Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
3.
Cureus ; 14(7): e27043, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35989841

ABSTRACT

Coronavirus disease 2019 (COVID-19) predominantly targets the respiratory tract; despite gastrointestinal (GI) symptoms that may present in many patients, colonic strictures in coronavirus disease (COVID-19) patients are extremely rare and, to our knowledge, have never been reported. We, herein, present a case of a 59-year-old lady who developed intestinal obstruction due to colonic strictures shortly after recovering from complicated COVID-19 pneumonia. Ultimately, she underwent laparoscopic subtotal colectomy with ileorectal anastomosis. After a long recovery period, she was discharged in good status. It has been more than two years since COVID-19 was declared as a pandemic by the World Health Organization. Infected individuals have highly variable clinical manifestations, yet the pathogenesis, diagnosis and ideal management of each of these complications is not well described in literature.

5.
Clin Exp Ophthalmol ; 49(9): 1018-1026, 2021 12.
Article in English | MEDLINE | ID: mdl-34382734

ABSTRACT

BACKGROUND: To determine the prevalence of zonulopathy in a large cohort of eyes with primary angle closure disease (PACD) that underwent cataract surgery. METHODS: Retrospective consecutive case series of PACD eyes (including primary angle closure suspect, primary angle closure, and primary angle closure glaucoma) that underwent phacoemulsification cataract surgery or clear lens extraction between 2009 and 2020 at a single ophthalmology centre. Those with risk factors for zonulopathy such as history of trauma, pseudoexfoliation syndrome, intraocular surgery, retinitis pigmentosa or connective tissue disorders were excluded. The primary outcomes included the prevalence of zonulopathy assessed intraoperatively and secondary pigment dispersion syndrome. RESULTS: In our cohort of 806 consecutive PACD eyes, the prevalence of zonulopathy was 7.3% (59 of 806 eyes) - significantly greater than the 0.46%-2.6% range reported for the general population (p < 0.001). Intraoperative signs of zonular weakness included floppy capsular bag (29 eyes, 3.6%), zonular laxity (25 eyes, 3.1%) and zonular dehiscence (11 eyes, 1.4%). Among these eyes, capsular tension ring was used in 23 eyes (39.0%), six eyes (10.2%) experienced vitreous prolapse intraoperatively and underwent anterior vitrectomy, and two eyes (3.4%) experienced posterior capsular rupture, one of which required a scleral-fixated intraocular lens. Secondary pigment dispersion syndrome was observed in 141 eyes (17.5%). CONCLUSIONS: This study evidenced a high prevalence of zonulopathy among a large cohort of PACD eyes and suggests zonulopathy as a possible under-recognised cause of angle closure. Until more sophisticated imaging modalities become available, awareness about the prevalence of zonulopathy in angle closure disease coupled with careful preoperative examinations can help minimise or prevent the complications of zonulopathy.


Subject(s)
Cataract , Lenses, Intraocular , Phacoemulsification , Humans , Lens Implantation, Intraocular , Prevalence , Retrospective Studies
6.
Vaccines (Basel) ; 9(4)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916009

ABSTRACT

Colorectal cancer (CRC) is influenced by infiltration of immune cell populations in the tumor microenvironment. While elevated levels of cytotoxic T cells are associated with improved prognosis, limited studies have reported associations between CD4+ T cells and disease outcomes. We recently performed transcriptomic profiling and comparative analyses of sorted CD4+ and CD8+ tumor-infiltrating lymphocytes (TILs) from bulk tumors of CRC patients with varying disease stages. In this study, we compared the transcriptomes of CD4+ with CD8+ TILs. Functional annotation pathway analyses revealed the downregulation of inflammatory response-related genes, while T cell activation and angiogenesis-related genes were upregulated in CD4+ TILs. The top 200 deregulated genes in CD4+ TILs were aligned with the cancer genome atlas (TCGA) CRC dataset to identify a unique gene signature associated with poor prognosis. Moreover, 69 upregulated and 20 downregulated genes showed similar trends of up/downregulation in the TCGA dataset and were used to calculate "poor prognosis score" (ppScore), which was significantly associated with disease-specific survival. High ppScore patients showed lower expression of Treg-, Th1-, and Th17-related genes, and higher expression of Th2-related genes. Our data highlight the significance of T cells within the TME and identify a unique candidate prognostic gene signature for CD4+ TILs in CRC patients.

