Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.495
Filter
1.
Front Immunol ; 15: 1393799, 2024.
Article in English | MEDLINE | ID: mdl-38975347

ABSTRACT

SOCS are a family of negative inhibitors of the molecular cascades induced by cytokines, growth factors and hormones. At molecular level, SOCS proteins inhibit the kinase activity of specific sets of receptor-associated Janus Activated Kinases (JAKs), thereby suppressing the propagation of intracellular signals. Of the eight known members, SOCS1 and SOCS3 inhibit activity of JAKs mainly induced by cytokines and can play key roles in regulation of inflammatory and immune responses. SOCS1 and SOCS3 are the most well-characterized SOCS members in skin inflammatory diseases, where their inhibitory activity on cytokine activated JAKs and consequent anti-inflammatory action has been widely investigated in epidermal keratinocytes. Structurally, SOCS1 and SOCS3 share the presence of a N-terminal domain containing a kinase inhibitory region (KIR) motif able to act as a pseudo-substrate for JAK and to inhibit its activity. During the last decades, the design and employment of SOCS1 and SOCS3-derived peptides mimicking KIR domains in experimental models of dermatoses definitively established a strong anti-inflammatory and ameliorative impact of JAK inhibition on skin inflammatory responses. Herein, we discuss the importance of the findings collected in the past on SOCS1 and SOCS3 function in the inflammatory responses associated to skin immune-mediated diseases and malignancies, for the development of the JAK inhibitor drugs. Among them, different JAK inhibitors have been introduced in the clinical practice for treatment of atopic dermatitis and psoriasis, and others are being investigated for skin diseases like alopecia areata and vitiligo.


Subject(s)
Cell Transformation, Neoplastic , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Humans , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Suppressor of Cytokine Signaling 3 Protein/metabolism , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Signal Transduction , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Dermatitis/immunology , Dermatitis/metabolism , Janus Kinases/metabolism , Skin/immunology , Skin/pathology , Skin/metabolism
2.
Front Immunol ; 15: 1376660, 2024.
Article in English | MEDLINE | ID: mdl-38903501

ABSTRACT

Although the chronic lymphocytic leukemia (CLL) treatment landscape has changed dramatically, unmet clinical needs are emerging, as CLL in many patients does not respond, becomes resistant to treatment, relapses during treatment, or transforms into Richter. In the majority of cases, transformation evolves the original leukemia clone into a diffuse large B-cell lymphoma (DLBCL). Richter transformation (RT) represents a dreadful clinical challenge with limited therapeutic opportunities and scarce preclinical tools. CLL cells are well known to highly depend on survival signals provided by the tumor microenvironment (TME). These signals enhance the frequency of immunosuppressive cells with protumor function, including regulatory CD4+ T cells and tumor-associated macrophages. T cells, on the other hand, exhibit features of exhaustion and profound functional defects. Overall immune dysfunction and immunosuppression are common features of patients with CLL. The interaction between malignant cells and TME cells can occur during different phases of CLL development and transformation. A better understanding of in vivo CLL and RT biology and the availability of adequate mouse models that faithfully recapitulate the progression of CLL and RT within their microenvironments are "conditio sine qua non" to develop successful therapeutic strategies. In this review, we describe the xenograft and genetic-engineered mouse models of CLL and RT, how they helped to elucidate the pathophysiology of the disease progression and transformation, and how they have been and might be instrumental in developing innovative therapeutic approaches to finally eradicate these malignancies.


Subject(s)
Cell Transformation, Neoplastic , Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell , Tumor Microenvironment , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Animals , Tumor Microenvironment/immunology , Humans , Mice , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/pathology
3.
Blood Cancer Discov ; 5(4): 229-233, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38713827

ABSTRACT

In this commentary, we discuss the investigation into reports of T-cell malignancies following chimeric antigen receptor T-cell therapy. We argue that although these cases should be thoroughly examined, current data suggest that such risks with autologous chimeric antigen receptor T cells are remarkably low compared with other cancer treatments. We also emphasize the importance of continued research, transparent reporting, and participation in postauthorization safety studies.


