Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 52(1): 83-95.e4, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31882362

ABSTRACT

Lymphoid tissue inducer (LTi) cells are regarded as a subset of innate lymphoid cells (ILCs). However, these cells are not derived from the ILC common progenitor, which generates other ILC subsets and is defined by the expression of the transcription factor PLZF. Here, we examined transcription factor(s) determining the fate of LTi progenitors versus non-LTi ILC progenitors. Conditional deletion of Gata3 resulted in the loss of PLZF+ non-LTi progenitors but not the LTi progenitors that expressed the transcription factor RORγt. Consistently, PLZF+ non-LTi progenitors expressed high amounts of GATA3, whereas GATA3 expression was low in RORγt+ LTi progenitors. The generation of both progenitors required the transcriptional regulator Id2, which defines the common helper-like innate lymphoid progenitor (ChILP), but not cytokine signaling. Nevertheless, low GATA3 expression was necessary for the generation of functionally mature LTi cells. Thus, differential expression of GATA3 determines the fates and functions of distinct ILC progenitors.


Subject(s)
GATA3 Transcription Factor/biosynthesis , Stem Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Lineage/immunology , Cells, Cultured , GATA3 Transcription Factor/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Stem Cells/immunology , T-Lymphocyte Subsets/immunology
2.
Immunity ; 46(1): 51-64, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099864

ABSTRACT

Despite the importance of programmed cell death-1 (PD-1) in inhibiting T cell effector activity, the mechanisms regulating its expression remain poorly defined. We found that the chromatin organizer special AT-rich sequence-binding protein-1 (Satb1) restrains PD-1 expression induced upon T cell activation by recruiting a nucleosome remodeling deacetylase (NuRD) complex to Pdcd1 regulatory regions. Satb1 deficienct T cells exhibited a 40-fold increase in PD-1 expression. Tumor-derived transforming growth factor ß (Tgf-ß) decreased Satb1 expression through binding of Smad proteins to the Satb1 promoter. Smad proteins also competed with the Satb1-NuRD complex for binding to Pdcd1 enhancers, releasing Pdcd1 expression from Satb1-mediated repression, Satb1-deficient tumor-reactive T cells lost effector activity more rapidly than wild-type lymphocytes at tumor beds expressing PD-1 ligand (CD274), and these differences were abrogated by sustained CD274 blockade. Our findings suggest that Satb1 functions to prevent premature T cell exhaustion by regulating Pdcd1 expression upon T cell activation. Dysregulation of this pathway in tumor-infiltrating T cells results in diminished anti-tumor immunity.


Subject(s)
Epigenetic Repression/immunology , Gene Expression Regulation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Matrix Attachment Region Binding Proteins/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Animals , Enzyme-Linked Immunospot Assay , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Matrix Attachment Region Binding Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism
3.
Immunity ; 41(2): 219-29, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25131532

ABSTRACT

The origin and developmental pathway of intestinal T cell receptor αß(+) CD4(-)CD8ß(-) intraepithelial lymphocytes (unconventional iIELs), a major population of innate-like resident cytolytic T cells, have remained elusive. By cloning and expressing several TCRs isolated from unconventional iIELs, we identified immature CD4(lo)CD8(lo)(DP(lo))CD69(hi)PD-1(hi) thymocytes as the earliest postsignaling precursors for these cells. Although these precursors displayed multiple signs of elevated TCR signaling, a sizeable fraction of them escaped deletion to selectively engage in unconventional iIEL differentiation. Conversely, TCRs cloned from DP(lo)CD69(hi)PD-1(hi) thymocytes, a population enriched in autoreactive thymocytes, selectively gave rise to unconventional iIELs upon transgenic expression. Thus, the unconventional iIEL precursor overlaps with the DP(lo) population undergoing negative selection, indicating that, concomitant with the downregulation of both CD4 and CD8 coreceptors, a balance between apoptosis and survival signals results in outcomes as divergent as clonal deletion and differentiation to the unconventional iIEL lineage.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Cells, Cultured , Clonal Deletion/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology
4.
Immunity ; 41(5): 802-14, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25464856