7.
Cancer Immunol Immunother ; 70(9): 2625-2638, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33582867

ABSTRACT

There is an increased risk of colorectal cancer (CRC) development in patients with non-insulin-dependent type 2 diabetes. CD8+ T cells have been implicated in diabetes and are crucial for anti-tumor immunity. However, transcriptomic profiling for CD8+ T cells from CRC diabetic patients has not been explored. We performed RNA sequencing and compared transcriptomic profiles of CD8+ tumor-infiltrating T lymphocytes (CD8+ TILs) in CRC diabetic patients with CRC nondiabetic patients. We found that genes associated with ribogenesis, epigenetic regulations, oxidative phosphorylation and cell cycle arrest were upregulated in CD8+ TILs from diabetic patients, while genes associated with PI3K signaling pathway, cytokine response and response to lipids were downregulated. Among the significantly deregulated 1009 genes, 342 (186 upregulated and 156 downregulated) genes were selected based on their link to diabetes, and their associations with the presence of specific CRC pathological parameters were assessed using GDC TCGA colon database. The 186 upregulated genes were associated with the presence of colon polyps history (P = 0.0007) and lymphatic invasion (P = 0.0025). Moreover, CRC patients with high expression of the 186 genes were more likely to have poorer disease-specific survival (DSS) (Mantel-Cox log-rank P = 0.024) than those with low score. Our data provide novel insights into molecular pathways and biological functions, which could be altered in CD8+ TILs from CRC diabetic versus nondiabetic patients, and reveal candidate genes linked to diabetes, which could predict DSS and pathological parameters associated with CRC progression. However, further investigations using larger patient cohorts and functional studies are required to validate these findings.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/etiology , Diabetes Mellitus, Type 2/complications , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Transcriptome , Biomarkers , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Computational Biology , Diabetes Mellitus, Type 2/diagnosis , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Prognosis , Protein Interaction Mapping
8.
Vaccines (Basel) ; 9(1)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33477864

ABSTRACT

Immune checkpoint inhibition is an effective anti-cancer therapeutic approach but has shown limited efficacy in treating colorectal cancer (CRC) patients. Importantly, immune constituents of the tumor microenvironment (TME) can influence therapy response and cancer progression. We investigated the expression of immune checkpoints (ICs) on lymphoid populations within the CRC TME and compared with cells from normal colon tissues using samples from 50 patients with varying disease stages. We found that the levels of B cells, T cells, and NK cells were similar, IC-expressing CD4+ and CD4+CD8+ double positive T cells were higher, while CD8+ T cells and CD4-CD8- double negative T cells were significantly lower in CRC tumors. Notably, patients with mismatch-repair deficiency/microsatellite instability-high tumors had higher levels of IC-expressing CD4+ and CD8+ T cells than patients with proficient MMR and microsatellite stable tumors. Lastly, The Cancer Genome Atlas Colon Adenocarcinoma datasets showed associations between low expression of selective genes and poorer progression-free interval. Our findings highlight differential expression of ICs on lymphoid cells in CRC tumors in the era of cancer immunotherapy, which at present is solely approved for anti-PD-1 therapy in patients with dMMR/MSI-H tumors. Further investigations into their functionality have potentials for deciphering resistance mechanisms to IC inhibition.