Subject(s)
Cell Transformation, Neoplastic , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/genetics , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Neoplasms/immunology , Neoplasms/therapy
4.
Mol Biol Rep ; 51(1): 571, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662155

ABSTRACT

Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.


Subject(s)
Cell Transformation, Neoplastic , Galectins , Hepatitis A Virus Cellular Receptor 2 , Leukemia , Humans , Galectins/metabolism , Leukemia/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/genetics , Animals , Immune Tolerance , Signal Transduction , Tumor Escape , Cell Proliferation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism
5.
J Leukoc Biol ; 116(1): 18-32, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38243586

ABSTRACT

B cell acute lymphoblastic leukemia (B-ALL) arises from genetic alterations impacting B cell progenitors, ultimately leading to clinically overt disease. Extensive collaborative efforts in basic and clinical research have significantly improved patient prognoses. Nevertheless, a subset of patients demonstrate resistance to conventional chemotherapeutic approaches and emerging immunotherapeutic interventions. This review highlights the mechanistic underpinnings governing B-ALL transformation. Beginning with exploring normative B cell lymphopoiesis, we delineate the influence of recurrent germline and somatic genetic aberrations on the perturbation of B cell progenitor differentiation and protumorigenic signaling, thereby facilitating the neoplastic transformation underlying B-ALL progression. Additionally, we highlight recent advances in the multifaceted landscape of B-ALL, encompassing metabolic reprogramming, microbiome influences, inflammation, and the discernible impact of socioeconomic and racial disparities on B-ALL transformation and patient survival.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Animals , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/genetics , Lymphopoiesis
6.
Nucleic Acids Res ; 51(10): 5193-5209, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37070602

ABSTRACT

The long non-coding RNA EPR is expressed in epithelial tissues, binds to chromatin and controls distinct biological activities in mouse mammary gland cells. Because of its high expression in the intestine, in this study we have generated a colon-specific conditional targeted deletion (EPR cKO) to evaluate EPR in vivo functions in mice. EPR cKO mice display epithelium hyperproliferation, impaired mucus production and secretion, as well as inflammatory infiltration in the proximal portion of the large intestine. RNA sequencing analysis reveals a rearrangement of the colon crypt transcriptome with strong reduction of goblet cell-specific factors including those involved in the synthesis, assembly, transport and control of mucus proteins. Further, colon mucosa integrity and permeability are impaired in EPR cKO mice, and this results in higher susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumor formation. Human EPR is down-regulated in human cancer cell lines as well as in human cancers, and overexpression of EPR in a colon cancer cell line results in enhanced expression of pro-apoptotic genes. Mechanistically, we show that EPR directly interacts with select genes involved in mucus metabolism whose expression is reduced in EPR cKO mice and that EPR deletion causes tridimensional chromatin organization changes.


Subject(s)
Cell Transformation, Neoplastic , Inflammation , Mucus , RNA, Long Noncoding , Animals , Humans , Mice , Cell Transformation, Neoplastic/immunology , Colon/metabolism , Disease Models, Animal , Inflammation/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
7.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35121655

ABSTRACT

The tumor microenvironment (TME) provides potential targets for cancer therapy. However, how signals originating in cancer cells affect tumor-directed immunity is largely unknown. Deletions in the CHUK locus, coding for IκB kinase α (IKKα), correlate with reduced lung adenocarcinoma (ADC) patient survival and promote KrasG12D-initiated ADC development in mice, but it is unknown how reduced IKKα expression affects the TME. Here, we report that low IKKα expression in human and mouse lung ADC cells correlates with increased monocyte-derived macrophage and regulatory T cell (Treg) scores and elevated transcription of genes coding for macrophage-recruiting and Treg-inducing cytokines (CSF1, CCL22, TNF, and IL-23A). By stimulating recruitment of monocyte-derived macrophages from the bone marrow and enforcing a TNF/TNFR2/c-Rel signaling cascade that stimulates Treg generation, these cytokines promote lung ADC progression. Depletion of TNFR2, c-Rel, or TNF in CD4+ T cells or monocyte-derived macrophages dampens Treg generation and lung tumorigenesis. Treg depletion also attenuates carcinogenesis. In conclusion, reduced cancer cell IKKα activity enhances formation of a protumorigenic TME through a pathway whose constituents may serve as therapeutic targets for KRAS-initiated lung ADC.