ABSTRACT

Protein kinase B (also known as AKT) and the mechanistic target of rapamycin (mTOR) are central regulators of T cell differentiation, proliferation, metabolism, and survival. Here, we show that during chronic murine lymphocytic choriomeningitis virus infection, activation of AKT and mTOR are impaired in antiviral cytotoxic T lymphocytes (CTLs), resulting in enhanced activity of the transcription factor FoxO1. Blockade of inhibitory receptor programmed cell death protein 1 (PD-1) in vivo increased mTOR activity in virus-specific CTLs, and its therapeutic effects were abrogated by the mTOR inhibitor rapamycin. FoxO1 functioned as a transcriptional activator of PD-1 that promoted the differentiation of terminally exhausted CTLs. Importantly, FoxO1-null CTLs failed to persist and control chronic viral infection. Collectively, this study shows that CTLs adapt to persistent infection through a positive feedback pathway (PD-1?FoxO1?PD-1) that functions to both desensitize virus-specific CTLs to antigen and support their survival during chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Chronic Disease , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Granzymes/biosynthesis , Humans , Interferon-gamma/biosynthesis , Jurkat Cells , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-akt/biosynthesis , Receptors, Antigen, T-Cell/immunology , Sirolimus/pharmacology , T-Lymphocytes, Cytotoxic/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/biosynthesis
5.
Nature ; 548(7669): 602-606, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28847007

ABSTRACT

Regulatory T cells (Treg cells) have a pivotal role in the establishment and maintenance of immunological self-tolerance and homeostasis. Transcriptional programming of regulatory mechanisms facilitates the functional activation of Treg cells in the prevention of diverse types of inflammatory responses. It remains unclear how Treg cells orchestrate their homeostasis and interplay with environmental signals. Here we show that liver kinase B1 (LKB1) programs the metabolic and functional fitness of Treg cells in the control of immune tolerance and homeostasis. Mice with a Treg-specific deletion of LKB1 developed a fatal inflammatory disease characterized by excessive TH2-type-dominant responses. LKB1 deficiency disrupted Treg cell survival and mitochondrial fitness and metabolism, but also induced aberrant expression of immune regulatory molecules including the negative co-receptor PD-1 and the TNF receptor superfamily proteins GITR and OX40. Unexpectedly, LKB1 function in Treg cells was independent of conventional AMPK signalling or the mTORC1-HIF-1α axis, but contributed to the activation of ß-catenin signalling for the control of PD-1 and TNF receptor proteins. Blockade of PD-1 activity reinvigorated the ability of LKB1-deficient Treg cells to suppress TH2 responses and the interplay with dendritic cells primed by thymic stromal lymphopoietin. Thus, Treg cells use LKB1 signalling to coordinate their metabolic and immunological homeostasis and to prevent apoptotic and functional exhaustion, thereby orchestrating the balance between immunity and tolerance.


Subject(s)
Homeostasis , Immune Tolerance , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , AMP-Activated Protein Kinases , Animals , Apoptosis , Cell Survival/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Mice , Mitochondria/metabolism , Mitochondria/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, OX40/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Th2 Cells/immunology , beta Catenin/metabolism , Thymic Stromal Lymphopoietin
6.
Nature ; 534(7607): 402-6, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281199

ABSTRACT

Successful treatment of many patients with advanced cancer using antibodies against programmed cell death 1 (PD-1; also known as PDCD1) and its ligand (PD-L1; also known as CD274) has highlighted the critical importance of PD-1/PD-L1-mediated immune escape in cancer development. However, the genetic basis for the immune escape has not been fully elucidated, with the exception of elevated PD-L1 expression by gene amplification and utilization of an ectopic promoter by translocation, as reported in Hodgkin and other B-cell lymphomas, as well as stomach adenocarcinoma. Here we show a unique genetic mechanism of immune escape caused by structural variations (SVs) commonly disrupting the 3' region of the PD-L1 gene. Widely affecting multiple common human cancer types, including adult T-cell leukaemia/lymphoma (27%), diffuse large B-cell lymphoma (8%), and stomach adenocarcinoma (2%), these SVs invariably lead to a marked elevation of aberrant PD-L1 transcripts that are stabilized by truncation of the 3'-untranslated region (UTR). Disruption of the Pd-l1 3'-UTR in mice enables immune evasion of EG7-OVA tumour cells with elevated Pd-l1 expression in vivo, which is effectively inhibited by Pd-1/Pd-l1 blockade, supporting the role of relevant SVs in clonal selection through immune evasion. Our findings not only unmask a novel regulatory mechanism of PD-L1 expression, but also suggest that PD-L1 3'-UTR disruption could serve as a genetic marker to identify cancers that actively evade anti-tumour immunity through PD-L1 overexpression.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Tumor Escape/genetics , Up-Regulation , Adenocarcinoma/genetics , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , Clonal Selection, Antigen-Mediated , Female , Genetic Markers/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics
7.
Prostate ; 81(1): 50-57, 2021 01.
Article in English | MEDLINE | ID: mdl-32986884