9.
Am J Ophthalmol ; 224: 310-320, 2021 04.
Article in English | MEDLINE | ID: mdl-33428885

ABSTRACT

PURPOSE: To compare 1-year outcomes of phacoemulsification alone (phaco-only) vs phacoemulsification with implantation of 2 trabecular microbypass stents (iStent or iStent inject; phaco-stent) in eyes with primary angle-closure glaucoma (PACG). DESIGN: Retrospective matched clinical cohort study. METHODS: PACG eyes that underwent phaco-only vs phaco-stent at a single ophthalmology center. Groups were matched for baseline intraocular pressure (IOP) and medication use with a tolerance of ±2 mm Hg and ±1 medication, respectively. Primary outcomes included postoperative change in the mean IOP and medications. One-year outcomes were assessed using generalized estimating equations corrected for baseline intergroup differences. RESULTS: One hundred fifty-eight eyes (79 per group) were included. At 1 year, IOP decreased by 13% (from 16.8 ± 3.1 mm Hg preoperatively) in the phaco-only group (P < .001) and by 27% (from 17.6 ± 3.2 mm Hg) in the phaco-stent group (P < .001). Medication use decreased by 11% (from 1.8 ± 1.3 medications preoperatively) in the phaco-only group (P < .001) and by 46% (from 2.2 ± 1.2 medications) in the phaco-stent group (P < .001). The phaco-stent group experienced significantly larger reductions in IOP and medications compared with the phaco-only group (P < .001). The incidence of IOP spikes was significantly greater in the phaco-only group (18%) compared with the phaco-stent group (4%; P = .005). Safety was favorable with few transient postoperative adverse events. CONCLUSION: The results of this study highlight that phacoemulsification with implantation of 2 trabecular microbypass stents is more effective and possibly more protective than phaco-only in PACG eyes, as evidenced by significantly larger IOP and medication reductions and smaller incidences of IOP spikes among the phaco-stent eyes.


Subject(s)
Glaucoma Drainage Implants , Glaucoma, Angle-Closure/surgery , Phacoemulsification , Stents , Trabecular Meshwork/surgery , Aged , Female , Glaucoma, Angle-Closure/physiopathology , Humans , Intraocular Pressure/physiology , Lens Implantation, Intraocular , Male , Matched-Pair Analysis , Middle Aged , Pseudophakia/physiopathology , Retrospective Studies , Tonometry, Ocular , Visual Acuity/physiology
10.
Cancer Immunol Immunother ; 70(3): 803-815, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33000418

ABSTRACT

Colorectal cancer (CRC) has high mortality rates, especially in patients with advanced disease stages, who often do not respond to therapy. The cellular components of the tumor microenvironment are essentially responsible for dictating disease progression and response to therapy. Expansion of different myeloid cell subsets in CRC tumors has been reported previously. However, tumor-infiltrating myeloid cells have both pro- and anti-tumor roles in disease progression. In this study, we performed transcriptomic profiling of cells of myeloid lineage (CD33+) from bulk CRC tumors at varying disease stages. We identified differentially expressed genes and pathways between CRC patients with advanced stage and early stages. We found that pro-angiogenic and hypoxia-related genes were upregulated, while genes related to immune and inflammatory responses were downregulated in CD33+ myeloid cells from patients with advanced stages, implying that immune cell recruitment and activation could be compromised in advanced disease stages. Moreover, we identified a unique "poor prognosis CD33+ gene signature" by aligning top upregulated and downregulated genes in tumor-infiltrating myeloid cells from our analyses with data from The Cancer Genome Atlas. Our results showed that this gene signature is an independent prognostic indicator for disease-specific survival in CRC patients, potentially reflecting its clinical importance.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Myeloid Cells/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Biomarkers , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Myeloid Cells/pathology , Neoplasm Grading , Neoplasm Staging , Sialic Acid Binding Ig-like Lectin 3/genetics
11.
Front Oncol ; 10: 604906, 2020.
Article in English | MEDLINE | ID: mdl-33312958

ABSTRACT

Elevated levels of myeloid-derived suppressor cells (MDSCs), including polymorphonuclear MDSCs (PMN-MDSCs) and immature MDSCs (I-MDSCs), are usually associated with disease progression in cancer patients, including colorectal cancer (CRC). However, biological mechanisms and molecular pathways regulated by MDSC subpopulations in the CRC tumor microenvironment (TME) have not been fully investigated. In this study, we performed transcriptomic analysis of tumor-infiltrating I-MDSCs and PMN-MDSCs isolated from tumor tissues of six CRC patients, compared to antigen-presenting cells (APCs). We also compared the transcriptomic profiles of tumor-infiltrating PMN-MDSCs to I-MDSCs. Our results showed different molecular pathways regulated by each MDSC subset, potentially reflecting their phenotypical/molecular/functional characteristics in the CRC TME. Moreover, we identified gene signatures in PMN-MDSC and I-MDSC of poor overall survival (OS) and disease-free survival (DFS) using the Cancer Genome Atlas (TCGA) dataset from patients with colon adenocarcinoma (COAD). However, functional studies are required to validate these findings.