Subject(s)
Adenocarcinoma of Lung/immunology , Cytokines/immunology , I-kappa B Kinase/immunology , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Humans , Immunosuppression Therapy/methods , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Signal Transduction/immunology
8.
Mol Cancer ; 21(1): 27, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35062950

ABSTRACT

Cellular metabolism constitutes a fundamental process in biology. During tumor initiation and progression, each cellular component in the cancerous niche undergoes dramatic metabolic reprogramming, adapting to a challenging microenvironment of hypoxia, nutrient deprivation, and other stresses. While the metabolic hallmarks of cancer have been extensively studied, the metabolic states of the immune cells are less well elucidated. Here we review the metabolic disturbance and fitness of the immune system in the tumor microenvironment (TME), focusing on the impact of oncometabolites to the function of immune cells and the clinical significance of targeting metabolism in anti-tumor immunotherapy. Metabolic alterations in the immune system of TME offer novel therapeutic insight into cancer treatment.


Subject(s)
Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Energy Metabolism , Neoplasms/etiology , Neoplasms/metabolism , Tumor Microenvironment/immunology , Adaptation, Biological , Animals , Cell Transformation, Neoplastic/genetics , Cellular Reprogramming , Combined Modality Therapy , Disease Management , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Immunomodulation , Immunotherapy , Neoplasms/diagnosis , Neoplasms/therapy , Treatment Outcome
9.
Eur J Surg Oncol ; 48(1): 245-252, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34620510

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most prevalent and life-threatening cancer among the world. Accumulated somatic mutations during malignant transformation process endow cancer cells with increased growth, invasiveness and immunogenicity. These highly immunogenic cancer cells develop multiple strategies to evade immune attack. Through post-transcriptional regulation, microRNAs (miRNAs) not only participate in cancer development and progression but also manipulate anti-cancer immune response. This study aims to identify miRNAs associated with the colorectal cell malignant transformation process and their association with immune cell population using synchronous adjacent normal, polyp and CRC specimens. METHODS: We conducted a Low Density Array to compare the miRNA expression profile of synchronous colorectal adenoma, adenocarcinoma and adjacent normal colon mucosa collected from 8 patients, in order to identify candidate miRNAs involved in CRC progression. These findings were further validated in 14 additional patients and GEO dataset GSE41655. The relative abundance of dendritic cells, natural killer cells, neutrophil and macrophage was determined and correlated with dysregulated miRNA levels. RESULTS: MicroRNA microarray identified 39 miRNAs aberrantly expressed during the colorectal cell transformation process. Seven novel miRNAs were shortlisted, and dysregulation of miR-149-3p, miR-192-3p, miR-335-5p and miR-425 were further validated by the qPCR validation experiment and data retrieved from the GEO dataset. Furthermore, these miRNAs demonstrated certain associations with level of dendritic cells, natural killer cells, neutrophil and macrophage within the polyp or CRC specimens. CONCLUSION: This study revealed miRNA dysregulated during stepwise malignant transformation of colorectal mucosal cells and their association with immune cell population.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Cell Transformation, Neoplastic/genetics , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , MicroRNAs/genetics , Tumor Escape/genetics , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenoma/immunology , Adenoma/metabolism , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/immunology , Colon/immunology , Colon/metabolism , Colonic Polyps/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Dendritic Cells/immunology , Female , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Male , MicroRNAs/metabolism , Middle Aged , Neutrophils/immunology , Tumor Escape/immunology , Tumor-Associated Macrophages/immunology
10.
Physiol Rev ; 102(1): 455-510, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34541899

