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cytokines/biosynthesis , Cytokines/immunology , Diet, High-Fat , Disease Progression , Hyperlipidemias/complications , Hyperlipidemias/immunology , Hyperlipidemias/pathology , Lymphocyte Depletion , Male , Mice , Mice, Knockout , Spleen/immunology , Spleen/pathology
14.
Circulation ; 127(9): 1028-39, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23395974

ABSTRACT

BACKGROUND: Heart attacks and strokes, leading causes of deaths globally, arise from thrombotic occlusion of ruptured vulnerable atherosclerotic plaques characterized by abundant apoptosis, large necrotic cores derived from inefficient apoptotic cell clearance, thin fibrous caps, and focal inflammation. The genesis of apoptosis and necrotic cores in these vulnerable atherosclerotic plaques remains unknown. Cytotoxic CD8(+) T lymphocytes represent up to 50% of leukocytes in advanced human plaques and dominate early immune responses in mouse lesions, yet their role in atherosclerosis also remains unresolved. METHODS AND RESULTS: CD8(+) T-lymphocyte depletion by CD8α or CD8ß monoclonal antibody in apolipoprotein E-deficient mice fed a high-fat diet ameliorated atherosclerosis by reducing lipid and macrophage accumulation, apoptosis, necrotic cores, and monocyte chemoattractant protein 1, interleukin 1ß, interferon γ, and vascular cell adhesion molecule 1. Transfer of CD8(+) T cells into lymphocyte-deficient, apolipoprotein E-deficient mice partially reconstituted CD8(+) T cells in lymphoid compartments and was associated with CD8(+) T-cell infiltration in lesions, increased lipid and macrophage accumulation, apoptotic cells, necrotic cores, and interleukin 1ß in atherosclerotic lesions. Transfer of CD8(+) T cells deficient in perforin, granzyme B, or tumor necrosis factor α but not interferon γ failed to increase atherosclerotic lesions despite partial reconstitution in the lymphoid system and the presence in atherosclerotic lesions. Macrophages, smooth muscle cells, and endothelial cells were identified as apoptotic targets. CONCLUSIONS: We conclude that CD8(+) T lymphocytes promote the development of vulnerable atherosclerotic plaques by perforin- and granzyme B-mediated apoptosis of macrophages, smooth muscle cells, and endothelial cells that, in turn, leads to necrotic core formation and further augments inflammation by tumor necrosis factor α secretion.


Subject(s)
Apolipoproteins E/deficiency , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Diet, High-Fat/adverse effects , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/genetics , T-Lymphocytes, Cytotoxic/metabolism
15.
PLoS One ; 7(1): e29371, 2012.
Article in English | MEDLINE | ID: mdl-22238605

ABSTRACT

We have recently identified conventional B2 cells as atherogenic and B1a cells as atheroprotective in hypercholesterolemic ApoE(-/-) mice. Here, we examined the development of atherosclerosis in BAFF-R deficient ApoE(-/-) mice because B2 cells but not B1a cells are selectively depleted in BAFF-R deficient mice. We fed BAFF-R(-/-) ApoE(-/-) (BaffR.ApoE DKO) and BAFF-R(+/+)ApoE(-/-) (ApoE KO) mice a high fat diet (HFD) for 8-weeks. B2 cells were significantly reduced by 82%, 81%, 94%, 72% in blood, peritoneal fluid, spleen and peripheral lymph nodes respectively; while B1a cells and non-B lymphocytes were unaffected. Aortic atherosclerotic lesions assessed by oil red-O stained-lipid accumulation and CD68+ macrophage accumulation were decreased by 44% and 50% respectively. B cells were absent in atherosclerotic lesions of BaffR.ApoE DKO mice as were IgG1 and IgG2a immunoglobulins produced by B2 cells, despite low but measurable numbers of B2 cells and IgG1 and IgG2a immunoglobulin concentrations in plasma. Plasma IgM and IgM deposits in atherosclerotic lesions were also reduced. BAFF-R deficiency in ApoE(-/-) mice was also associated with a reduced expression of VCAM-1 and fewer macrophages, dendritic cells, CD4+ and CD8+ T cell infiltrates and PCNA+ cells in lesions. The expression of proinflammatory cytokines, TNF-α, IL1-ß and proinflammatory chemokine MCP-1 was also reduced. Body weight and plasma cholesterols were unaffected in BaffR.ApoE DKO mice. Our data indicate that B2 cells are important contributors to the development of atherosclerosis and that targeting the BAFF-R to specifically reduce atherogenic B2 cell numbers while preserving atheroprotective B1a cell numbers may be a potential therapeutic strategy to reduce atherosclerosis by potently reducing arterial inflammation.