12.
Epigenomics ; 12(21): 1871-1882, 2020 11.
Article in English | MEDLINE | ID: mdl-33169618

ABSTRACT

Aim: To elucidate the epigenetic alterations behind the upregulation of immune checkpoints and T cell exhaustion markers in colorectal cancer (CRC) patients. Materials & methods: mRNA expressions of different immune checkpoint/exhaustion markers were analyzed by quantitative real-time reverse transcriptase PCR and epigenetic investigations were performed using bisulfite sequencing and chromatin immunoprecipitation quantitative PCR. Results: mRNA expressions of PD-1, TIM-3, CTLA-4, PD-L1 and TOX2 were significantly upregulated in CD4+ and CD8+ tumor-infiltrating lymphocytes and bulk CRC tumor tissues. Histone 3 lysine 9 trimethylation was downregulated and histone 3 lysine 4 trimethylation was upregulated in PD-L1 and TOX2 promoters in tumor tissues, suggesting that PD-L1 and TOX2 upregulation in CRC tumors could be mediated by activating histone 3 lysine 4 trimethylation. Conclusion: Epigenetic modifications in promoters of immune checkpoint and T cell exhaustion genes could induce their upregulation, and potentially implicate the use of epigenetic modifiers to enhance antitumor immunity in CRC patients.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Immune Checkpoint Proteins/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Histones/metabolism , Humans , Immune Checkpoint Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Promoter Regions, Genetic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
13.
Oncoimmunology ; 9(1): 1825178, 2020 09 30.
Article in English | MEDLINE | ID: mdl-33101776

ABSTRACT

Tumor-infiltrating lymphocytes (TILs) play indispensable roles in the progression and response to treatment of solid tumors. However, the prognostic significance of CD4+ TILs is not fully disclosed in cancers generally and in CRC in particular, mainly due to the existence of different functional subsets of CD4+ T cells. We performed transcriptomic profiling of CD4+ TILs isolated from CRC patients in order to identify differentially expressed genes and their functional pathways in early versus advanced disease stages. We found that in advanced stages, genes related to immune and inflammatory responses, in particular Th1-mediated immune response and cytotoxicity-mediated genes, were downregulated; while epigenetic-mediated silencing genes were upregulated. Interestingly, we identified genes, which were steadily upregulated or downregulated in CD4+ TILs with CRC progression from stage I to IV. Additionally, of the top 200 deregulated genes, 43 upregulated and 64 downregulated genes showed similar deregulation trends in the cancer genome atlas CRC dataset. From these 97 deregulated genes, we identified a "poor prognosis CD4 gene signature (ppCD4sig)". Patients with high ppCD4sig score showed shorter disease-specific survival (DSS) and progression-free interval (PFI). The ppCD4sig was an independent prognostic indicator for DSS (HR = 1.73, 95% CI 1.32-2.27, P = 0.0001) and PFI (HR = 1.75, 95% CI 1.3-2.35, P = 0.0016). Additionally, patients at advanced stages and at a younger age (<55 years) were more likely to have a high ppCD4sig score. Altogether, our data provide novel insights and a unique prognostic gene signature of CD4+ TILs in the CRC microenvironment.


Subject(s)
Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , CD4-Positive T-Lymphocytes , Colorectal Neoplasms/genetics , Gene Expression , Humans , Infant, Newborn , Prognosis , Tumor Microenvironment
14.
J Immunother Cancer ; 8(2)2020 09.
Article in English | MEDLINE | ID: mdl-32948653