ABSTRACT

Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. Cancer is a multistep disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signaling in cancer in every step of disease progression. Rho GTPases contribute to tumor initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence, and cancer cell stemness. Rho GTPases play a major role in cell migration and in the metastatic process. They are also involved in interactions with the tumor microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/drug therapy , Tumor Microenvironment/physiology , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement/physiology , Cell Transformation, Neoplastic/immunology , Humans , Signal Transduction/genetics
11.
Virology ; 566: 136-142, 2022 01.
Article in English | MEDLINE | ID: mdl-34922257

ABSTRACT

High mobility group box 1 (HMGB1) is an important chromatin protein and a pro-inflammatory molecule. Though shown to enhance target DNA binding by the Epstein-Barr virus (EBV) lytic switch protein ZEBRA, whether HMGB1 actually contributes to gammaherpesvirus biology is not known. In investigating the contribution of HMGB1 to the lytic phase of EBV, important for development of EBV-mediated diseases, we find that compared to latently-infected cells, lytic phase Burkitt lymphoma-derived cells and peripheral blood lytic cells during primary EBV infection express high levels of HMGB1. Our experiments place HMGB1 upstream of ZEBRA and reveal that HMGB1, through the NLRP3 inflammasome, sustains the expression of ZEBRA. These findings indicate that in addition to the NLRP3 inflammasome's recently discovered role in turning the EBV lytic switch on, NLRP3 cooperates with the danger molecule HMGB1 to also maintain ZEBRA expression, thereby sustaining the lytic signal.


Subject(s)
Burkitt Lymphoma/genetics , Epstein-Barr Virus Infections/genetics , HMGB1 Protein/genetics , Herpesvirus 4, Human/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Trans-Activators/genetics , B-Lymphocytes/immunology , B-Lymphocytes/virology , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , HMGB1 Protein/immunology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/genetics , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Primary Cell Culture , Signal Transduction , Trans-Activators/immunology , Virus Activation/genetics , Virus Activation/immunology , Virus Latency/genetics , Virus Latency/immunology
12.
Front Endocrinol (Lausanne) ; 12: 785050, 2021.
Article in English | MEDLINE | ID: mdl-34925244

ABSTRACT

Macrophages are one of the most common infiltrating immune cells and an essential component of tumor microenvironment. Macrophages and the soluble cytokines and chemokines produced play an important role in tumorigenesis, progression, invasion and metastasis in solid tumors. Despite the multiple studies in other solid tumors, there is little known about macrophages in pituitary adenomas. Recently, studies about pituitary adenoma-infiltrated macrophages have been emerging, including the immunohistochemical and immunophenotypic analysis of the pituitary adenomas and further studies into the mechanism of the crosstalk between macrophages and tumor cells in vivo and in vitro. These studies have offered us new insights into the polarization of macrophages and its role in tumorigenesis, progression and invasion of pituitary adenomas. This review describes the advances in the field of pituitary adenoma-infiltrated macrophages and the prospect of targeting macrophages as cancer therapy in pituitary adenoma.


Subject(s)
Adenoma/metabolism , Cell Transformation, Neoplastic/metabolism , Pituitary Neoplasms/metabolism , Tumor Microenvironment/physiology , Tumor-Associated Macrophages/metabolism , Adenoma/immunology , Adenoma/pathology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Humans , Pituitary Neoplasms/immunology , Pituitary Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
13.
Cell Rep ; 37(10): 110099, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879282

ABSTRACT

Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/immunology , Epithelial Cells/immunology , Lymphocyte Activation , Mammary Glands, Animal/immunology , Mammary Neoplasms, Experimental/immunology , Natural Killer T-Cells/immunology , Parity , Animals , Antigens, CD1d/metabolism , Cell Communication , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Genes, myc , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Natural Killer T-Cells/metabolism , Pregnancy , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , Tumor Microenvironment
14.
PLoS One ; 16(12): e0261082, 2021.
Article in English | MEDLINE | ID: mdl-34910746