ABSTRACT

BACKGROUND: Characterization of markers of both immune suppression and activation may provide more prognostic information than assessment of single markers in localized prostate cancer. We therefore sought to determine the association between CD8 and PD-L1 expression in localized prostate tumors and biochemical recurrence (BCR) and metastasis-free survival (MFS). METHODS: Tissue microarrays were constructed on 109 men undergoing radical prostatectomy (RP) for localized prostate cancer at Dana-Farber Cancer Institute between 1991 and 2008. Fluorescence immunohistochemistry was used to evaluate the expression of six immune markers (CD3, CD4, CD8, PD-1, PD-L1, FOXP3). Quantitative multispectral imaging analysis was used to calculate the density of each marker, which was dichotomized by the median as "high" or "low." Cox proportional hazards regression models and Kaplan-Meier analyses were used to analyze associations between immune marker densities and time to BCR and MFS. RESULTS: Over a median follow-up of 8.1 years, 55 (51%) and 39 (36%) men developed BCR and metastases, respectively. Median time to BCR was shorter in men with low CD8 (hazard ratio [HR] = 2.27 [1.27-4.08]) and high PD-L1 expression (HR = 2.03 [1.17-3.53]). While neither low CD8 or high PD-L1 alone were independent predictors of BCR or MFS on multivariable analysis, men with low CD8 and/or high PD-L1 had a significantly shorter time to BCR (median 3.5 years vs. NR) and MFS (median 10.8 vs. 18.4 years) compared to those with high CD8 and low PD-L1 expression. The main limitation is the retrospective and singe-center nature of the study. CONCLUSION: The presence of higher CD8 and lower PD-L1 expression in prostatectomy specimens was associated a low risk of biochemical relapse and metastatic disease. These findings are hypothesis-generating and further study is needed.


Subject(s)
B7-H1 Antigen/biosynthesis , CD8 Antigens/biosynthesis , Prostatic Neoplasms/immunology , B7-H1 Antigen/immunology , CD3 Complex/biosynthesis , CD3 Complex/immunology , CD8 Antigens/immunology , Cohort Studies , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/immunology , Proportional Hazards Models , Prostatectomy , Prostatic Neoplasms/surgery , Retrospective Studies , Tissue Array Analysis
8.
J Virol ; 94(14)2020 07 01.
Article in English | MEDLINE | ID: mdl-32350076

ABSTRACT

While the relationship of protective human leukocyte antigen (HLA) class I alleles and HIV progression is well defined, the interaction of HLA-mediated protection and CD8 T-cell exhaustion is less well characterized. To gain insight into the influence of HLA-B*57:01 on the deterioration of CD8 T-cell responses during HIV infection in the absence of antiretroviral treatment, we compared HLA-B*57:01-restricted HIV-specific CD8 T-cell responses to responses restricted by other HLA class I alleles longitudinally after control of peak viremia. Detailed characterization of polyfunctionality, differentiation phenotypes, transcription factor, and inhibitory receptor expression revealed progression of CD8 T-cell exhaustion over the course of the infection in both patient groups. However, early effects on the phenotype of the total CD8 T-cell population were apparent only in HLA-B*57-negative patients. The HLA-B*57:01-restricted, HIV epitope-specific CD8 T-cell responses showed beneficial functional patterns and significantly lower frequencies of inhibitory receptor expression, i.e., PD-1 and coexpression of PD-1 and TIGIT, within the first year of infection. Coexpression of PD-1 and TIGIT was correlated with clinical markers of disease progression and declining percentages of the T-bethi Eomesdim CD8 T-cell population. In accordance with clinical and immunological deterioration in the HLA-B*57:01 group, the difference in PD-1 and TIGIT receptor expression did not persist to later stages of the disease.IMPORTANCE Given the synergistic nature of TIGIT and PD-1, the coexpression of those inhibitory receptors should be considered when evaluating T-cell pathogenesis, developing immunomodulatory therapies or vaccines for HIV, and when using immunotherapy or vaccination for other causes in HIV-infected patients. HIV-mediated T-cell exhaustion influences the patient´s disease progression, immune system and subsequently non-AIDS complications, and efficacy of vaccinations against other pathogens. Consequently, the possibilities of interfering with exhaustion are numerous. Expanding the use of immunomodulatory therapies to include HIV treatment depends on information about possible targets and their role in the deterioration of the immune system. Furthermore, the rise of immunotherapies against cancer and elevated cancer incidence in HIV-infected patients together increase the need for detailed knowledge of T-cell exhaustion and possible interactions. A broader approach to counteract immune exhaustion to alleviate complications and improve efficacy of other vaccines also promises to increase patients' health and quality of life.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , HIV Infections/metabolism , HIV-1/metabolism , HLA-B Antigens/metabolism , Programmed Cell Death 1 Receptor/biosynthesis , Receptors, Immunologic/biosynthesis , Adult , CD8-Positive T-Lymphocytes/pathology , Female , HIV Infections/pathology , Humans , Male , Middle Aged
9.
Cell Immunol ; 369: 104435, 2021 11.
Article in English | MEDLINE | ID: mdl-34482260