ABSTRACT

BACKGROUND: Cytotoxic CD8+ T cell-mediated response is the most important arm of adaptive immunity, which dictates the capacity of the host immune response in eradicating tumor cells. Due to tumor intrinsic and/or extrinsic factors, the density and function of CD8+ tumor-infiltrating lymphocytes (TILs) could be compromised, leading to poor prognosis and survival. METHODS: Using RNA-Seq, transcriptomes of sorted CD3+CD8+ TILs from treatment-naïve colorectal cancer (CRC) patients at advanced stages (III and IV) were compared with those from patients with early stages (I and II). A signature referred to as 'poor prognosis CD8 gene signature (ppCD8sig)' was identified and analyzed in The Cancer Genome Atlas CRC dataset. Scores for the ppCD8sig were calculated and classified as high, intermediate and low, and its prognostic significance was assessed using multivariate analysis and Cox proportional hazard model. Densities of CD3+ and CD8+ T cell infiltration in tumors from patients with high and low ppCD8sig scores were assessed by flow cytometry and immunostaining. RESULTS: Genes related to epigenetic regulation and response to hypoxia were upregulated in CD8+ TILs from patients with advanced stages, while genes related to T cell activation, cell proliferation and cell cycle were downregulated. Patients with high ppCD8sig score had poorer disease-specific survival (DSS) and shorter progression-free interval (PFI). The ppCD8sig was an independent prognostic indicator for DSS (HR 1.83, 95% CI 1.40 to 2.38, p<0.0001) and PFI (HR 1.42, 95% CI 1.04 to 1.93, p=0.026). Additionally, patients with high ppCD8sig score were more likely to have advanced stages (χ2 p<0.0001) and residual disease after primary therapy (χ2 p=0.046). Patients with high ppCD8sig score had reduced levels of CD3+ and CD8+ TILs and low Immunoscores (IS), compared to patients with low ppCD8sig score. CONCLUSIONS: Our data provided insights into the altered regulation of biological mechanisms and signaling pathways in CD8+ TILs during CRC progression, and revealed a gene signature as an independent prognostic indicator. Patients with high ppCD8sig score had lower levels of TILs and low IS. These data further confirm the prognostic value of the identified ppCD8sig and potentially highlight its clinical relevance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/genetics , Gene Expression/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Prognosis , Young Adult
15.
J Immunol Res ; 2020: 9678168, 2020.
Article in English | MEDLINE | ID: mdl-32626789

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of cells that have been implicated in the development of an immunosuppressive environment, which promotes tumorigenesis and tumor progression. Numerous studies have reported expansion of MDSCs in circulation and the tumor microenvironment (TME) of cancer patients. However, due to the heterogenic nature of MDSCs and the different approaches for their identification, their detailed characterization and impact on disease progression in cancer patients are warranted. In this study, we investigated the levels of different myeloid cell subsets and antigen-presenting cells (APCs) using flow cytometry in unfractionated whole blood (WB), peripheral blood mononuclear cells (PBMCs), tumor tissue (TT), and adjacent normal tissue (NT) of colorectal cancer (CRC) patients. We found high levels of granulocytic myeloid cells (GMCs) in whole blood, but their levels were significantly lower in PBMCs. Importantly, we found significantly higher levels of GMCs in the TME compared to NT. In addition, monocytic myeloid cells (MMCs) showed significantly higher levels in PBMCs of CRC patients, compared to healthy donors (HDs). Notably, patients with advanced disease stages showed significantly higher levels of GMCs compared to early stages in whole blood, but PBMCs and tumor-infiltrating myeloid cells did not show any significant differences. Lastly, we found that levels of GMCs decreased, while IMCs increased in the TME with tumor budding. Our results highlight the importance of investigating the levels of different myeloid cell subsets in PBMCs versus whole blood of cancer patients and improve current knowledge on the potential prognostic significance of myeloid cells in CRC patients.


Subject(s)
Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Leukocyte Count , Myeloid Cells , Tumor Microenvironment , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Colorectal Neoplasms/therapy , Female , Humans , Immunophenotyping , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Young Adult
16.
Cancer Immunol Immunother ; 69(10): 1989-1999, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32393998

ABSTRACT

Despite recent advances in colorectal cancer (CRC) treatment, a large proportion of patients show limited responses to therapies, especially in advanced stages. There is an urgent need to identify prognostic biomarkers and/or therapeutic targets in advanced stages, aiming to improve the efficacy of current treatments. We aimed to determine prognostic biomarkers in tumor tissue and circulation of CRC patients, with a special focus on T cell exhaustion markers. We found that mRNA levels of PD-1, TIM-3, CTLA-4, TIGIT, CD160, CD244, KLRG1, TOX2, TOX3, Ki-67, and PRDM1 were elevated in CRC tumor tissues. We also investigated differences in gene expression between early and advanced disease stages. We found that TOX and potentially TIM-3, CTLA-4, VISTA, TIGIT, KLRG1, TOX2, SIRT1, Ki-67, and Helios mRNA levels in tumor tissue were elevated in advanced disease stages, suggesting their potential roles in CRC progression. In contrast, PD-1 and CD160 levels in tumor tissue were downregulated in advanced stages. In the circulation of CRC patients, mRNA levels of PD-1, VISTA and LAG-3 were higher than those of healthy individuals. Moreover, in circulation, PD-1, CTLA-4 and TIGIT mRNA levels were reduced in advanced stages. Interestingly, levels of PD-1 in both tumor tissue and circulation were reduced in advanced stages, suggesting that targeting PD-1 in patients with advanced stages could be less effective. Altogether, these findings suggest some potential T cell exhaustion markers that could be utilized as prognostic biomarkers and/or therapeutic targets for CRC. However, further investigations and validations in larger cohorts are required to confirm these findings.