ABSTRACT

INTRODUCTION: Multiple previous studies have shown the monoclonal antibody Das-1 (formerly called 7E12H12) is specifically reactive towards metaplastic and carcinomatous lesions in multiple organs of the gastrointestinal system (e.g. Barrett's esophagus, intestinal-type metaplasia of the stomach, gastric adenocarcinoma, high-grade pancreatic intraepithelial neoplasm, and pancreatic ductal adenocarcinoma) as well as in other organs (bladder and lung carcinomas). Beyond being a useful biomarker in tissue, mAb Das-1 has recently proven to be more accurate than current paradigms for identifying cysts harboring advanced neoplasia. Though this antibody has been used extensively for clinical, basic science, and translational applications for decades, its epitope has remained elusive. METHODS: In this study, we chemically deglycosylated a standard source of antigen, which resulted in near complete loss of the signal as measured by western blot analysis. The epitope recognized by mAb Das-1 was determined by affinity to a comprehensive glycan array and validated by inhibition of a direct ELISA. RESULTS: The epitope recognized by mAb Das-1 is 3'-Sulfo-Lewis A/C (3'-Sulfo-LeA/C). 3'-Sulfo-LeA/C is broadly reexpressed across numerous GI epithelia and elsewhere during metaplastic and carcinomatous transformation. DISCUSSION: 3'-Sulfo-LeA/C is a clinically important antigen that can be detected both intracellularly in tissue using immunohistochemistry and extracellularly in cyst fluid and serum by ELISA. The results open new avenues for tumorigenic risk stratification of various gastrointestinal lesions.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Transformation, Neoplastic/immunology , Epitopes, B-Lymphocyte/immunology , Gastrointestinal Neoplasms/immunology , Intestinal Mucosa/immunology , Lewis Blood Group Antigens/immunology , Oligosaccharides/immunology , Antibody Specificity , Biomarkers, Tumor/immunology , Cell Line, Tumor , Humans , Immunohistochemistry
15.
Front Immunol ; 12: 719954, 2021.
Article in English | MEDLINE | ID: mdl-34721383

ABSTRACT

The constant exposure of the liver to gut derived foreign antigens has resulted in this organ attaining unique immunological characteristics, however it remains susceptible to immune mediated injury. Our understanding of this type of injury, in both the native and transplanted liver, has improved significantly in recent decades. This includes a greater awareness of the tolerance inducing CD4+ CD25+ CD127low T-cell lineage with the transcription factor FoxP3, known as regulatory T-Cells (Tregs). These cells comprise 5-10% of CD4+ T cells and are known to function as an immunological "braking" mechanism, thereby preventing immune mediated tissue damage. Therapies that aim to increase Treg frequency and function have proved beneficial in the setting of both autoimmune diseases and solid organ transplantations. The safety and efficacy of Treg therapy in liver disease is an area of intense research at present and has huge potential. Due to these cells possessing significant plasticity, and the potential for conversion towards a T-helper 1 (Th1) and 17 (Th17) subsets in the hepatic microenvironment, it is pre-requisite to modify the microenvironment to a Treg favourable atmosphere to maintain these cells' function. In addition, implementation of therapies that effectively increase Treg functional activity in the liver may result in the suppression of immune responses and will hinder those that destroy tumour cells. Thus, fine adjustment is crucial to achieve this immunological balance. This review will describe the hepatic microenvironment with relevance to Treg function, and the role these cells have in both native diseased and transplanted livers.