ABSTRACT

The PD-1/PD-L1 axis plays a crucial role in regulating the anti-tumour immune response. A soluble PD-1 protein (sPD-1) has previously been observed, which could block the binding of PD-L1 to PD-1. Tumour associated macrophages are abundant in tumours, and evidence suggest they express PD-1. However, whether they also express sPD-1 remains unclear. The objective of this study was to investigate expression of sPD-1 in two in vitro models of human macrophages: THP-1 cells and monocyte-derived macrophages (MDM). Cells were polarised with either LPS + IFN-γ or IL-4 + IL-13 or left unpolarised. PD-1 and sPD-1 mRNAs were measured using droplet digital PCR, sPD-1 protein by electrochemiluminescence immunoassay and PD-1 by flow cytometry. sPD-1 mRNA was induced in both THP-1 cells and MDM after polarisation with LPS + IFN-γ, while IL-4 + IL-13 induced sPD-1 mRNA in MDM only. sPD-1 protein was measurable in culture supernatants. These findings show that macrophages can be induced to express sPD-1.


Subject(s)
Programmed Cell Death 1 Receptor/biosynthesis , Tumor-Associated Macrophages/immunology , Humans , Macrophage Activation/immunology , Protein Isoforms , THP-1 Cells
10.
Microb Pathog ; 153: 104779, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33548481

ABSTRACT

BACKGROUND: During viral infection, inhibitory receptors play a key role in regulating CD8 T-cell activity. The objective of this research was to investigate programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin domain-containing protein-3 (TIM-3), and CD39 exhaustion markers in CD8 T cells of new coronavirus disease-2019 (COVID-19) patients. METHODS: A total of 44 patients with COVID-19 (17 subjects in a critical group and 27 patients in a non-critical group) and 14 healthy controls, who were admitted to Hospitals in Babol, were recruited to the study. In subjects' peripheral blood mononuclear cells (PBMCs), we compared the phenotype of CD8 T lymphocytes, expressing PD-1, TIM-3, or CD39, both alone and in various combinations. RESULTS: The findings showed that the percentage of CD8+ cells was significantly lower in patients. Critical and non-critical patients were more likely than healthy controls to have an escalated frequency of CD8+ TIM-3+, CD8+ CD39+, and CD8+ TIM-3+ CD39+ cells. No significant differences were observed between all groups in the CD8+ PD-1+ cell counts. There was also no difference between three groups regarding the counts of CD8+ TIM-3+ PD-1+, CD8+ PD-1+ CD39+, and CD8+ TIM-3+ PD-1+ CD39+ cells. The counts of non-exhausted cells were significantly lower in critical and non-critical individuals compared to the healthy individuals' value. CONCLUSION: Patients, infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), altered exhausted CD8 T lymphocytes with CD39 and TIM-3 exhaustion markers, which may account the dysregulated immune response found in COVID-19.