Subject(s)
Biomarkers, Tumor/blood , CTLA-4 Antigen/blood , Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/blood , Receptors, Immunologic/blood , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , CTLA-4 Antigen/genetics , Case-Control Studies , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging , Programmed Cell Death 1 Receptor/genetics , Receptors, Immunologic/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Young Adult
17.
Clin Immunol ; 215: 108429, 2020 06.
Article in English | MEDLINE | ID: mdl-32320745

ABSTRACT

T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) is an inhibitory immune checkpoint, which suppresses anti-tumor immune responses. TIM-3 expression on different immune cells in periphery and tumor microenvironment (TME) of colorectal cancer (CRC) patients has not been fully investigated. We found that TIM-3 was mainly expressed on monocytic myeloid cells (MMCs) and antigen-presenting cells (APCs) in circulation but was mainly expressed on T cells and APCs in the TME. Additionally, TIM-3- T cells co-expressed higher levels of PD-1 than TIM-3+ T cells in normal tissue. In contrast, TIM-3+ T cells in the TME showed significantly higher PD-1 expression. Interestingly, there was a trend towards increased levels of TIM-3+ APCs with disease stages; however, levels of TIM-3+ T cells were decreased with disease stages in the TME. This study shows the differential expression of TIM-3 on different immune cells in circulation and TME of CRC patients, and their associations with disease stages.


Subject(s)
Colorectal Neoplasms/blood , Colorectal Neoplasms/metabolism , Hepatitis A Virus Cellular Receptor 2/blood , Tumor Microenvironment/physiology , Antigen-Presenting Cells/metabolism , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Cells, Cultured , Humans , Monocytes/metabolism , Myeloid Cells/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/metabolism
18.
Vaccines (Basel) ; 8(1)2020 02 06.
Article in English | MEDLINE | ID: mdl-32041340

ABSTRACT

T cell immunoglobulin mucin-3 (TIM-3) is an immune checkpoint identified as one of the key players in regulating T-cell responses. Studies have shown that TIM-3 is upregulated in the tumor microenvironment (TME). However, the precise role of TIM-3 in colorectal cancer (CRC) TME is yet to be elucidated. We performed phenotypic and molecular characterization of TIM-3+ T cells in the TME and circulation of CRC patients by analyzing tumor tissues (TT, TILs), normal tissues (NT, NILs), and peripheral blood mononuclear cells (PBMC). TIM-3 was upregulated on both CD4+ and CD3+CD4- (CD8+) TILs. CD4+TIM-3+ TILs expressed higher levels of T regulatory cell (Tregs)-signature genes, including FoxP3 and Helios, compared with their TIM-3- counterparts. Transcriptomic and ingenuity pathway analyses showed that TIM-3 potentially activates inflammatory and tumor metastatic pathways. Moreover, NF-κB-mediated transcription factors were upregulated in CD4+TIM-3+ TILs, which could favor proliferation/invasion and induce inflammatory and T-cell exhaustion pathways. In addition, we found that CD4+TIM-3+ TILs potentially support tumor invasion and metastasis, compared with conventional CD4+CD25+ Tregs in the CRC TME. However, functional studies are warranted to support these findings. In conclusion, this study discloses some of the functional pathways of TIM-3+ TILs, which could improve their targeting in more specific therapeutic approaches in CRC patients.