Subject(s)
Immunotherapy/methods , Liver Diseases/therapy , Liver Transplantation , T-Lymphocytes, Regulatory/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cellular Microenvironment/immunology , Chronic Disease , Disease Management , Disease Susceptibility/etiology , Disease Susceptibility/metabolism , Humans , Immunomodulation , Immunotherapy/adverse effects , Liver Diseases/diagnosis , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Transplantation/adverse effects , Liver Transplantation/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome
16.
Front Immunol ; 12: 687874, 2021.
Article in English | MEDLINE | ID: mdl-34675913

ABSTRACT

Soluble tumor necrosis factor-α (sTNF-α) plays an important role in colitis-associated cancer (CAC); however, little is known about transmembrane TNF-α (tmTNF-α). Here, we observed an increase in sTNF-α mainly in colitis tissues from an azoxymethane/dextran sodium sulfate (DSS)-induced CAC mouse model whereas tmTNF-α levels were chiefly increased on epithelial cells at the tumor stage. The ratio of intracolonic tmTNF-α/sTNF-α was negatively correlated with the levels of pro-inflammatory mediators (IL-1ß, IL-6, and NO) and M1 macrophages but positively correlated with the infiltration of myeloid-derived suppressor cells, regulatory T cells, and the level of the anti-inflammatory cytokine IL-10, suggesting an anti-inflammatory effect of tmTNF-α. This effect of tmTNF-α was confirmed again by the induction of resistance to LPS in colonic epithelial cell lines NCM460 and HCoEpiC through the addition of exogenous tmTNF-α or transfection of the tmTNF-α leading sequence that lacks the extracellular segment but retains the intracellular domain of tmTNF-α. A tmTNF-α antibody was used to block tmTNF-α shedding after the first or second round of inflammation induction by DSS drinking to shift the time window of tmTNF-α expression ahead to the inflammation stage. Antibody treatment significantly alleviated inflammation and suppressed subsequent adenoma formation, accompanied by increased apoptosis. An antitumor effect was also observed when the antibody was administered at the malignant phase of CAC. Our results reveal tmTNF-α as a novel molecular marker for malignant transformation in CAC and provide a new insight into blocking the pathological process by targeting tmTNF-α processing.


Subject(s)
Adenoma/prevention & control , Anti-Inflammatory Agents/pharmacology , Antibodies/pharmacology , Anticarcinogenic Agents/pharmacology , Cell Membrane/drug effects , Colitis-Associated Neoplasms/prevention & control , Colon/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adenoma/immunology , Adenoma/metabolism , Adenoma/pathology , Animals , Apoptosis/drug effects , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis-Associated Neoplasms/immunology , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
17.
Cell Rep ; 37(3): 109858, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686350

ABSTRACT

Cell death provides host defense and maintains homeostasis. Zα-containing molecules are essential for these processes. Z-DNA binding protein 1 (ZBP1) activates inflammatory cell death, PANoptosis, whereas adenosine deaminase acting on RNA 1 (ADAR1) serves as an RNA editor to maintain homeostasis. Here, we identify and characterize ADAR1's interaction with ZBP1, defining its role in cell death regulation and tumorigenesis. Combining interferons (IFNs) and nuclear export inhibitors (NEIs) activates ZBP1-dependent PANoptosis. ADAR1 suppresses this PANoptosis by interacting with the Zα2 domain of ZBP1 to limit ZBP1 and RIPK3 interactions. Adar1fl/flLysMcre mice are resistant to development of colorectal cancer and melanoma, but deletion of the ZBP1 Zα2 domain restores tumorigenesis in these mice. In addition, treating wild-type mice with IFN-γ and the NEI KPT-330 regresses melanoma in a ZBP1-dependent manner. Our findings suggest that ADAR1 suppresses ZBP1-mediated PANoptosis, promoting tumorigenesis. Defining the functions of ADAR1 and ZBP1 in cell death is fundamental to informing therapeutic strategies for cancer and other diseases.