Subject(s)
Apyrase/biosynthesis , CD8-Positive T-Lymphocytes/immunology , COVID-19/pathology , Hepatitis A Virus Cellular Receptor 2/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Female , Humans , Iran , Lymphocyte Count , Male , Middle Aged , SARS-CoV-2/immunology , Young Adult
11.
Histopathology ; 79(1): 36-46, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33326644

ABSTRACT

AIMS: Immunotherapies represent a new alternative therapeutic approach for hepatocellular carcinomas (HCCs), and have shown promising results when used in combination therapy. The aim of this study was to evaluate the potential of transarterial chemoembolisation (TACE) to modulate programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) expression profiles in a cohort of surgically treated HCCs. METHODS AND RESULTS: A total of 82 surgically treated HCCs from patients who had undergone (n = 32) or not undergone (n = 50) preoperative TACE were included in the study. Immunohistochemical expression of PD-1 and of PD-L1 were analysed and compared according to TACE treatment. Pretreatment biopsies, which were available for 30 cases (20 with TACE and 10 without), were similarly analysed. Follow-up data were retrieved from patients' charts. PD-1 expression (≥1%) in intratumoral inflammatory cells (ICs) was observed in 46% of HCCs, and PD-L1 expression (≥1%) in ICs and PD-L1 expression in tumour cells (TCs) were observed in 46% and 16% of HCCs, respectively. A low level of PD-1 expression (<1%) was associated with strong and diffuse glutamine synthetase overexpression (8% versus 27%, P = 0.024). HCCs from patients with TACE pretreatment showed significantly higher PD-L1 expression in TCs than those from patients without TACE pretreatment (2% versus 0.4%, P = 0.027). PD-1 expression in ICs and PD-L1 expression in both ICs and TCs were higher in TACE-resected tumours than in corresponding pre-TACE biopsies (respectively: 1.8% versus 8.1%, P = 0.034; 0.8% versus 7.1%, P = 0.032; and 0% versus 2.4%, P = 0.043). CONCLUSION: Our results, showing increases in PD-1 expression and PD-L1 expression in HCCs following TACE, support the use of TACE in combination with immunotherapy in selected cases to optimise tumour response.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Hepatocellular/metabolism , Chemoembolization, Therapeutic/methods , Liver Neoplasms/therapy , Programmed Cell Death 1 Receptor/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/therapy , Cohort Studies , Female , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , Transcriptome
12.
Mol Cell Biochem ; 476(4): 1717-1727, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33428059

ABSTRACT

Endometriosis is an estrogen-dependent, inflammatory gynecological disorder characterized by the growth of endometrial cells in lesions outside the uterus. Bone marrow-derived cells (BMDCs) engraft lesions and increase lesion size. Do endometriosis cells regulate differentiation of engrafted BMDCs in the pathogenesis and growth of endometriosis? Here, we report endometriosis derived stromal cells promote the differentiation of BMDCs to stromal, epithelial and leukocyte cell fates through paracrine signaling. In-vitro studies demonstrated that both mRNA and protein levels of vimentin, cytokeratin and PD-1 were significantly increased in BMDCs cocultured with stromal cells from endometriosis (ENDO) patients compared to stromal cells from normal endometrium (CNTL). Increased expression of PD-1 has been reported in malignancy where it promotes T cell quiescence and immune tolerance. Increased PD-1 was also confirmed in-vivo where we showed that PD-1 expression was induced in BMDCs engrafted into endometriotic lesions in a murine model of endometriosis. AMD3100, an antagonist for CXCR4 receptor inhibited PD-1 expression in BMDCs suggesting that PD-1 induction requires CXCL12. These results suggest that endometriosis stimulated BMDC differentiation through paracrine signaling and increased T cell PD-1 expression. Increased PD-1 expression may be one mechanism by which endometriosis avoids immune surveillance.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Endometriosis/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Paracrine Communication , Programmed Cell Death 1 Receptor/biosynthesis , Adolescent , Adult , Bone Marrow Cells/pathology , Coculture Techniques , Endometriosis/pathology , Female , Humans , Middle Aged , Stromal Cells/metabolism , Stromal Cells/pathology
13.
J Immunol ; 203(4): 899-910, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31285277