19.
Clin Epigenetics ; 12(1): 13, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31941522

ABSTRACT

BACKGROUND: Increased numbers of myeloid-derived suppressor cells (MDSCs) are positively correlated with poor prognosis and reduced survivals of cancer patients. They play central roles in tumor immune evasion and tumor metastasis. However, limited data are available on phenotypic/transcriptomic characteristics of the different MDSCs subsets in cancer. These cells include immature (I-MDSCs), monocytic (M-MDSCs), and polymorphonuclear/granulocytic (PMN-MDSCs). METHODS: Phenotypic characterization of myeloid subsets from 27 colorectal cancer (CRC) patients was assessed by flow cytometric analyses. RNA-sequencing of sorted I-MDSCs, PMN-MDSCs, and antigen-presenting cells (APCs) was also performed. RESULTS: We found that the levels of I-MDSCs and PMN-MDSCs were increased in tumor tissues (TT), compared with normal tissues (NT) in colorectal cancer. Our functional annotation analyses showed that genes associated with histone deacetylase (HDAC) activation- and DNA methylation-mediated transcriptional silencing were upregulated, and histone acetyl transferase (HAT)-related genes were downregulated in tumor-infiltrating I-MDSCs. Moreover, pathways implicated in cell trafficking and immune suppression, including Wnt, interleukin-6 (IL-6), and mitogen-activated protein kinase (MAPK) signaling, were upregulated in I-MDSCs. Notably, PMN-MDSCs showed downregulation in genes related to DNA methylation and HDAC binding. Using an ex vivo model, we found that inhibition of HDAC activation or neutralization of IL-6 in CRC tumor tissues downregulates the expression of genes associated with immunosuppression and myeloid cell chemotaxis, confirming the importance of HDAC activation and IL-6 signaling pathway in MDSC function and chemotaxis. CONCLUSIONS: This study provides novel insights into the epigenetic regulations and other molecular pathways in different myeloid cell subsets within the CRC tumor microenvironment (TME), giving opportunities to potential targets for therapeutic benefits.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histone Code , Myeloid-Derived Suppressor Cells/metabolism , Adult , Aged , Aged, 80 and over , Cell Movement/genetics , Chemotaxis/genetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Gene Expression Profiling , Histone Deacetylases/metabolism , Humans , Immune Tolerance/genetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/physiology , Male , Middle Aged , Myeloid-Derived Suppressor Cells/enzymology
20.
Front Immunol ; 10: 2936, 2019.
Article in English | MEDLINE | ID: mdl-31921188

ABSTRACT

Blockade of inhibitory immune checkpoints (ICs) is a promising therapeutic approach; however, it has shown limited success in some cancers including colorectal cancer (CRC). The tumor microenvironment (TME) is largely responsible for response to therapy, and its constituents may provide robust biomarkers for successful immunotherapeutic approaches. In this study, we performed phenotypical characterization and critical analyses of key inhibitory ICs and T regulatory cell (Treg)-related markers on CD4+ T cell subsets in CRC patients, and compared with normal colon tissues and peripheral blood from the same patients. We also investigated correlations between the levels of different CD4+ T cell subsets and the clinicopathologic features including disease stage and tumor budding. We found a significant increase in the levels of CD4+FoxP3+Helios+ T cells, which represent potentially highly immunosuppressive Tregs, in the CRC TME. Additionally, tumor-infiltrating CD4+ T cells upregulated programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), T cell immunoglobulin and mucin domain-3 (TIM-3) and lymphocyte-activation gene 3 (LAG-3). We also characterized the expression of PD-1, CTLA-4, TIM-3, and LAG-3 on different CD4+FoxP3-/+Helios-/+ T cell subsets. Interestingly, we found that CTLA-4, TIM-3, and LAG-3 were mainly co-expressed on FoxP3+Helios+ Tregs in the TME. Additionally, FoxP3high Tregs expressed higher levels of Helios, CTLA-4 and TIM-3 than FoxP3low T cells. These results highlight the significance of Tregs in the CRC TME and suggest that Tregs may hamper response to IC blockade in CRC patients, but effects of different IC inhibition regimes on Treg levels or activity warrants further investigations. We also found that CD4+CTLA-4+ T cells in circulation are increased in patients with advanced disease stage. This study simultaneously provides important insights into the differential levels of CD4+ T cell subpopulations and IC expression in CRC TME, compared to periphery and associations with clinicopathologic features, which could be used as potential biomarkers for CRC progression and response to therapy.


Subject(s)
Biomarkers, Tumor/immunology , CD4-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/pathology , Colorectal Neoplasms/pathology , Female , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging
SELECTION OF CITATIONS
SEARCH DETAIL
...