Subject(s)
Adenosine Deaminase/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/enzymology , Melanoma, Experimental/enzymology , RNA-Binding Proteins/metabolism , Skin Neoplasms/enzymology , Adenosine Deaminase/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hydrazines/pharmacology , Interferon-gamma/pharmacology , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Necroptosis , Pyroptosis , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Triazoles/pharmacology
18.
Biomed Pharmacother ; 144: 112277, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34624674

ABSTRACT

Increasing evidence suggested that cholesterol is an important integrant of cell membranes, that plays a key role in tumor progression, immune dysregulation, and pathological changes in epigenetic mechanisms. Based on these theories, there is a growing interest on targeting cholesterol in the treatment of cancer. Here, we comprehensively reviewed the major function of cholesterol on oncogenicity, the therapeutic targets of cholesterol and its metabolites in cancer, and provide detailed insight into the essential roles of cholesterol in mediating immune and epigenetic mechanisms of the tumor microenvironment. It is also worth mentioning that the gut microbiome is an indispensable component of cancer mediation because of its role in cholesterol metabolism. Finally, we summarized recent studies on the potential targets of cholesterol and their metabolism, to provide more therapeutic interventions in oncology.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cholesterol/metabolism , Neoplasms/metabolism , Animals , Bacteria/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Disease Progression , Epigenesis, Genetic , Gastrointestinal Microbiome , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/immunology , Tumor Microenvironment/immunology
19.
Front Immunol ; 12: 680955, 2021.
Article in English | MEDLINE | ID: mdl-34566954

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a malignant tumor of the nasopharynx mainly characterized by geographic distribution and EBV infection. Metabolic reprogramming, one of the cancer hallmarks, has been frequently reported in NPCs to adapt to internal energy demands and external environmental pressures. Inevitably, the metabolic reprogramming within the tumor cell will lead to a decreased pH value and diverse nutritional supplements in the tumor-infiltrating micro-environment incorporating immune cells, fibroblasts, and endothelial cells. Accumulated evidence indicates that metabolic reprogramming derived from NPC cells may facilitate cancer progression and immunosuppression by cell-cell communications with their surrounding immune cells. This review presents the dysregulated metabolism processes, including glucose, fatty acid, amino acid, nucleotide metabolism, and their mutual interactions in NPC. Moreover, the potential connections between reprogrammed metabolism, tumor immunity, and associated therapy would be discussed in this review. Accordingly, the development of targets on the interactions between metabolic reprogramming and immune cells may provide assistances to overcome the current treatment resistance in NPC patients.


Subject(s)
Disease Susceptibility , Energy Metabolism , Immune Evasion , Nasopharyngeal Neoplasms/etiology , Nasopharyngeal Neoplasms/metabolism , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Humans , Metabolic Networks and Pathways , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/metabolism , Nasopharyngeal Neoplasms/pathology , Tumor Microenvironment
20.
Front Immunol ; 12: 720025, 2021.
Article in English | MEDLINE | ID: mdl-34484227

ABSTRACT

Inborn Errors of Immunity (IEI) comprise more than 450 inherited diseases, from which selected patients manifest a frequent and early incidence of malignancies, mainly lymphoma and leukemia. Primary antibody deficiency (PAD) is the most common form of IEI with the highest proportion of malignant cases. In this review, we aimed to compare the oncologic hallmarks and the molecular defects underlying PAD with other IEI entities to dissect the impact of avoiding immune destruction, genome instability, and mutation, enabling replicative immortality, tumor-promoting inflammation, resisting cell death, sustaining proliferative signaling, evading growth suppressors, deregulating cellular energetics, inducing angiogenesis, and activating invasion and metastasis in these groups of patients. Moreover, some of the most promising approaches that could be clinically tested in both PAD and IEI patients were discussed.


Subject(s)
Disease Susceptibility , Genetic Diseases, Inborn/complications , Genetic Diseases, Inborn/genetics , Immune System Diseases/complications , Neoplasms/etiology , Neoplasms/metabolism , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/immunology , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Disease Susceptibility/immunology , Energy Metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Genomic Instability , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immunity/genetics , Inflammation/complications , Inflammation/etiology , Inflammation/metabolism , Mutation , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...