ABSTRACT

The kynurenine pathway (KP) is a key regulator of many important physiological processes and plays a harmful role in cancer, many neurologic conditions, and chronic viral infections. In HIV infection, KP activity is consistently associated with reduced CD4 T cell counts and elevated levels of T cell activation and viral load; it also independently predicts mortality and morbidity from non-AIDS events. Kynurenine 3-monooxygenase (KMO) is a therapeutically important target in the KP. Using the nonhuman primate model of SIV infection in rhesus macaques, we investigated whether KMO inhibition could slow the course of disease progression. We used a KMO inhibitor, CHDI-340246, to perturb the KP during early acute infection and followed the animals for 1 y to assess clinical outcomes and immune phenotype and function during pre-combination antiretroviral therapy acute infection and combination antiretroviral therapy-treated chronic infection. Inhibition of KMO in acute SIV infection disrupted the KP and prevented SIV-induced increases in downstream metabolites, improving clinical outcome as measured by both increased CD4+ T cell counts and body weight. KMO inhibition increased naive T cell frequency and lowered PD-1 expression in naive and memory T cell subsets. Importantly, early PD-1 expression during acute SIV infection predicted clinical outcomes of body weight and CD4+ T cell counts. Our data indicate that KMO inhibition in early acute SIV infection provides clinical benefit and suggest a rationale for testing KMO inhibition as an adjunctive treatment in SIV/HIV infection to slow the progression of the disease and improve immune reconstitution.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , Pyrimidines/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Anti-Retroviral Agents/pharmacology , Body Weight/drug effects , CD4-Positive T-Lymphocytes/drug effects , Enzyme Inhibitors/pharmacology , Macaca mulatta , Programmed Cell Death 1 Receptor/drug effects , Simian Acquired Immunodeficiency Syndrome/metabolism
14.
J Immunol ; 203(3): 696-704, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31209101

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune disease with no curative treatment. The immune regulatory properties of type I IFNs have led to the approval of IFN-ß for the treatment of relapsing-remitting MS. However, there is still an unmet need to improve the tolerability and efficacy of this therapy. In this work, we evaluated the sustained delivery of IFN-α1, either alone or fused to apolipoprotein A-1 by means of an adeno-associated viral (AAV) system in the mouse model of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. These in vivo experiments demonstrated the prophylactic and therapeutic efficacy of the AAV-IFN-α or AAV-IFN-α fused to apolipoprotein A-1 vectors in experimental autoimmune encephalomyelitis, even at low doses devoid of hematological or neurologic toxicity. The sustained delivery of such low-dose IFN-α resulted in immunomodulatory effects, consisting of proinflammatory monocyte and T regulatory cell expansion. Moreover, encephalitogenic T lymphocytes from IFN-α-treated mice re-exposed to the myelin oligodendrocyte glycoprotein peptide in vitro showed a reduced proliferative response and cytokine (IL-17A and IFN-γ) production, in addition to upregulation of immunosuppressive molecules, such as IL-10, IDO, or PD-1. In conclusion, the results of the present work support the potential of sustained delivery of low-dose IFN-α for the treatment of MS and likely other T cell-dependent chronic autoimmune disorders.


Subject(s)
Apolipoprotein A-I/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-alpha/therapeutic use , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes, Regulatory/cytology
15.
J Cutan Pathol ; 48(2): 221-228, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32779238

ABSTRACT

BACKGROUND: Kaposi sarcoma (KS) is a mesenchymal tumor with distinct histopathological features according to stage of progression. Programmed death-1 (PD-1) and its ligand PD-L1 play major roles in the immune escape strategy of tumors. OBJECTIVES: This study evaluated expression of PD-1 and PD-L1 in various stages of KS and investigated associations between their expression and clinical characteristics. METHODS: Fifty cases with histopathologically diagnosed KS were classified as early or late stage. These specimens were stained with anti-PD-1 and anti-PD-L1 antibodies. The extent of expression in the intratumoral and peritumoral areas was judged by two dermatopathologists. RESULTS: PD-1 and PD-L1 were expressed in 72.2% (13/18) and 11.1% (2/18) of early-stage cases, respectively, compared with 43.8% (14/32) and 28.1% (9/32) of late-stage cases, respectively. At the late stage, PD-1 expression was significantly higher in the peritumoral area than in the intratumoral area (P = 0.001). PD-1 expression in the intratumoral area was significantly higher at the early stage than at the late stage (P = 0.013). PD-L1 expression in the peritumoral area was significantly higher at the late stage than at the early stage (P = 0.038). CONCLUSIONS: The pattern of PD-1 and PD-L1 expression differs according to the stage of KS, but is unaffected by clinical variables.