Subject(s)
Apolipoproteins E/genetics , Arteritis/prevention & control , Atherosclerosis/pathology , Cytoprotection/genetics , Endothelial Cells/physiology , Animals , Apolipoproteins E/physiology , Arteries/cytology , Arteries/metabolism , Arteries/pathology , Arteritis/complications , Arteritis/genetics , Arteritis/pathology , Atherosclerosis/complications , Atherosclerosis/genetics , Atherosclerosis/metabolism , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/physiology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/physiology , Endothelial Cells/classification , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Circ Res ; 109(8): 830-40, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21868694

ABSTRACT

RATIONALE: Aggravated atherosclerosis in B lymphocyte-deficient chimeric mice and reduced atherosclerosis after transfer of unfractionated spleen B lymphocytes into splenectomized mice have led to the widely held notion that B lymphocytes are atheroprotective. However, B lymphocytes can be pathogenic, because their depletion by anti-CD20 antibody ameliorated atherosclerosis, and transfer of B2 lymphocytes aggravated atherosclerosis. These observations raise the question of the identity of the atheroprotective B-lymphocyte population. OBJECTIVE: The purpose of the study was to identify an atheroprotective B-lymphocyte subset and mechanisms by which they confer atheroprotection. METHODS AND RESULTS: Splenectomy of apolipoprotein E-deficient mice selectively reduced peritoneal B1a lymphocytes, plasma IgM, and oxidized low-density lipoprotein IgM levels and lesion IgM deposits. These reductions were accompanied by increased oil red O-stained atherosclerotic lesions and increased necrotic cores, oxidized low-density lipoproteins, and apoptotic cells in lesions. Plasma lipids, body weight, collagen, and smooth muscle content were unaffected. Transfer of B1a lymphocytes into splenectomized mice increased peritoneal B1a lymphocytes; restored plasma IgM, oxidized low-density lipoprotein IgM levels, and lesion IgM deposits; and potently attenuated atherosclerotic lesions, with reduced lesion necrotic cores, oxidized low-density lipoprotein, and apoptotic cells. In contrast, transfer of B1a lymphocytes that cannot secrete IgM failed to protect against atherosclerosis development in splenectomized mice despite reconstitution in the peritoneum. CONCLUSIONS: B1a lymphocytes are an atheroprotective B-lymphocyte population. Our data suggest that natural IgM secreted by these lymphocytes offers protection by depositing IgM in atherosclerotic lesions, which reduces the necrotic cores of lesions.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/prevention & control , B-Lymphocyte Subsets/metabolism , Immunoglobulin M/metabolism , Tunica Intima/pathology , Animals , Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/transplantation , Immunoglobulin M/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Splenectomy
17.
J Immunol ; 185(7): 4410-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20817865

ABSTRACT

Atherosclerosis is a chronic inflammatory arterial disease characterized by focal accumulation of lipid and inflammatory cells. It is the number one cause of deaths in the Western world because of its complications of heart attacks and strokes. Statins are effective in only approximately one third of patients, underscoring the urgent need for additional therapies. B cells that accumulate in atherosclerotic lesions and the aortic adventitia of humans and mice are considered to protect against atherosclerosis development. Unexpectedly, we found that selective B cell depletion in apolipoprotein E-deficient (ApoE(-/-)) mice using a well-characterized mAb to mouse CD20 reduced atherosclerosis development and progression without affecting the hyperlipidemia imposed by a high-fat diet. Adoptive transfer of 5 × 10(6) or 5 × 10(7) conventional B2 B cells but not 5 × 10(6) B1 B cells to a lymphocyte-deficient ApoE(-/-) Rag-2(-/-) common cytokine receptor γ-chain-deficient mouse that was fed a high-fat diet augmented atherosclerosis by 72%. Transfer of 5 × 10(6) B2 B cells to an ApoE(-/-) mouse deficient only in B cells aggravated atherosclerosis by >300%. Our findings provide compelling evidence for the hitherto unrecognized proatherogenic role of conventional B2 cells. The data indicate that B2 cells can potently promote atherosclerosis development entirely on their own in the total absence of all other lymphocyte populations. Additionally, these B2 cells can also significantly augment atherosclerosis development in the presence of T cells and all other lymphocyte populations. Our findings raise the prospect of B cell depletion as a therapeutic approach to inhibit atherosclerosis development and progression in humans.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , Lymphocyte Depletion , Adoptive Transfer/adverse effects , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived , Atherosclerosis/pathology , Atherosclerosis/therapy , Cell Separation , Flow Cytometry , Immunohistochemistry , Lymphocyte Depletion/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Rituximab
SELECTION OF CITATIONS
SEARCH DETAIL
...