Subject(s)
B7-H1 Antigen/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Sarcoma, Kaposi , Skin Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Staging , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
16.
J Clin Apher ; 36(4): 645-648, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33648026

ABSTRACT

Novel immune-modulating anticancer drugs are being used with increasing frequency. With increased use, there are more frequent cases of toxicities caused by these drugs, termed immune-related adverse events (irAEs). We present a case in which we successfully treated a case of severe, steroid-refractory, nivolumab-induced myocarditis with therapeutic plasma exchange (TPE). Nivolumab is an immune checkpoint inhibitor (ICI) which blocks programmed death receptor-1 (PD-1). This blockade allows for enhanced T-cell function and increased anti-tumor response. The patient presented with signs and symptoms of heart failure and was found to have a significantly depressed cardiac ejection fraction. Over the course of her five TPE procedures, she improved clinically and was discharged home with improved left ventricular ejection function. This case suggests an emerging role of TPE in the management of severe ICI-induced toxicity, such as myocarditis.


Subject(s)
Immune Checkpoint Inhibitors/toxicity , Plasma Exchange/methods , Abatacept , Adrenal Cortex Hormones/therapeutic use , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/secondary , Aged , Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/drug therapy , Female , Humans , Immune System , Mycophenolic Acid/adverse effects , Myocarditis/chemically induced , Neoplasms/drug therapy , Nivolumab/adverse effects , Programmed Cell Death 1 Receptor/biosynthesis , Steroids/chemistry
17.
Am J Respir Cell Mol Biol ; 62(6): 767-782, 2020 06.
Article in English | MEDLINE | ID: mdl-32048861

ABSTRACT

Pneumocystis is an unusual, opportunistic fungal pathogen capable of causing Pneumocystis pneumonia (PCP) in immunocompromised hosts. Although PCP was discovered >100 years ago, its pathogenesis remains unclear. The inhibitory receptor PD-1 (programmed death 1), a negative regulator of activated T cells, has been reported to take part in tumor escape, immune tolerance, and infection immunity. In this study, we examined the role of the PD-1/PD-L1 (programmed death-ligand 1) pathway in patients with PCP and in mice. The expression levels of PD-1/PD-L1 in patients with PCP and in mice were measured by real-time PCR and flow cytometry. The effects of PD-1 deficiency are demonstrated using wild-type and PD-1-/- mice. Our data show that Pneumocystis infection promotes PD-1/PD-L1 expression; PD-1 deficiency enhances the phagocytic function of macrophages and the pulmonary T-helper cell type 1 (Th1)/Th17 response, which might contribute to Pneumocystis clearance; and PD-1 deficiency affects the polarization of macrophages. PCP mice treated with anti-PD-1 antibody showed improved pulmonary clearance of Pneumocystis. Collectively, our results demonstrate that the PD-1/PD-L1 pathway plays a role in regulating the innate and adaptive immune responses, suggesting that manipulation of this pathway may constitute an immunotherapeutic strategy for PCP.


Subject(s)
B7-H1 Antigen/physiology , Macrophage Activation/physiology , Pneumonia, Pneumocystis/immunology , Programmed Cell Death 1 Receptor/deficiency , Th1 Cells/immunology , Th17 Cells/immunology , Adaptive Immunity , Adult , Aged , Animals , Antibodies, Fungal/blood , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Female , Humans , Immunity, Innate , Immunocompromised Host , Immunotherapy , Lung/immunology , Lung/metabolism , Lung/microbiology , Macrophages/microbiology , Male , Mice , Mice, Knockout , Middle Aged , Nitric Oxide/metabolism , Opportunistic Infections/immunology , Pneumocystis/immunology , Pneumonia, Pneumocystis/genetics , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction
18.
Clin Immunol ; 214: 108382, 2020 05.
Article in English | MEDLINE | ID: mdl-32169439

ABSTRACT

With the successful treatment of B-cell lymphomas using rituximab, a monoclonal antibody targeting CD20, novel immunotherapies have developed rapidly in recent years. Immune checkpoint blockade and chimeric antigen receptor-T (CAR-T) cell therapy, which are antibody-based therapy and cell-based therapy, respectively, show promising efficacy and have been approved by the Food and Drug Administration for treating hematological malignancies. However, considering severe side effects and short-term clinical remission, the combination of CAR-T cell therapy and programmed cell-death protein-1 (PD-1) blockade has been applied to enhance therapeutic efficacy in preclinical models and clinical trials. Herein, we review the mechanism of the two therapies, show their toxicities and clinical use respectively, address their combined application, and discuss the scope of further investigations of this mechanism-based combination therapy.


Subject(s)
Hematologic Neoplasms/therapy , Immunotherapy, Adoptive , Lymphoma, B-Cell/therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Hematologic Neoplasms/drug therapy , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/drug therapy , Molecular Targeted Therapy/adverse effects , Multicenter Studies as Topic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nivolumab/adverse effects , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Up-Regulation
19.
Cancer Immunol Immunother ; 69(11): 2223-2232, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32474769

ABSTRACT

Malignant cells can increase in number using immune escape mechanisms such as immune checkpoints. In this study, we evaluated the expression of an immune checkpoint programmed death 1 (PD-1) on T-cell subsets in chronic myeloid leukemia (CML). We obtained bone marrow aspirate samples from CML patients and from individuals without evidence of hematologic malignancies (controls). PD-1 expression on T-cell subsets was measured using flow cytometric analysis. PD-1 expression levels on CD8+ T-cells were significantly lower in complete hematologic response (CHR) than in controls, chronic phase, and blast phase (BP). In CML patients receiving imatinib and dasatinib, PD-1 expression levels on CD8+ T-cells were lower than that at diagnosis. PD-1 expression levels on CD8+ T-cells were positively correlated with quantitative levels of the BCR/ABL fusion gene. PD-1 expression levels on CD4+ T-cells were higher in BP than in CHR. PD-1 expression levels on CD4+ T-cells did not differ significantly according to different medications or quantitative BCR/ABL1 fusion gene levels. Low PD-1 expression on CD8+ T-cells might play a role in maintaining CHR in CML patients. Immune monitoring of PD-1 expression on CD8+ T-cells may predict the disease course. In cases of refractory disease or resistance to imatinib or dasatinib, the use of PD-1 inhibitors would be helpful.


Subject(s)
Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Programmed Cell Death 1 Receptor/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Child , Dasatinib/therapeutic use , Female , Humans , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Young Adult
20.
Crit Care Med ; 48(9): 1365-1374, 2020 09.
Article in English | MEDLINE | ID: mdl-32706554

ABSTRACT

OBJECTIVES: Checkpoint inhibitors have been proposed for sepsis following reports of increased checkpoint molecule expression in septic patients. To determine whether clinical studies investigating checkpoint molecule expression provide strong evidence supporting trials of checkpoint inhibitors for sepsis. DATA SOURCES: PubMed, EMBASE, Scopus, Web of Science, inception through October 2019. STUDY SELECTION: Studies comparing checkpoint molecule expression in septic patients versus healthy controls or critically ill nonseptic patients or in sepsis nonsurvivors versus survivors. DATA EXTRACTION: Two investigators extracted data and evaluated study quality. DATA SYNTHESIS: Thirty-six studies were retrieved. Across 26 studies, compared with healthy controls, septic patients had significantly (p ≤ 0.05) increased CD4+ lymphocyte programmed death-1 and monocyte programmed death-ligand-1 expression in most studies. Other checkpoint molecule expressions were variable and studied less frequently. Across 11 studies, compared with critically ill nonseptic, septic patients had significantly increased checkpoint molecule expression in three or fewer studies. Septic patients had higher severity of illness scores, comorbidities, and mortality in three studies providing analysis. Across 12 studies, compared with septic survivors, nonsurvivors had significantly increased expression of any checkpoint molecule on any cell type in five or fewer studies. Of all 36 studies, none adjusted for nonseptic covariates reported to increase checkpoint molecule expression. CONCLUSIONS: Although sepsis may increase some checkpoint molecule expression compared with healthy controls, the data are limited and inconsistent. Further, data from the more informative patient comparisons are potentially confounded by severity of illness. These clinical checkpoint molecule expression studies do not yet provide a strong rationale for trials of checkpoint inhibitor therapy for sepsis.


Subject(s)
Critical Illness , Immune Checkpoint Proteins/biosynthesis , Sepsis/physiopathology , B7-H1 Antigen/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/biosynthesis , Sepsis/drug therapy , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL