1
|
Auti A, Tathode M, Marino MM, Vitiello A, Ballini A, Miele F, Mazzone V, Ambrosino A, Boccellino M. Nature's weapons: Bioactive compounds as anti-cancer agents. AIMS Public Health 2024; 11:747-772. [PMID: 39416904 PMCID: PMC11474324 DOI: 10.3934/publichealth.2024038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 10/19/2024] Open
Abstract
Cancer represents a major global health burden, prompting continuous research for effective therapeutic strategies. Natural compounds derived from plants have emerged as potential strategies for preventing cancer and treatment because of their inherent pharmacological properties. This comprehensive review aimed to evaluate the therapeutic potential of five key natural compounds: apigenin, quercetin, piperine, curcumin, and resveratrol in cancer prevention and therapy. By examining their molecular mechanisms and preclinical evidence, this review seeks to elucidate their role as potential adjuvants or stand-alone therapies in cancer management. The exploration of natural compounds as cancer therapeutics offers several advantages, including low toxicity, wide availability, and compatibility with conventional chemotherapeutic agents. We highlighted the current understanding of their anticancer mechanisms and clinical applications for advancing personalized cancer care to improve patient outcomes. We discussed the empirical findings from in vitro, in vivo, and clinical studies reporting biological activity and therapeutic efficacy in antioxidant, immunomodulatory, anti-carcinogenic, and chemo-sensitizing modes. Innovative delivery systems and personalized treatment approaches may further enhance their bioavailability and therapeutic utility in a synergistic approach with chemo- and radiotherapeutic disease management. This review underscores the importance of natural compounds in cancer prevention and treatment, promoting a multidisciplinary approach to the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Amogh Auti
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Madhura Tathode
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Maria Michela Marino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Antonio Vitiello
- Ministry of Health, Directorate-General for Health Prevention, 00144 Rome, Italy
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122, Foggia, Italy
| | - Francesco Miele
- General Surgery Unit, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Valeria Mazzone
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessia Ambrosino
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariarosaria Boccellino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
2
|
Cao L, Liu Q, Ma Y, Wang S. Identification of immune-related signature with prognosis in children with stage 4 and 4S neuroblastoma. Clin Transl Oncol 2024; 26:905-916. [PMID: 37709978 DOI: 10.1007/s12094-023-03320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE Spontaneous regression of tumors is an attractive phenomenon that most commonly occurs in stage 4S neuroblastoma (NB). However, the mechanism underlying this phenomenon remains unclear. METHODS Datasets correlated with NB were downloaded from online public databases, the differentially expressed genes (DEGs) between stage 4 and 4S associated with immunity were identified, and functional enrichment analysis was utilized to explore the potential functions and signaling pathways of these DEGs. In addition, based on these DEGs, a prognostic signature was constructed and validated, and differences in immune cell infiltration were analyzed. RESULTS A total of 13 DEGs were finally identified, and functional enrichment analysis revealed that these DEGs were primarily enriched in the positive regulation of neuron differentiation and TGF-β signaling pathway. The signature successfully stratifies patients into two risk score groups and performs well in judging prognosis and predicting overall survival time. In addition, the prognostic value of the risk score calculated by the signature was independent of clinical factors. The results of immune cell infiltration showed that patients with a high infiltration of resting CD4 + memory T cells had a better prognosis, while plasma cells had a worse prognosis. CONCLUSION The results of the functional enrichment analysis of these identified DEGs suggested that these DEGs may be related to spontaneous regression of NB. In addition, the prognostic signature has the potential to create new risk stratification in patients with NB.
Collapse
Affiliation(s)
- Lijian Cao
- Department of Pediatric Surgical Oncology, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University and the National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, People's Republic of China
| | - Qingqing Liu
- Department of Pediatric Surgical Oncology, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University and the National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, People's Republic of China
| | - Yue Ma
- Department of Pediatric Surgical Oncology, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University and the National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, People's Republic of China
| | - Shan Wang
- Department of Pediatric Surgical Oncology, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University and the National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, People's Republic of China.
| |
Collapse
|
3
|
Adetutu A, Aborisade AB, Ogunsina FA, Adegbola PI, Olaniyi TD. Ginger mitigated the health risks associated with arsenic-contamination of rats feed via inflammatory and apoptosis regulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115768. [PMID: 38064790 DOI: 10.1016/j.ecoenv.2023.115768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 01/12/2024]
Abstract
Millions of people around the world are inadvertently exposed to arsenic through drinking water and food. However, food spices possess antioxidants and anti-inflammatory potentials. Therefore, this study evaluated the protective potentials of Zingiber officinale (ginger) against the toxic effects of arsenic in male Wistar rats. Thirty-six Wistar rats were assigned into 6 groups (n = 6); group A1 and A2 (control), group B1 and B2 were fed with arsenic-contaminated feed (3.45x10-3 mg/kg), group C1 and C2 were feed with arsenic-contaminated feed (3.45x10-3 mg) supplemented with ginger respectively for 12 and 24 weeks. The blood, bone marrow, and liver of rats were harvested and prepared for various analyses. Micronucleus and Comet analysis were performed for the genotoxicity assessment every 4 weeks. Activities of AST, ALT, GGT, and SOD, and the concentration of GSH, MDA, protein carbonyl, protein thiol, and total protein, were measured by spectrophotometric methods. Quantification of IL-10, 1 L-1β, TNF-α, TGF-β NF-Ƙβ, and 8-oxodeoxyguanosine was done by ELISA method while Bax, Bcl2, and Erk 1/2 were quantified by immuno-histochemical staining. mRNA expression of cyclin D1 was quantified using qRT-PCR. Statistical analysis was performed with SPSS and statistical significance was accepted when p<0.05. Result showed significant (p<0.05) decrease in the haemoglobin concentration, red blood cell, lymphocyte counts, tail DNA and MnPCE of rats fed arsenic-contaminated feed compared with control. The supplementation with ginger significantly reduced serum activities of AST and GGT (p<0.05). Ginger supplementation also lowered the arsenic indued increases in liver MDA, protein carbonyl and 8-OXdG levels. Ginger restores to near normal the histological changes due to arsenic exposure. In the arsenic-exposed group, liver IL-10, IL-1β and TNF-α decreased significantly (p<0.05) at week 24 whereas, NF-Ƙβ and TGF-β increased significantly (p 0.05) at weeks 12 and 24 and TNF-α, Bcl2 at week 24. mRNA expression of cyclin D1 was significantly (p<0.05) downregulated in the arsenic and ginger-supplemented groups. This study showed that long-term consumption of arsenic resulted in immunosuppression, anaemia and activated anti-apoptotic process that was mitigated due to ginger supplementation.
Collapse
Affiliation(s)
- Adewale Adetutu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abiodun Bukunmi Aborisade
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Faith Ayotunde Ogunsina
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Peter Ifeoluwa Adegbola
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Department of Biochemistry and Forensic Science, First Technical University, Ibadan, Nigeria.
| | - Temitope Deborah Olaniyi
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
4
|
Doi H, Atsumi J, Baratz D, Miyamoto Y. A Phase I Study of TRK-250, a Novel siRNA-Based Oligonucleotide, in Patients with Idiopathic Pulmonary Fibrosis. J Aerosol Med Pulm Drug Deliv 2023; 36:300-308. [PMID: 37738329 DOI: 10.1089/jamp.2023.0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023] Open
Abstract
Purpose: TRK-250 is a novel single-stranded oligonucleotide carrying a human Transforming growth factor-beta 1-targeting siRNA motif tethered by two proline linkers. Nonclinical studies have shown that TRK-250 may have potency to prevent the progression of pulmonary fibrosis. Herein, a phase I study was conducted to investigate the safety and pharmacokinetics (PKs) of TRK-250 in patients with idiopathic pulmonary fibrosis (IPF). Method: In the phase I study, 34 IPF patients were partially randomized to receive a placebo or TRK-250 in 4 single doses of 2, 10, 30, and 60 mg or multiple rising doses of 10, 30, and 60 mg once per week for 4 weeks by oral inhalation. For both the single- and multiple-dose studies, the primary endpoint was safety, and the secondary endpoint was PKs. Result: In all IPF patients who orally inhaled TRK-250, no significant drug-related adverse events (AEs) were observed. The AEs were mild or moderate, except for one severe case with acute exacerbation. One of the more common AEs was coughing. One patient discontinued treatment before the last dose because of coughing. There were no medically important findings related to safety endpoints based on clinical laboratory data (clinical chemistry, hematology, or urinalysis), vital signs data, electrocardiogram data, physical examination findings, pulse oximetry data, spirometry data, or diffusing capacity of the lung for carbon monoxide data. All the bioanalytical results of PKs in the blood were below the lower limit of quantification. Conclusions: Both the single and multiple doses of TRK-250 were safe and well tolerated in this first study done in IPF patients. Furthermore, TRK-250 was not detected in the systemic circulation following inhalation, indicating low or virtually nonexistent systemic exposure. This study is registered at ClinicalTrials.gov with identifier number NCT03727802.
Collapse
Affiliation(s)
- Hiroyuki Doi
- Clinical Research Department, Toray Industries, Inc., Tokyo, Japan
| | - Jun Atsumi
- Clinical Research Department, Toray Industries, Inc., Tokyo, Japan
| | | | - Yohei Miyamoto
- Clinical Research Department, Toray Industries, Inc., Tokyo, Japan
| |
Collapse
|
5
|
Cao J, Xu H, Yu Y, Xu Z. Regulatory roles of cytokines in T and B lymphocytes-mediated immunity in teleost fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 144:104621. [PMID: 36801469 DOI: 10.1016/j.dci.2022.104621] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 06/05/2023]
Abstract
T and B lymphocytes (T and B cells) are immune effector cells that play critical roles in adaptive immunity and defend against external pathogens in most vertebrates, including teleost fish. In mammals, the development and immune response of T and B cells is associated with cytokines including chemokines, interferons, interleukins, lymphokines, and tumor necrosis factors during pathogenic invasion or immunization. Given that teleost fish have evolved a similar adaptive immune system to mammals with T and B cells bearing unique receptors (B-cell receptors (BCRs) and T-cell receptors (TCRs)) and that cytokines in general have been identified, whether the regulatory roles of cytokines in T and B cell-mediated immunity are evolutionarily conserved between mammalians and teleost fish is a fascinating question. Thus, the purpose of this review is to summarize the current knowledge of teleost cytokines and T and B cells as well as the regulatory roles of cytokines on these two types of lymphocytes. This may provide important information on the parallelisms and dissimilarities of the functions of cytokines in bony fish versus higher vertebrates, which may aid in the evaluation and development of adaptive immunity-based vaccines or immunostimulants.
Collapse
Affiliation(s)
- Jiafeng Cao
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Haoyue Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yongyao Yu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhen Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
6
|
Hernandez-Cazares F, Maqueda-Alfaro RA, Lopez-Saucedo C, Martinez-Barnetche J, Yam-Puc JC, Estrada-Parra S, Flores-Romo L, Estrada-Garcia T. Elevated levels of enteric IgA in an unimmunised mouse model of Hyper IgM syndrome derived from gut-associated secondary lymph organs even in the absence of germinal centres. Front Cell Infect Microbiol 2023; 13:1172021. [PMID: 37457961 PMCID: PMC10339347 DOI: 10.3389/fcimb.2023.1172021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Patients with Human Hyper IgM syndromes (HIGM) developed pulmonary and gastrointestinal infections since infancy and most patients have mutations in the CD40 ligand (CD40L) gene. Most HIGM patients compared to healthy subjects have higher/similar IgM and lower IgG, and IgA serum concentrations but gut antibody concentrations are unknown. CD40L on activated T-cells interacts with CD40 on B-cells, essential for the formation of germinal centres (GCs) inside secondary lymphoid organs (SLOs), where high-affinity antibodies, long-lived antibody-secreting plasma cells, and memory B-cells, are produced. C57BL6-CD40 ligand deficient mice (C57BL6-cd40l -/-), are a model of HIGM, because serum immunoglobulin concentrations parallel levels observed in HIGM patients and have higher faecal IgA concentrations. In mice, TGFβ and other cytokines induce IgA production. Aims To compare and evaluate B-cell populations and IgA-producing plasma cells in peritoneal lavage, non-gut-associated SLOs, spleen/inguinal lymph nodes (ILN), and gut-associated SLOs, mesenteric lymph nodes (MLN)/Peyer´s patches (PP) of unimmunised C57BL6-cd40l -/- and C57BL6-wild-type (WT) mice. Material and methods Peritoneal lavages, spleens, ILN, MLN, and PP from 8-10 weeks old C57BL6-cd40l -/- and WT mice, were obtained. Organ cryosections were analysed by immunofluorescence and B-cell populations and IgA-positive plasma cell suspensions by flow cytometry. Results In unimmunised WT mice, GCs were only observed in the gut-associated SLOs, but GCs were absent in all C57BL6-cd40l -/- SLOs. PP and MLN of C57BL6-cd40l -/- mice exhibited a significantly higher number of IgA-producing cells than WT mice. In the spleen and ILN of C57BL6-cd40l- /- mice IgA-producing cells significantly decreased, while IgM-positive plasma cells increased. C57BL6-cd40l -/- B-1 cells were more abundant in all analysed SLOs, whereas in WT mice most B-1 cells were contained within the peritoneal cavity. C57BL6-cd40l -/- B-cells in MLN expressed a higher TGFβ receptor-1 than WT mice. Mouse strains small intestine microvilli (MV), have a similar frequency of IgA-positive cells. Discussion Together our results confirm the role of PP and MLN as gut inductive sites, whose characteristic features are to initiate an IgA preferential immune response production in these anatomical sites even in the absence of GCs. IgA antibodies play a pivotal role in neutralising, eliminating, and regulating potential pathogens and microorganisms in the gut.
Collapse
Affiliation(s)
| | | | | | - Jesus Martinez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | | | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | | |
Collapse
|
7
|
Devlin MJ, Miller R, Laforets F, Kotantaki P, Garsed DW, Kristeleit R, Bowtell DD, McDermott J, Maniati E, Balkwill FR. The Tumor Microenvironment of Clear-Cell Ovarian Cancer. Cancer Immunol Res 2022; 10:1326-1339. [PMID: 36095166 PMCID: PMC9627265 DOI: 10.1158/2326-6066.cir-22-0407] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/01/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023]
Abstract
Some patients with advanced clear-cell ovarian cancer (CCOC) respond to immunotherapy; however, little is known about the tumor microenvironment (TME) of this relatively rare disease. Here, we describe a comprehensive quantitative and topographical analysis of biopsies from 45 patients, 9 with Federation Internationale des Gynaecologistes et Obstetristes (FIGO) stage I/II (early CCOC) and 36 with FIGO stage III/IV (advanced CCOC). We investigated 14 immune cell phenotype markers, PD-1 and ligands, and collagen structure and texture. We interrogated a microarray data set from a second cohort of 29 patients and compared the TMEs of ARID1A-wildtype (ARID1Awt) versus ARID1A-mutant (ARID1Amut) disease. We found significant variations in immune cell frequency and phenotype, checkpoint expression, and collagen matrix between the malignant cell area (MCA), leading edge (LE), and stroma. The MCA had the largest population of CD138+ plasma cells, the LE had more CD20+ B cells and T cells, whereas the stroma had more mast cells and αSMA+ fibroblasts. PD-L2 was expressed predominantly on malignant cells and was the dominant PD-1 ligand. Compared with early CCOC, advanced-stage disease had significantly more fibroblasts and a more complex collagen matrix, with microarray analysis indicating "TGFβ remodeling of the extracellular matrix" as the most significantly enriched pathway. Data showed significant differences in immune cell populations, collagen matrix, and cytokine expression between ARID1Awt and ARID1Amut CCOC, which may reflect different paths of tumorigenesis and the relationship to endometriosis. Increased infiltration of CD8+ T cells within the MCA and CD4+ T cells at the LE and stroma significantly associated with decreased overall survival.
Collapse
Affiliation(s)
- Michael-John Devlin
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- Department of Medical Oncology, St Bartholomew's Hospital, London, United Kingdom
| | - Rowan Miller
- Department of Medical Oncology, St Bartholomew's Hospital, London, United Kingdom
- Department of Medical Oncology, University College London Hospital, London, United Kingdom
| | - Florian Laforets
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Dale W. Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Rebecca Kristeleit
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Jacqueline McDermott
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Frances R. Balkwill
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
8
|
Maheshwari A, Traub TM, Garg PM, Ethawi Y, Buonocore G. Necrotizing Enterocolitis: Clinical Features, Histopathological Characteristics, and Genetic Associations. Curr Pediatr Rev 2022; 18:210-225. [PMID: 35125082 DOI: 10.2174/1573396318666220204113858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis seen in premature infants. Although the etiopathogenesis of NEC is unclear, genetic factors may alter a patient's susceptibility, clinical course, and outcomes. This review draws from existing studies focused on individual genes and others based on microarray-based high-throughput discovery techniques. We have included evidence from our own studies and from an extensive literature search in the databases PubMed, EMBASE, and Scopus. To avoid bias in the identification of studies, keywords were short-listed a priori from anecdotal experience and PubMed's Medical Subject Heading (MeSH) thesaurus.
Collapse
Affiliation(s)
| | - Terri M Traub
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Parvesh M Garg
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, University of Mississippi, Jackson, Mississippi, USA
| | - Yahya Ethawi
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, Saudi American Hospital, Ajman, United Arab Emirates
| | - Giuseppe Buonocore
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics/ Neonatology, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Sun Z, Sun X, Chen Z, Du J, Wu Y. Head and Neck Squamous Cell Carcinoma: Risk Factors, Molecular Alterations, Immunology and Peptide Vaccines. Int J Pept Res Ther 2021; 28:19. [PMID: 34903958 PMCID: PMC8653808 DOI: 10.1007/s10989-021-10334-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/29/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) arises from the epithelial lining of the oral cavity, hypopharynx, oropharynx, and larynx. There are several potential risk factors that cause the generation of HNSCC, including cigarette smoking, alcohol consumption, betel quid chewing, inadequate nutrition, poor oral hygiene, HPV and Epstein–Barr virus, and Candida albicans infections. HNSCC has causative links to both environmental factors and genetic mutations, with the latter playing a more critical role in cancer progression. These molecular changes to epithelial cells include the inactivation of cancer suppressor genes and proto-oncogenes overexpression, resulting in tumour cell proliferation and distant metastasis. HNSCC patients have impaired dendritic cell (DC) and natural killer (NK) cell functions, increased production of higher immune-suppressive molecules, loss of regulatory T cells and co-stimulatory molecules and major histocompatibility complex (MHC) class Ι molecules, lower number of lymphocyte subsets, and a poor response to antigen-presenting cells. At present, the standard treatment modalities for HNSCC patients include surgery, chemotherapy and radiotherapy, and combinatorial therapy. Despite advances in the development of novel treatment modalities over the last few decades, survival rates of HNSCC patients have not increased. To establish effective immunotherapies, a greater understanding of interactions between the immune system and HNSCC is required, and there is a particular need to develop novel therapeutic options. A therapeutic cancer vaccine has been proposed as a promising method to improve outcome by inducing a powerful adaptive immune response that leads to cancer cell elimination. Compared with other vaccines, peptide cancer vaccines are more robust and specific. In the past few years, there have been remarkable achievements in peptide-based vaccines for HNSCC patients. Here, we summarize the latest molecular alterations in HNSCC, explore the immune response to HNSCC, and discuss the latest developments in peptide-based cancer vaccine strategies. This review highlights areas for valuable future research focusing on peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, Shandong 250000 China
| | - Zhanwei Chen
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Juan Du
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| | - Yihua Wu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China
| |
Collapse
|
10
|
Hoffmann JC, Schön MP. Integrin α E(CD103)β 7 in Epithelial Cancer. Cancers (Basel) 2021; 13:6211. [PMID: 34944831 PMCID: PMC8699740 DOI: 10.3390/cancers13246211] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 01/22/2023] Open
Abstract
Interactions of both the innate and the adaptive immune system with tumors are complex and often influence courses and therapeutic treatments in unanticipated ways. Based on the concept that CD8+T cells can mediate important antitumor effects, several therapies now aim to amplify their specific activity. A subpopulation of CD8+ tissue-resident T lymphocytes that express the αE(CD103)β7 integrin has raised particular interest. This receptor presumably contributes to the recruitment and retention of tumor-infiltrating immune cells through interaction with its ligand, E-cadherin. It appears to have regulatory functions and is thought to be a component of some immunological synapses. In TGF-rich environments, the αE(CD103)β7/E-cadherin-interaction enhances the binding strength between tumor cells and infiltrating T lymphocytes. This activity facilitates the release of lytic granule contents and cytokines as well as further immune responses and the killing of target cells. Expression of αE(CD103)β7 in some tumors is associated with a rather favorable prognosis, perhaps with the notable exception of squamous cell carcinoma of the skin. Although epithelial skin tumors are by far the most common tumors of fair-skinned people, there have been very few studies on the distribution of αE(CD103)β7 expressing cells in these neoplasms. Given this background, we describe here that αE(CD103)β7 is scarcely present in basal cell carcinomas, but much more abundant in squamous cell carcinomas with heterogeneous distribution. Notwithstanding a substantial number of studies, the role of αE(CD103)β7 in the tumor context is still far from clear. Here, we summarize the essential current knowledge on αE(CD103)β7 and outline that it is worthwhile to further explore this intriguing receptor with regard to the pathophysiology, therapy, and prognosis of solid tumors.
Collapse
Affiliation(s)
- Johanna C. Hoffmann
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Michael P. Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, 37075 Göttingen, Germany;
- Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
11
|
Grases-Pintó B, Abril-Gil M, Torres-Castro P, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. Rat Milk and Plasma Immunological Profile throughout Lactation. Nutrients 2021; 13:nu13041257. [PMID: 33920419 PMCID: PMC8070501 DOI: 10.3390/nu13041257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
The composition of bioactive factors with immune activity in human breast milk is widely studied. However, the knowledge on rat milk immune factors during the whole lactation period is still scarce. This study aimed to analyze rat breast milk’s immunoglobulin (Ig) content and some critical adipokines and growth factors throughout the lactation period, and to assess relationships with corresponding plasma levels. During lactation, milk concentration of the transforming growth factor (TGF)-β2 and -β3 showed a punctual increase in the first week, whereas adiponectin and leptin remained stable. In the second period of lactation (d14–21), despite the increase in the milk epidermal growth factor (EGF), a decrease in fibroblast growth factor 21 (FGF21) was detected at day 21. Milk IgA concentration had a progressive increase during lactation, while no significant changes were found in IgM and IgG. Regarding plasma levels, a decrease in all studied adipokines was observed in the second period of lactation, with the exception of IgA and TGF-β1, which reached their highest values at the end of the study. A positive correlation in IgM, IgG, and adipokine concentration was detected between milk and plasma compartments. In summary, the changes in the pattern of these bioactive compounds in rat milk and plasma and their relationships during lactation are established.
Collapse
Affiliation(s)
- Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Mar Abril-Gil
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Paulina Torres-Castro
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
12
|
Hamed A, Todd I, Tighe PJ, Powell RJ, Harrison T, Fairclough LC. Array-based measurements of aero-allergen-specific IgE correlate with skin-prick test reactivity in asthma regardless of specific IgG4 or total IgE measurements. J Immunol Methods 2021; 492:112999. [PMID: 33609533 DOI: 10.1016/j.jim.2021.112999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/28/2020] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Skin prick testing (SPT) and measurement of serum allergen-specific IgE (sIgE) are used to investigate asthma and other allergic conditions. Measurement of serum total IgE (tIgE) and allergen-specific IgG4 (sIgG4) may also be useful. The aim was to ascertain the correlation between these serological parameters and SPT. Sera from 60 suspected asthmatic patients and 18 healthy controls were assayed for sIgE and sIgG4 reactivity against a panel of 70 SPT allergen preparations, and for tIgE. The patients were also assessed by skin prick tests for reactivity to cat, dog, house dust mite and grass allergens. Over 50% of the patients had tIgE levels above the 75th percentile of the controls. 58% of patients and 39% of controls showed sIgE reactivity to ≥1 allergen. The mean number of allergens detected by sIgE was 3.1 in suspected asthma patients and 0.9 in controls. 58% of patients and 50% of controls showed sIgG4 reactivity to ≥1 allergen. The mean number of allergens detected by sIgG4 was 2.5 in patients and 1.7 in controls. For the patients, a strong correlation was observed between clinical SPT reactivity and serum sIgE levels to cat, dog, house dust mite (HDM) and grass allergens. SPT correlations using sIgE/sIgG4 or sIgE/tIgE ratios were not markedly higher. The measurement of serum sIgE by microarray using SPT allergen preparations showed good correlation with clinical SPT reactivity to cat, dog, HDM and grass allergens. This concordance was not improved by measuring tIgE or sIgG4.
Collapse
Affiliation(s)
- Aljali Hamed
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2UH, UK; Department of Laboratory Medicine, Faculty of Medical Technology, Omar Al-Mukhtar University, Al Bayda City, Libya
| | - Ian Todd
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2UH, UK
| | - Patrick J Tighe
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2UH, UK
| | - Richard J Powell
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2UH, UK
| | - Tim Harrison
- School of Medicine, Division of Respiratory Medicine, Clinical Sciences Building, City Hospital Campus, University of Nottingham, Nottingham NG5 1PB, UK
| | - Lucy C Fairclough
- School of Life Sciences, The University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
13
|
Gerlach MM, Stelling-Germani A, Ting Wu C, Newrzela S, Döring C, Vela V, Müller A, Hartmann S, Tzankov A. SMAD1 promoter hypermethylation and lack of SMAD1 expression in Hodgkin lymphoma: a potential target for hypomethylating drug therapy. Haematologica 2021; 106:619-621. [PMID: 32299904 PMCID: PMC7849564 DOI: 10.3324/haematol.2020.249276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/14/2020] [Indexed: 11/16/2022] Open
Affiliation(s)
- Magdalena M Gerlach
- Institute of Medical Genetics and Pathology, University of Basel, Basel, Switzerland
| | | | - Cheuk Ting Wu
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Sebastian Newrzela
- Institute of Pathology, Department of Pathology, Goethe University Frankfurt on Main, Germany
| | - Claudia Döring
- Institute of Pathology, Department of Pathology, Goethe University Frankfurt on Main, Germany
| | - Visar Vela
- Institute of Medical Genetics and Pathology, University of Basel, Basel, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Sylvia Hartmann
- Institute of Pathology, Department of Pathology, Goethe University Frankfurt on Main, Germany
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
Zidan AA, Al-Hawwas M, Perkins GB, Mourad GM, Stapledon CJM, Bobrovskaya L, Zhou XF, Hurtado PR. Characterization of Urine Stem Cell-Derived Extracellular Vesicles Reveals B Cell Stimulating Cargo. Int J Mol Sci 2021; 22:E459. [PMID: 33466423 PMCID: PMC7796485 DOI: 10.3390/ijms22010459] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 12/31/2020] [Accepted: 01/01/2021] [Indexed: 12/22/2022] Open
Abstract
Elucidation of the biological functions of extracellular vesicles (EVs) and their potential roles in physiological and pathological processes is an expanding field of research. In this study, we characterized USC-derived EVs and studied their capacity to modulate the human immune response in vitro. We found that the USC-derived EVs are a heterogeneous population, ranging in size from that of micro-vesicles (150 nm-1 μm) down to that of exosomes (60-150 nm). Regarding their immunomodulatory functions, we found that upon isolation, the EVs (60-150 nm) induced B cell proliferation and IgM antibody secretion. Analysis of the EV contents unexpectedly revealed the presence of BAFF, APRIL, IL-6, and CD40L, all known to play a central role in B cell stimulation, differentiation, and humoral immunity. In regard to their effect on T cell functions, they resembled the function of mesenchymal stem cell (MSC)-derived EVs previously described, suppressing T cell response to activation. The finding that USC-derived EVs transport a potent bioactive cargo opens the door to a novel therapeutic avenue for boosting B cell responses in immunodeficiency or cancer.
Collapse
Affiliation(s)
- Asmaa A. Zidan
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt;
- Centre of Excellence for Research in Regenerative Medicine Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt
| | - Mohammed Al-Hawwas
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Griffith B. Perkins
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA 5000, Australia;
| | - Ghada M. Mourad
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt;
- Centre of Excellence for Research in Regenerative Medicine Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt
| | | | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Plinio R. Hurtado
- Department of Renal Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
15
|
Yu Z, Zhou T, Luo Y, Dong L, Li C, Liu J, Luo J, Yan R, Xu L, Song X, Li X. Modulation Effects of Toxoplasma gondii Histone H2A1 on Murine Macrophages and Encapsulation with Polymer as a Vaccine Candidate. Vaccines (Basel) 2020; 8:vaccines8040731. [PMID: 33287313 PMCID: PMC7761694 DOI: 10.3390/vaccines8040731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/21/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is the most common zoonotic protozoa and has infected about one-third of the population worldwide. Recombinant epitopes encapsulated in nanospheres have advantages over traditional T. gondii vaccines. For an efficient delivery system, poly (DL-lactide-co-glycolide) (PLGA) and chitosan are the most frequently used biodegradable polymeric nanospheres with strong safety profiles. In the present study, we first expressed and purified histone H2A1 of T. gondii using the prokaryotic expression system. The effects of recombinant TgH2A1 on the functions of murine macrophages were then studied. Purified recombinant TgH2A1 was then encapsulated in nanospheres with PLGA and chitosan. After subcutaneous vaccination in mice, the immune response was evaluated by double antibody sandwich ELISA kits. The results from this study showed that PLGA and chitosan loaded with rTgH2A1 could trigger a stronger Th1 oriented immune response and prolong the survival time of mice effectively. In conclusion, PLGA and chitosan nanospheres loaded with histone H2A1 are an effective method for the development of vaccines against T. gondii. Further studies should focus on evaluating the regulatory mechanism of TgH2A1, vaccine potency, and cellular response in chronic T. gondii infections.
Collapse
Affiliation(s)
- Zhengqing Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Tianyuan Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Yanxin Luo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Lu Dong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Chunjing Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Junlong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (J.L.); (J.L.)
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (J.L.); (J.L.)
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Z.Y.); (T.Z.); (Y.L.); (L.D.); (C.L.); (R.Y.); (L.X.); (X.S.)
- Correspondence:
| |
Collapse
|
16
|
Pal S, Nath S, Meininger CJ, Gashev AA. Emerging Roles of Mast Cells in the Regulation of Lymphatic Immuno-Physiology. Front Immunol 2020; 11:1234. [PMID: 32625213 PMCID: PMC7311670 DOI: 10.3389/fimmu.2020.01234] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Mast cells (MCs) are abundant in almost all vascularized tissues. Furthermore, their anatomical proximity to lymphatic vessels and their ability to synthesize, store and release a large array of inflammatory and vasoactive mediators emphasize their significance in the regulation of the lymphatic vascular functions. As a major secretory cell of the innate immune system, MCs maintain their steady-state granule release under normal physiological conditions; however, the inflammatory response potentiates their ability to synthesize and secrete these mediators. Activation of MCs in response to inflammatory signals can trigger adaptive immune responses by dendritic cell-directed T cell activation. In addition, through the secretion of various mediators, cytokines and growth factors, MCs not only facilitate interaction and migration of immune cells, but also influence lymphatic permeability, contractility, and vascular remodeling as well as immune cell trafficking through the lymphatic vessels. In summary, the consequences of these events directly affect the lymphatic niche, influencing inflammation at multiple levels. In this review, we have summarized the recent advancements in our understanding of the MC biology in the context of the lymphatic vascular system. We have further highlighted the MC-lymphatic interaction axis from the standpoint of the tumor microenvironment.
Collapse
Affiliation(s)
- Sarit Pal
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Bryan, TX, United States
| | - Shubhankar Nath
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Cynthia J Meininger
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Bryan, TX, United States
| | - Anatoliy A Gashev
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Bryan, TX, United States
| |
Collapse
|
17
|
Ehsan M, Gadahi JA, Liu T, Lu M, Wang Y, Hasan MW, Haseeb M, Yan R, Xu L, Song X, Zhu XQ, Li X. Identification of a novel methyltransferase-type 12 protein from Haemonchus contortus and its effects on functions of goat PBMCs. Parasit Vectors 2020; 13:154. [PMID: 32228657 PMCID: PMC7106832 DOI: 10.1186/s13071-020-04028-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/17/2020] [Indexed: 01/06/2023] Open
Abstract
Background Methyltransferases (MTFs) are broad range of enzymes, which are ubiquitously expressed in diverse organisms ranging from bacteria to animals. MTFs proteins have been associated with various biological/cellular processes including transcriptional regulation, subcellular protein and RNA localization, signal transduction and DNA-damage repair. However, the role of MTFs in immune mechanism during host–parasite interaction has not been addressed yet. Results An open reading frame (764 bp) of methyltransferase-type 12 gene of H. contortus denoted as HcMTF-12, was successfully cloned using reverse transcriptase-polymerase chain reaction (RT-PCR) followed by prokaryotic expression in Escherichia coli BL21 (DE3 strain). The recombinant HcMTF-12 protein (rHcMTF-12) was about 47 kDa along with a fusion vector protein of 18 kDa. Immunoblot results identified the native protein MTF-12 with antibodies produced in rats against rHcMT-12, whereas rHcMTF-12 protein was recognized with sera of goat experimentally infected with H. contortus. Immunohistochemical analysis revealed that the native MTF-12 protein was mainly located in the periphery (cuticle) of parasite sections as well as within the pharynx and intestinal region. An immunofluorescence assay validated that rHcMTF-12 attached to the surface of goat PBMCs. Furthermore, the cytokines transcription of IL-2, IFN-γ and IL-4 transcripts of PBMCs incubated with rHcMTF-12 were enhanced in a dose-dependent manner. The secretion of TGF-β1 and IL-10 was significantly decreased. However, IL-6 production was not significantly different as compared to the control groups. Moreover, the migration activity and nitric oxide (NO) production by PBMCs were induced considerably, whereas the proliferation of PBMCs cells was negatively affected when incubated with the rHcMTF-12 protein. Conclusions Our findings suggest that HcMTF-12 significantly mediated the functions of PBMCs, and it might be a potential candidate for therapeutic interventions against haemonchosis.![]()
Collapse
Affiliation(s)
- Muhammad Ehsan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Javaid A Gadahi
- Department of Veterinary Parasitology, Sindh Agriculture University Tandojam, Hyderabad, Pakistan
| | - Tingqi Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Mingmin Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Yujian Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Muhammad W Hasan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Muhammad Haseeb
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, People's Republic of China
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Characterization of Haemonchus contortus Excretory/Secretory Antigen (ES-15) and Its Modulatory Functions on Goat Immune Cells In Vitro. Pathogens 2020; 9:pathogens9030162. [PMID: 32120801 PMCID: PMC7157690 DOI: 10.3390/pathogens9030162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 01/11/2023] Open
Abstract
Small size excretory/secretory (ES) antigens of the Haemonchus contortus parasite have intense interest among researchers for understanding the molecular basis of helminths immune regulation in term of control strategies. Immunomodulatory roles of H. contortus ES-15 kDa (HcES-15) on host immune cells during host–parasite interactions are unknown. In this study, the HcES-15 gene was cloned and expression of recombinant protein (rHcES-15) was induced by isopropyl-ß-d-thiogalactopyranoside (IPTG). Binding activity of rHcES-15 to goat peripheral blood mononuclear cells (PBMCs) was confirmed by immunofluorescence assay (IFA) and immunohistochemical analysis showed that H. contortus 15 kDa protein localized in the outer and inner structure of the adult worm, clearly indicated as the parasite’s ES antigen. The immunoregulatory role on cytokines production, cell proliferation, cell migration, nitric oxide (NO) production, apoptosis, and phagocytosis were observed by co-incubation of rHcES-15 with goat PBMCs. The results showed that cytokines IL-4, IL-10, IL-17, the production of nitric oxide (NO), PBMCs apoptosis, and monocytes phagocytosis were all elevated after cells incubated with rHcES-15 at differential protein concentrations. We also found that IFN-γ, TGF-β1, cells proliferation and migration were significantly suppressed with the interaction of rHcES-15 protein. Our findings indicated that low molecular ES antigens of H. contortus possessed discrete immunoregulatory roles, which will help to understand the mechanisms involved in immune evasion by the parasite during host–parasite interactions.
Collapse
|
19
|
Positive and negative cooperativity of TNF and Interferon-γ in regulating synovial fibroblast function and B cell survival in fibroblast/B cell co-cultures. Sci Rep 2020; 10:780. [PMID: 31964950 PMCID: PMC6972945 DOI: 10.1038/s41598-020-57772-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/07/2020] [Indexed: 12/21/2022] Open
Abstract
Synovial fibroblasts (SF) were reported to produce B cell activating factor (BAFF) in response to stimulation with interferon-γ (IFN-γ) or tumor necrosis factor (TNF). However, the influence of these pro-inflammatory cytokines on other receptors/ligands of the TNF superfamily or associated cytokine receptors in SF has not been investigated yet. Here we show the differential regulation of BAFF (CD257), Fn14 (CD266), TACI (CD267), BAFF-R (CD268), BCMA (CD269), CD40 ligand (CD40L, CD154), IFN-γR (CD119), Leptin receptor (ObR, CD295), VCAM-1 (CD106) and membrane TGF-β in isolated SF and the impact of IFN-γ/TNF co-incubation on proliferation, IL-6 and IL-8 production. In addition, the impact of differentially stimulated SF on B cell survival in co-cultures was assessed. Surface cytokines and cytokine receptors were detected by flow cytometry. Soluble cytokine receptors and cytokines were quantified by ELISA. Proliferation was assessed by cell titer blue. Murine B cell survival in fibroblast/ B cell co-cultures was determined by annexin V/propidium iodide staining and flow cytometry. IFN-γ together with TNF synergistically and significantly increased the cell surface levels of BAFF, Fn14, TACI, BAFF-R, BCMA, CD40L, ObR and IFN-γR in rheumatoid arthritis SF after 72 h incubation. Soluble BAFF was only induced by IFN-γ and inhibited by TNF. Addition of TWEAK had no influence on proliferation or IL-8 production but decreased TNF-induced IL-6 production, whereas APRIL, BAFF and leptin did not modulate TNF or TNF/IFN-γ-induced proliferation or cytokine production. Proliferation was increased by TNF and further enhanced by the addition of IFN-γ. In co-culture experiments, SF stimulated with TNF/IFN but not TNF or IFN-γ alone increased shedding of VCAM-1 and expression of membrane TGFβ, which was associated with reduced survival of murine B cells. IFN-γ and TNF regulate the expression of TNF family member cytokines and associated receptors. Ligation of IFN-γR and Fn14 under pro-inflammatory conditions modulated IL-6/IL-8 production and proliferation. In B cell/SF co-cultures, the combination of TNF/IFN reduced B cell survival possibly via enhanced VCAM-1 shedding and/or increased TGF-β production. IFN-γ is necessary for the observed effects on B cell survival and SF cytokine production and emphasizes its anti-inflammatory role in rheumatoid arthritis.
Collapse
|
20
|
Wu L, Yang Y, Kong L, Bian X, Guo Z, Fu S, Liang F, Li B, Ye J. Comparative transcriptome analysis of the transcriptional heterogeneity in different IgM + cell subsets from peripheral blood of Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2019; 93:612-622. [PMID: 31408730 DOI: 10.1016/j.fsi.2019.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
In teleost fish, IgM+ B cells play important roles in innate and adaptive immunity. Different IgM+ B cells are detected in teleost, named IgMlo and IgMhi B cell subsets, according to the distinct expression levels of membrane IgM (mIgM). However, the study on the heterogeneity in IgM+ B cell subsets remains poorly understood. In this study, the comparative transcriptomic profiles of IgM-, IgMlo and IgMhi from peripheral blood of Nile tilapia (Oreochromis niloticus) were carried out by using RNA-sequencing technique. A total of 6045 and 5470 differentially expressed genes (DEGs) were detected in IgMlo and IgMhi cells, respectively, as compared with IgM- lymphocytes, whereas 3835 genes were differentially expressed when IgMlo compared to IgMhi cells. Analysis of the KEGG database indicated that the DEGs were enriched in immune system categories and signaling transduction and interaction in IgM- vs IgMhi, IgM- vs IgMlo and IgMlo vs IgMhi. Comparatively, in IgMlo vs IgMhi, GO enrichment analysis indicated that the DEGs enriched in nucleic acid binding transcription factor activity. Analysis of crucial transcription factors for B cell differentiation indicated that IgMlo and IgMhi cell clusters belonged to the different B cell subsets. The data generated in this study may provide insights into understanding the heterogeneity of IgM+ cells in teleost, and suggest that IgM+ B cells play a crucial role in innate immunity.
Collapse
Affiliation(s)
- Liting Wu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Yanjian Yang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Linghe Kong
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Xia Bian
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Zheng Guo
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Shengli Fu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Fang Liang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Bingxi Li
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Jianmin Ye
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China.
| |
Collapse
|
21
|
Shukla A, Chen C, Jellusova J, Leung CR, Kao E, Bhat N, Lin WW, Apgar JR, Rickert RC. Self-reactive B cells in the GALT are actively curtailed to prevent gut inflammation. JCI Insight 2019; 5:130621. [PMID: 31335327 DOI: 10.1172/jci.insight.130621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Immune homeostasis in the gut associated lymphoid tissues (GALT) is critical to prevent the development of inadvertent pathologies. B cells as the producers of antibodies and cytokines plays an important role in maintaining the GALT homeostasis. However, the mechanism by which B cells specifically direct their responses towards non-self-antigens and become ignorant to self-antigens in the GALT is not known. Therefore, we developed a novel mouse model by expressing Duck Egg Lysozyme (DEL) in gut epithelial cells in presence of HEL reactive B cells. Notably, we observed a transient activation and rapid deletion of self-reactive B cells in Peyers Patches and Mesenteric lymph nodes upon self-antigen exposure. The survival of self-reactive B cells upon exposure to their self-antigen was partially rescued by blocking receptor editing but could be completely rescued by stronger survival signal like ectopic expression of BCL2. Importantly, rescuing the self-reactive B cells promoted production of auto-antibodies and gut inflammation. Mechanistically, we identify a specific activation of TGFβ signaling in self-reactive B cells in the gut and a critical role of this pathway in maintaining peripheral tolerance. Collectively, our studies describe functional consequences and fate of self-reactive B cells in GALT and provide novel mechanistic insights governing self-tolerance of B cells in the gut.
Collapse
|
22
|
Wang M, Gu Z, Yang J, Zhao H, Cao Z. Changes among TGF-β1+ Breg cells and helper T cell subsets in a murine model of allergic rhinitis with prolonged OVA challenge. Int Immunopharmacol 2019; 69:347-357. [DOI: 10.1016/j.intimp.2019.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/18/2018] [Accepted: 01/07/2019] [Indexed: 01/08/2023]
|
23
|
Maskarinec G, Ju D, Fong J, Horio D, Chan O, Loo LWM, Hernandez BY. Mammographic density and breast tissue expression of inflammatory markers, growth factors, and vimentin. BMC Cancer 2018; 18:1191. [PMID: 30497427 PMCID: PMC6267911 DOI: 10.1186/s12885-018-5088-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Background Mammographic density is a known risk factor for breast cancer, but the underlying pathologic characteristics are not well understood. The current analysis investigated the expression of several markers of interest, e.g., inflammation and growth, with mammographic density (MD) in normal and malignant breast tissue specimens from 279 women of the Multiethnic Cohort (MEC). Methods Breast cancer cases, recruited from a nested case-control study within the MEC, provided mammograms for density evaluation. Protein expression (COX-2, TNF-α, TGF-β, IGF-1R, IGFBP-2, and vimentin) was assessed by immunohistochemical detection. Linear regression was applied to evaluate the relation between marker expression and percent density and to compute adjusted means with 95% confidence intervals (CI) by marker status while adjusting for confounders. Results Due to missing cores and tissue, normal tissue could only be evaluated for COX-2 and vimentin. No significant associations with mammographic density were detected for all markers analyzed. For inflammatory markers (TNF-α, COX-2, and TGF-β) in tumor tissue, MD were non-significantly higher with stronger expression but the differences were very small. For example, the mean MD values for no, weak, and strong TNF-α expression were 35% (95% CI 24–47%), 39% (95% CI 29–48%), and 38% (95% CI 27–50%). In a posthoc analysis among postmenopausal women only, the difference across categories of TNF-α expression increased to 25% (95% CI 12–39%), 35% (95% CI 23–48%), and 35% (95% CI 20–49%). Conclusions The current analysis offers little support for an involvement of immunohistochemical markers representing inflammatory and growth factor pathways as predictors of breast density. Electronic supplementary material The online version of this article (10.1186/s12885-018-5088-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gertraud Maskarinec
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Dan Ju
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Jaimie Fong
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - David Horio
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Owen Chan
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Lenora W M Loo
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Brenda Y Hernandez
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| |
Collapse
|
24
|
Ehsan M, Wang W, Gadahi JA, Hasan MW, Lu M, Wang Y, Liu X, Haseeb M, Yan R, Xu L, Song X, Li X. The Serine/Threonine-Protein Phosphatase 1 From Haemonchus contortus Is Actively Involved in Suppressive Regulatory Roles on Immune Functions of Goat Peripheral Blood Mononuclear Cells. Front Immunol 2018; 9:1627. [PMID: 30061894 PMCID: PMC6054924 DOI: 10.3389/fimmu.2018.01627] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Serine/threonine-protein phosphatases (STPs), as integral constituents of parasitic excretory/secretory proteins, are assumed to be released during the host–parasite interactions. However, knowledge about these phosphatases and their immunoregulatory and immune protective efficiencies with host peripheral blood mononuclear cells (PBMCs) is scant. In this study, an open reading frame of STP from Haemonchus contortus designated as HcSTP-1 was amplified and cloned using reverse-transcription-polymerase chain reaction (RT-PCR) method. The 951-bp nucleotides sequence was encoded to a protein of 316 amino acid residues, conserved in characteristics motifs GDXHG, GDYVDRG, GNHE, HGG, RG, and H. The HcSTP-1 protein was detected at approximately 35 kDa as recombinant protein fused in an expression vector system and resolved on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Immunohistochemically, HcSTP-1 was found to be localized in both male and female adult worm sections. Using immunofluorescence assay, the binding activity of rHcSTP-1 was confirmed on surface of goat PBMCs, which resulted in expression of multiple cytokines and various immunoregulatory activities in vitro. The RT-PCR results showed that mRNA level of interleukin-2, TGF-β1, IFN-γ, and IL-17 (with 10 µg/ml) was upregulated and IL-10 was decreased. However, IL-6 showed no change after PBMCs incubated with rHcSTP-1 protein. Further functional analysis showed that migratory activity of cells, intracellular nitrite production (NO), and apoptotic efficiency of PBMCs were elevated at significant level, whereas the proliferation of goat PBMCs and monocytes-associated major histocompatibility complex (MHC)-I and MHC-II expressions were decreased significantly at concentration-dependent fashion. Our results showed that the HcSTP-1 protein engaged in vital suppressive regulatory roles on host immune cells, which might represent a potential molecular target for controlling H. contortus infection in future.
Collapse
Affiliation(s)
- Muhammad Ehsan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - WenJuan Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Javaid Ali Gadahi
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Muhammad Waqqas Hasan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - MingMin Lu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - YuJian Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - XinChao Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Muhammad Haseeb
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - RuoFeng Yan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - LiXin Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - XiaoKai Song
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - XiangRui Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
25
|
The effect of lipopolysaccharide on the expression level of immunomodulatory and immunostimulatory factors of human amniotic epithelial cells. BMC Res Notes 2018; 11:343. [PMID: 29843819 PMCID: PMC5975661 DOI: 10.1186/s13104-018-3411-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/07/2018] [Indexed: 11/30/2022] Open
Abstract
Objective Human amniotic epithelial cells (hAECs) are a novel source of stem cells and have immunomodulatory effects on both the innate and adoptive immune system. hAECs can differentiate into multiple cell lineages that make them a suitable cell source for regenerative medicine. These cells express multiple toll-like receptors (TLRs) and respond to various TLR ligands. This study aimed to evaluate the effect of lipopolysaccharide (LPS), a TLR4 ligand, on the level of immunomodulatory and immunostimulatory factors of hAECs. Results Our results indicated that LPS had the ability to up-regulate the expression of prostaglandin E2 synthase and transforming growth factor-beta1 in hAECs. However, there was no change in the level of interleukin-1beta, interleukin-6 and interleukin-10 in hAECs when were stimulated with LPS. In addition, we observed tumor necrosis factor-alpha was only expressed at very low level in some of hAECs samples which its expression was independent of the effects of LPS. Electronic supplementary material The online version of this article (10.1186/s13104-018-3411-9) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Wallace CH, Wu BX, Salem M, Ansa-Addo EA, Metelli A, Sun S, Gilkeson G, Shlomchik MJ, Liu B, Li Z. B lymphocytes confer immune tolerance via cell surface GARP-TGF-β complex. JCI Insight 2018; 3:99863. [PMID: 29618665 PMCID: PMC5928869 DOI: 10.1172/jci.insight.99863] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
GARP, a cell surface docking receptor for binding and activating latent TGF-β, is highly expressed by platelets and activated Tregs. While GARP is implicated in immune invasion in cancer, the roles of the GARP-TGF-β axis in systemic autoimmune diseases are unknown. Although B cells do not express GARP at baseline, we found that the GARP-TGF-β complex is induced on activated human and mouse B cells by ligands for multiple TLRs, including TLR4, TLR7, and TLR9. GARP overexpression on B cells inhibited their proliferation, induced IgA class-switching, and dampened T cell-independent antibody production. In contrast, B cell-specific deletion of GARP-encoding gene Lrrc32 in mice led to development of systemic autoimmune diseases spontaneously as well as worsening of pristane-induced lupus-like disease. Canonical TGF-β signaling more readily upregulates GARP in Peyer patch B cells than in splenic B cells. Furthermore, we demonstrated that B cells are required for the induction of oral tolerance of T cell-dependent antigens via GARP. Our studies reveal for the first time to our knowledge that cell surface GARP-TGF-β is an important checkpoint for regulating B cell peripheral tolerance, highlighting a mechanism of autoimmune disease pathogenesis.
Collapse
Affiliation(s)
| | - Bill X. Wu
- Department of Microbiology and Immunology
| | | | | | | | - Shaoli Sun
- Department of Pathology and Laboratory Medicine, and
| | - Gary Gilkeson
- Department of Microbiology and Immunology
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bei Liu
- Department of Microbiology and Immunology
| | - Zihai Li
- Department of Microbiology and Immunology
- First Affiliated Hospital, Zhengzhou University School of Medicine, Zhengzhou, China
| |
Collapse
|
27
|
Safer approaches to therapeutic modulation of TGF-β signaling for respiratory disease. Pharmacol Ther 2018; 187:98-113. [PMID: 29462659 DOI: 10.1016/j.pharmthera.2018.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The transforming growth factor (TGF)-β cytokines play a central role in development and progression of chronic respiratory diseases. TGF-β overexpression in chronic inflammation, remodeling, fibrotic process and susceptibility to viral infection is established in the most prevalent chronic respiratory diseases including asthma, COPD, lung cancer and idiopathic pulmonary fibrosis. Despite the overwhelming burden of respiratory diseases in the world, new pharmacological therapies have been limited in impact. Although TGF-β inhibition as a therapeutic strategy carries great expectations, the constraints in avoiding compromising the beneficial pleiotropic effects of TGF-β, including the anti-proliferative and immune suppressive effects, have limited the development of effective pharmacological modulators. In this review, we focus on the pathways subserving deleterious and beneficial TGF-β effects to identify strategies for selective modulation of more distal signaling pathways that may result in agents with improved safety/efficacy profiles. Adverse effects of TGF-β inhibitors in respiratory clinical trials are comprehensively reviewed, including those of the marketed TGF-β modulators, pirfenidone and nintedanib. Precise modulation of TGF-β signaling may result in new safer therapies for chronic respiratory diseases.
Collapse
|
28
|
Su E, Han X, Jiang G. The Transforming Growth Factor Beta 1/SMAD Signaling Pathway Involved in Human Chronic Myeloid Leukemia. TUMORI JOURNAL 2018; 96:659-66. [DOI: 10.1177/030089161009600503] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factor beta 1 (TGF-β1) is the prototypic member of a large family of structurally related pleiotropic-secretedcytokines. The TGF-β1/SMAD signaling pathway usually participates in a wide range of cellular processes such as growth, proliferation, differentiation and apoptosis. Upon binding onTGF-β1, the dimerized TGF-β type II receptors recruit and phosphorylate the TGF-β type I receptors, which phosphorylate the receptor-regulated SMAD (SMAD2 and SMAD3) presented by the SMAD anchor for receptor activation. The phosphorylated receptor-regulated SMAD form heterologous complexes with the common-mediator SMAD (SMAD4) and subsequently translocate into the nucleus, where they interact with other transcription factors to regulate the expression of target genes. This multi-functional signaling pathway modulated by various elements with complex mechanisms at different levels is also inevitably involved in cancer. We herein present data on the role of the TGF-β1/SMAD signaling pathway in human chronic myeloid leukemia and explain the potent biological effects of TGF-β1 on leukemia cells. The paper is based on a review of articles selected from Cancerline and Medline data bases. The constitutively active tyrosine kinase produced by the specific Bcr-Abl fusion gene on the Philadelphia chromosome can enhance the resistance of malignant cells to TGF-β1-induced growth inhibition and apoptosis, which contributes to enhancement of proteasomal degradation of p27. However, overexpression of the EVI1 gene, which is also caused by Bcr-Abl, can recruit the C-terminal binding protein and histone deacetylase to prevent the MH2 domain on SMAD3. The later is essential for transcription activation on target genes and leads to blockage of the TGF-β1/SMAD signaling pathway. Some studies have indicated that certain therapeutic agents applied in clinical treatment can inhibit proliferation and promote differentiation of leukemia cells by way of modulation of the TGF-β1/SMAD signal pathway. For example, arsenic trioxide can promote specific degradation of the AML1/MDS1/EVI1 oncoprotein and inhibit the proliferation of leukemia cells. However, specific histone deacetylase inhibitors can interrupt the effect of histone deacetylase to alleviate EVI1-mediated suppression of TGF-β1/SMAD signaling. The tyrosine kinase inhibitor in the target therapy of chronic myeloid leukemia can effectively inhibit the tyrosine kinase activity of Bcr-Abl and induce suppression on the TGF-β1/SMAD signaling pathway. The TGF-β1/SMAD signaling pathway plays an important role in chronic myeloid leukemia cells and leads the leukemia cells to growth inhibition, differentiation and apoptosis. The positive influence of the TGF-β1/SMAD signaling pathway in chronic myeloid leukemia is fairly significant, and its potential effects in clinical treatment will bring about definite benefits. Since it is a complex signaling pathway widely involved in many aspects of cellular activities, further study and comprehensive analysis of the TGF-β1/SMAD signaling pathway are imperative and will have a guiding significance in research and clinical applications. It is an exciting area for future research. Free full text available at www.tumorionline.it
Collapse
Affiliation(s)
- Enyu Su
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine
| | - Xiao Han
- Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Guosheng Jiang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine
| |
Collapse
|
29
|
Tso FY, Kossenkov AV, Lidenge SJ, Ngalamika O, Ngowi JR, Mwaiselage J, Wickramasinghe J, Kwon EH, West JT, Lieberman PM, Wood C. RNA-Seq of Kaposi's sarcoma reveals alterations in glucose and lipid metabolism. PLoS Pathog 2018; 14:e1006844. [PMID: 29352292 PMCID: PMC5792027 DOI: 10.1371/journal.ppat.1006844] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/31/2018] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi’s sarcoma (KS). It is endemic in a number of sub-Saharan African countries with infection rate of >50%. The high prevalence of HIV-1 coupled with late presentation of advanced cancer staging make KS the leading cancer in the region with poor prognosis and high mortality. Disease markers and cellular functions associated with KS tumorigenesis remain ill-defined. Several studies have attempted to investigate changes of the gene profile with in vitro infection of monoculture models, which are not likely to reflect the cellular complexity of the in vivo lesion environment. Our approach is to characterize and compare the gene expression profile in KS lesions versus non-cancer tissues from the same individual. Such comparisons could identify pathways critical for KS formation and maintenance. This is the first study that utilized high throughput RNA-seq to characterize the viral and cellular transcriptome in tumor and non-cancer biopsies of African epidemic KS patients. These patients were treated anti-retroviral therapy with undetectable HIV-1 plasma viral load. We found remarkable variability in the viral transcriptome among these patients, with viral latency and immune modulation genes most abundantly expressed. The presence of KSHV also significantly affected the cellular transcriptome profile. Specifically, genes involved in lipid and glucose metabolism disorder pathways were substantially affected. Moreover, infiltration of immune cells into the tumor did not prevent KS formation, suggesting some functional deficits of these cells. Lastly, we found only minimal overlaps between our in vivo cellular transcriptome dataset with those from in vitro studies, reflecting the limitation of in vitro models in representing tumor lesions. These findings could lead to the identification of diagnostic and therapeutic markers for KS, and will provide bases for further mechanistic studies on the functions of both viral and cellular genes that are involved. Kaposi’s sarcoma-associated herpesvirus (KSHV) is endemic in sub-Saharan Africa and cause Kaposi’s sarcoma (KS). KS is one of the most common cancer among HIV-1 patients in this region. Despite anti-retroviral treatment, prognosis for KS is poor with high mortality often due to presentation of late cancer stage. In order to identify biomarkers or therapeutic targets against KS, a better understanding of the viral and cellular genes expression/transcriptome in the tumor will be necessary. We used RNA-seq, a highly efficient method to sequence transcriptome, to characterize and compare the viral and cellular transcriptome in tumor and non-cancer tissues from KS patients. We found that viral genes involved in latency and immune modulation are most commonly expressed among KS patients. Additionally, cellular genes involved in lipid and glucose metabolism disorder pathways are significantly affected by the presence of KSHV. Despite the detection of immune cells in the tumor, it did not prevent the tumor progression, suggesting some level of immune cell dysfunctions in KS patients. Lastly, we found limited overlap of our data, derived from actual KS biopsy, with other cell culture models, suggesting that the complexity of tumor is difficult to be reflected in cell line models.
Collapse
Affiliation(s)
- For Yue Tso
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | | | - Salum J. Lidenge
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Owen Ngalamika
- Dermatology and Venereology section, University Teaching Hospitals, University of Zambia School of Medicine, Lusaka, Zambia
| | - John R. Ngowi
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Eun Hee Kwon
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - John T. West
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Paul M. Lieberman
- Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Charles Wood
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
30
|
Wei Y, Zhang Z, Wang F, Zhou S. Assessment of tumor growth factor-β1 neutralizing antibody in the treatment of allergic rhinitis and asthma. Exp Ther Med 2018; 15:649-656. [PMID: 29399067 PMCID: PMC5772626 DOI: 10.3892/etm.2017.5501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 04/10/2017] [Indexed: 02/06/2023] Open
Abstract
To identify a novel and effective therapy for allergic rhinitis and asthma (ARA), the present study focused on treatment with tumor growth factor (TGF)-β1 neutralizing antibody. In the present study, four medications were administered to mice with ovalbumin-induced allergic inflammation. Allergic symptoms in the lungs and nasal mucosa were evaluated by detecting the secretion of cytokines from helper T cells (Th) in the peripheral blood, nasal lavage fluid and bronchoalveolar lavage fluid using ELISA. Defects in regulatory T (Treg) cells in peripheral blood mononuclear cells were also detected using flow cytometry. Furthermore, the expression of TGF-β1 and activation of Smad2/3 pathways were assessed using immunohistochemical staining, reverse transcription-quantitative polymerase chain reaction, and western blotting. It was observed that TGF-β1 neutralizing antibody inhibited symptoms of inflammation in the upper and lower airways. TGF-β1 neutralizing antibody also restored the Th1/Th2 balance and ameliorated Treg cell defects induced by ARA. Furthermore, the therapeutic effects of TGF-β1 neutralizing antibody were related to its inhibitory effects on TGF-β1 expression and Smad2/3 signaling in nasal and lung tissues. Therefore, TGF-β1 neutralizing antibody may be an effective medicine for the treatment of ARA.
Collapse
Affiliation(s)
- Yan Wei
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of The Medical School of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Zhili Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of The Medical School of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Feng Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of The Medical School of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shuihong Zhou
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of The Medical School of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
31
|
Dasgupta S, Jain SK. Protective effects of amniotic fluid in the setting of necrotizing enterocolitis. Pediatr Res 2017; 82:584-595. [PMID: 28609432 DOI: 10.1038/pr.2017.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
Abstract
Necrotizing enterocolitis (NEC) is the most common life threatening condition affecting preterm infants. NEC occurs in 1-5% of all neonatal intensive care admissions and 5-10% of very low birth weight infants. The protective role of human breast milk (BM) has been well established. It has also been shown that amniotic fluid (AF) and BM have many similarities in terms of presence of growth and other immune-modulatory factors. This finding led to the initial hypothesis that AF may exert similar protective effects against the development of NEC, as does BM. Multiple studies have elucidated the presence of growth factors in AF and the protective effect of AF against NEC. Studies have also described possible mechanisms how AF protects against NEC. At present, research in this particular area is extremely active and robust. This review summarizes the various studies looking at the protective effects of AF against the development of NEC. It also provides an insight into future directions, the vast potential of AF as a readily available biologic medium, and the ethical barriers that must be overcome before using AF.
Collapse
Affiliation(s)
- Soham Dasgupta
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Sunil Kumar Jain
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
32
|
Hardenberg JHB, Braun A, Schön MP. A Yin and Yang in Epithelial Immunology: The Roles of the α E(CD103)β 7 Integrin in T Cells. J Invest Dermatol 2017; 138:23-31. [PMID: 28941625 DOI: 10.1016/j.jid.2017.05.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/21/2017] [Accepted: 05/31/2017] [Indexed: 01/22/2023]
Abstract
The proper function(s) of cell-surface receptors is crucial for the regulation of adaptive immune responses. One such receptor is the αE(CD103)β7 integrin, whose history in science is closely linked with the evolution of our knowledge of immune regulation. Initially described as a marker of intraepithelial T-lymphocytes, this leukocyte integrin is now seen as a dynamically regulated receptor involved in the functional differentiation of some cytotoxic T cells as well as regulatory T cells, thus presumably contributing to the fine-tuning of immune reactions in epithelial compartments. In this brief overview, we delineate our current view on αE(CD103)β7 in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Jan-Hendrik B Hardenberg
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg August University, Göttingen, Germany
| | - Andrea Braun
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Germany.
| |
Collapse
|
33
|
Kassambara A, Jourdan M, Bruyer A, Robert N, Pantesco V, Elemento O, Klein B, Moreaux J. Global miRNA expression analysis identifies novel key regulators of plasma cell differentiation and malignant plasma cell. Nucleic Acids Res 2017; 45:5639-5652. [PMID: 28459970 PMCID: PMC5449613 DOI: 10.1093/nar/gkx327] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/14/2017] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that attenuate expression of their mRNA targets. Here, we developed a new method and an R package, to easily infer candidate miRNA–mRNA target interactions that could be functional during a given biological process. Using this method, we described, for the first time, a comprehensive integrated analysis of miRNAs and mRNAs during human normal plasma cell differentiation (PCD). Our results reveal 63 miRNAs with significant temporal changes in their expression during normal PCD. We derived a high-confidence network of 295 target relationships comprising 47 miRNAs and 141 targets. These relationships include new examples of miRNAs that appear to coordinately regulate multiple members of critical pathways associated with PCD. Consistent with this, we have experimentally validated a role for the miRNA-30b/c/d-mediated regulation of key PCD factors (IRF4, PRDM1, ELL2 and ARID3A). Furthermore, we found that 24 PCD stage-specific miRNAs are aberrantly overexpressed in multiple myeloma (MM) tumor plasma cells compared to their normal counterpart, suggesting that MM cells frequently acquired expression changes in miRNAs already undergoing dynamic expression modulation during normal PCD. Altogether, our analysis identifies candidate novel key miRNAs regulating networks of significance for normal PCD and malignant plasma cell biology.
Collapse
Affiliation(s)
- Alboukadel Kassambara
- Department of Biological Hematology, CHRU Montpellier, 34000 Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, 34000 Montpellier, France
| | - Michel Jourdan
- Institute of Human Genetics, CNRS-UPR1142, 34000 Montpellier, France
| | - Angélique Bruyer
- Department of Biological Hematology, CHRU Montpellier, 34000 Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, 34000 Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHRU Montpellier, 34000 Montpellier, France
| | | | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Bernard Klein
- Department of Biological Hematology, CHRU Montpellier, 34000 Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, 34000 Montpellier, France.,University of Montpellier 1, UFR de Médecine, 34000 Montpellier, France
| | - Jérôme Moreaux
- Department of Biological Hematology, CHRU Montpellier, 34000 Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, 34000 Montpellier, France.,University of Montpellier 1, UFR de Médecine, 34000 Montpellier, France
| |
Collapse
|
34
|
Thomas BJ, Kan-O K, Loveland KL, Elias JA, Bardin PG. In the Shadow of Fibrosis: Innate Immune Suppression Mediated by Transforming Growth Factor-β. Am J Respir Cell Mol Biol 2017; 55:759-766. [PMID: 27603223 DOI: 10.1165/rcmb.2016-0248ps] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor-β (TGFB) regulates cell proliferation, differentiation, apoptosis, and matrix homeostasis and is intimately involved in fibrosis. TGFB expression is increased in fibrotic lung diseases, such as idiopathic pulmonary fibrosis, and in chronic inflammatory conditions, such as chronic obstructive pulmonary disease and asthma. In addition to exhibiting profibrotic activities, the protein exhibits profound immune-suppressive actions involving both innate and adaptive responses, but often this aspect of TGFB biology is overlooked. Recent investigations have demonstrated that TGFB causes wide-ranging immune suppression, including blunting of pivotal early innate IFN responses. These activities permit severe virus infections, often followed by secondary bacterial infections, which may last longer, with augmented inflammation, scarring, fibrosis, and loss of lung function. Strategies to oppose TGFB actions or to enhance IFN responses may help ameliorate the detrimental consequences of infection in patients with diseases characterized by TGFB overexpression, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Belinda J Thomas
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Keiko Kan-O
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Kate L Loveland
- 4 Centre of Reproductive Health, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Jack A Elias
- 5 Division of Biology and Medicine, Brown University, Providence, Rhode Island
| | - Philip G Bardin
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| |
Collapse
|
35
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 410] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
36
|
MohanKumar K, Namachivayam K, Ho TT, Torres BA, Ohls RK, Maheshwari A. Cytokines and growth factors in the developing intestine and during necrotizing enterocolitis. Semin Perinatol 2017; 41:52-60. [PMID: 27832931 PMCID: PMC5334139 DOI: 10.1053/j.semperi.2016.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines and growth factors play diverse roles in the uninflamed fetal/neonatal intestinal mucosa and in the development of inflammatory bowel injury during necrotizing enterocolitis (NEC). During gestational development and the early neonatal period, the fetal/premature intestine is exposed to high levels of many "inflammatory" cytokines and growth factors, first via swallowed amniotic fluid in utero and then, after birth, in colostrum and mother's milk. This article reviews the dual, seemingly counter-intuitive roles of cytokines, where these agents play a "trophic" role and promote maturation of the uninflamed mucosa, but can also cause inflammation and promote intestinal injury during NEC.
Collapse
Affiliation(s)
| | | | - Thao T.B. Ho
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL
| | | | - Robin K. Ohls
- Department of Pediatrics, University of New Mexico, Albuquerque, NM
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL; Departments of Molecular Medicine, Morsani College of Medicine, Tampa, FL; Department of Community and Family Health, College of Public Health, University of South Florida, 1 Tampa General Circle, Suite F170, Tampa, FL.
| |
Collapse
|
37
|
Kovalova N, Nault R, Crawford R, Zacharewski TR, Kaminski NE. Comparative analysis of TCDD-induced AhR-mediated gene expression in human, mouse and rat primary B cells. Toxicol Appl Pharmacol 2016; 316:95-106. [PMID: 27913140 DOI: 10.1016/j.taap.2016.11.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 12/21/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental pollutant that activates the aryl hydrocarbon receptor (AhR) resulting in altered gene expression. In vivo, in vitro, and ex vivo studies have demonstrated that B cells are directly impaired by TCDD, and are a sensitive target as evidenced by suppression of antibody responses. The window of sensitivity to TCDD-induced suppression of IgM secretion among mouse, rat and human B cells is similar. Specifically, TCDD must be present within the initial 12h post B cell stimulation, indicating that TCDD disrupts early signaling network(s) necessary for B lymphocyte activation and differentiation. Therefore, we hypothesized that TCDD treatment across three different species (mouse, rat and human) triggers a conserved, B cell-specific mechanism that is involved in TCDD-induced immunosuppression. RNA sequencing (RNA-Seq) was used to identify B cell-specific orthologous genes that are differentially expressed in response to TCDD in primary mouse, rat and human B cells. Time course studies identified TCDD-elicited differential expression of 515 human, 2371 mouse and 712 rat orthologous genes over the 24-h period. 28 orthologs were differentially expressed in response to TCDD in all three species. Overrepresented pathways enriched in all three species included cytokine-cytokine receptor interaction, ECM-receptor interaction, focal adhesion, regulation of actin cytoskeleton and pathways in cancer. Differentially expressed genes functionally associated with cell-cell signaling in humans, immune response in mice, and oxidation reduction in rats. Overall, these results suggest that despite the conservation of the AhR and its signaling mechanism, TCDD elicits species-specific gene expression changes.
Collapse
Affiliation(s)
- Natalia Kovalova
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, Lansing, MI 48824, USA.
| | - Rance Nault
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI 48824, USA; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| | - Robert Crawford
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI 48824, USA.
| | - Timothy R Zacharewski
- Institute for Integrative Toxicology, Michigan State University, Lansing, MI 48824, USA; Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| | - Norbert E Kaminski
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI 48824, USA; Institute for Integrative Toxicology, Michigan State University, Lansing, MI 48824, USA.
| |
Collapse
|
38
|
Tsuboi I, Morimoto K, Hirabayashi Y, Li GX, Aizawa S, Mori KJ, Kanno J, Inoue T. Senescent B Lymphopoiesis is Balanced in Suppressive Homeostasis: Decrease in Interleukin-7 and Transforming Growth Factor-β Levels in Stromal Cells of Senescence-Accelerated Mice. Exp Biol Med (Maywood) 2016; 229:494-502. [PMID: 15169968 DOI: 10.1177/153537020422900607] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The suppression of the B cell population during senescence has been considered to be due to the suppression of interleukin-7 (IL-7) production and responsiveness to IL-7; however, the upregulation of transforming growth factor-β (TGF-β) was found to contribute to B cell suppression. To investigate the mechanism of this suppression based on the interrelationship between IL-7 and TGF-β during senescence, senescence-accelerated mice (SAMs), the mouse model of aging, were used in this study to elucidate the mechanisms of B lymphopoietic suppression during aging. Similar to regular senescent mice, SAMs showed a decrease in the number of IL-7–responding B cell progenitors (i.e., colony-forming unit pre-B [CFU-pre-B] cells in the femoral bone marrow [BM]). A co-culture system of B lymphocytes and stromal cells that the authors established showed a significantly lower number of CFU-pre-B cells harvested when BM cells were co-cultured with senescent stromal cells than when they were co-cultured with young stromal cells. Interestingly, cells harvested from a senescent stroma and those from the control culture without stromal cells were higher in number than those harvested from a young stroma, thereby implying that an altered senescent stromal cell is unable to maintain self-renewal of the stem cell compartment. Because TGF-β is supposed to suppress the proliferative capacity of pro-B/pre-B cells, we added a neutralizing anti-TGF-β antibody to the co-culture system with a pro-B/pre-B cell-rich population to determine whether such suppression may be rescued. However, unexpectedly, any rescue was not observed and the number of CFU-pre-B cells remained unchanged when BM cells were co-cultured with senescent stromal cells compared with the co-culture with young stromal cells, which essentially showed an increase in the number of CFU-pre-B cells (P < 0.001 in 5 μg/ml). Furthermore, TGF-β protein level in the supernatant of cultured senescent stroma cells was evaluated by enzyme-linked immunoabsorbent assay, but surprisingly, it was found that TGF-β concentration was significantly lower than that of cultured young stromal cells. Thus, TGF-β activity was assumed to decline particularly in a senescent stroma, which means a distinct difference between the senescent suppression of B lymphopoiesis and secondary B lymphocytopenia. Concerning proliferative signaling, on the other hand, the level of IL-7 gene expression in cells from freshly isolated BM decreased significantly with age. Therefore, the acceleration of proliferative signaling and the deceleration of suppressive signaling may both be altered and weakened in a senescent stroma (i.e., homeosupression).
Collapse
Affiliation(s)
- Isao Tsuboi
- Division of Cellular and Molecular Toxicology, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Tang JB, Zhou YL, Wu YF, Liu PY, Wang XT. Gene therapy strategies to improve strength and quality of flexor tendon healing. Expert Opin Biol Ther 2016; 16:291-301. [PMID: 26853840 DOI: 10.1517/14712598.2016.1134479] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Rupture of the repair and adhesion around a tendon are two major problems after tendon surgery. Novel biological therapies which enhance healing and reduce adhesions are goals of many investigations. Gene therapy offers a new and promising approach to tackle these difficult problems. In the past decade, we sought to develop methods to augment tendon healing and reduce tendon adhesion through gene therapy. AREAS COVERED This review discusses the methods and results of adeno-associated viral (AAV) type 2 vector gene therapy to increase tendon healing strength and reduce adhesions in a chicken model. Micro-RNA related gene therapy is also discussed. We also developed a controlled release system, which incorporates nanoparticles to deliver micro-RNAs to regulate tendon healing. EXPERT OPINION We obtained promising results of enhancement of tendon healing strength in a chicken model using AAV2-mediated gene transfer. AAV2-mediated micro-RNA transfer also limited adhesions around the tendon. Controlled release systems incorporating nanoparticles have ideally delivered genes to the healing tendons and resulted in a moderate (but incomplete) reduction of adhesions. It remains to be determined what the best doses are and what other factors are in play in adhesion formation. These are two targets in our future investigations.
Collapse
Affiliation(s)
- Jin Bo Tang
- a Department of Hand Surgery , The Hand Surgery Research Center, Affiliated Hospital of Nantong University , Nantong , Jiangsu , China
| | - You Lang Zhou
- a Department of Hand Surgery , The Hand Surgery Research Center, Affiliated Hospital of Nantong University , Nantong , Jiangsu , China
| | - Ya Fang Wu
- a Department of Hand Surgery , The Hand Surgery Research Center, Affiliated Hospital of Nantong University , Nantong , Jiangsu , China
| | - Paul Y Liu
- b Department of Plastic Surgery, Rhode Island Hospital , The Alpert Medical School of Brown University , Providence , RI , USA
| | - Xiao Tian Wang
- b Department of Plastic Surgery, Rhode Island Hospital , The Alpert Medical School of Brown University , Providence , RI , USA
| |
Collapse
|
40
|
Alawad A, Altuwaijri S, Aljarbu A, Kryczek I, Niu Y, Al-sobayil FA, Chang C, Bayoumi A, Zou W, Rudat V, Hammad M. Depletion of androgen receptor (AR) in mesenchymal stem cells (MSCs) inhibits induction of CD4+CD25+FOX3+ regulatory T (Treg) cells via androgen TGF-β interaction. J Appl Biomed 2015. [DOI: 10.1016/j.jab.2015.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
41
|
Namachivayam K, Coffing HP, Sankaranarayanan NV, Jin Y, MohanKumar K, Frost BL, Blanco CL, Patel AL, Meier PP, Garzon SA, Desai UR, Maheshwari A. Transforming growth factor-β2 is sequestered in preterm human milk by chondroitin sulfate proteoglycans. Am J Physiol Gastrointest Liver Physiol 2015; 309:G171-80. [PMID: 26045614 PMCID: PMC4525106 DOI: 10.1152/ajpgi.00126.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/22/2015] [Indexed: 01/31/2023]
Abstract
Human milk contains biologically important amounts of transforming growth factor-β2 isoform (TGF-β2), which is presumed to protect against inflammatory gut mucosal injury in the neonate. In preclinical models, enterally administered TGF-β2 can protect against experimental necrotizing enterocolitis, an inflammatory bowel necrosis of premature infants. In this study, we investigated whether TGF-β bioactivity in human preterm milk could be enhanced for therapeutic purposes by adding recombinant TGF-β2 (rTGF-β2) to milk prior to feeding. Milk-borne TGF-β bioactivity was measured by established luciferase reporter assays. Molecular interactions of TGF-β2 were investigated by nondenaturing gel electrophoresis and immunoblots, computational molecular modeling, and affinity capillary electrophoresis. Addition of rTGF-β2 (20-40 nM) to human preterm milk samples failed to increase TGF-β bioactivity in milk. Milk-borne TGF-β2 was bound to chondroitin sulfate (CS) containing proteoglycan(s) such as biglycan, which are expressed in high concentrations in milk. Chondroitinase treatment of milk increased the bioactivity of both endogenous and rTGF-β2, and consequently, enhanced the ability of preterm milk to suppress LPS-induced NF-κB activation in macrophages. These findings provide a mechanism for the normally low bioavailability of milk-borne TGF-β2 and identify chondroitinase digestion of milk as a potential therapeutic strategy to enhance the anti-inflammatory effects of preterm milk.
Collapse
Affiliation(s)
- Kopperuncholan Namachivayam
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| | - Hayley P. Coffing
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,3Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois;
| | - Nehru Viji Sankaranarayanan
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Yingzi Jin
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Krishnan MohanKumar
- 1Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| | - Brandy L. Frost
- 6Department of Pediatrics, NorthShore University Health System, Evanston, Illinois;
| | - Cynthia L. Blanco
- 7Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Aloka L. Patel
- 8Department of Pediatrics, Rush University Medical Center, Chicago, Illinois; and
| | - Paula P. Meier
- 8Department of Pediatrics, Rush University Medical Center, Chicago, Illinois; and ,9Department of Women Children and Family Nursing, Rush University Medical Center, Chicago, Illinois
| | - Steven A. Garzon
- 10Department of Pathology, University of Illinois at Chicago, Chicago, Illinois;
| | - Umesh R. Desai
- 4Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia; ,5Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia;
| | - Akhil Maheshwari
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Pediatrics, Morsani College of Medicine, University of South Florida Health, Tampa, Florida; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Health, Tampa, Florida;
| |
Collapse
|
42
|
All-Trans Retinoic Acid Induces TGF-β2 in Intestinal Epithelial Cells via RhoA- and p38α MAPK-Mediated Activation of the Transcription Factor ATF2. PLoS One 2015. [PMID: 26225425 PMCID: PMC4520553 DOI: 10.1371/journal.pone.0134003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Objective We have shown previously that preterm infants are at risk of necrotizing enterocolitis (NEC), an inflammatory bowel necrosis typically seen in infants born prior to 32 weeks’ gestation, because of the developmental deficiency of transforming growth factor (TGF)-β2 in the intestine. The present study was designed to investigate all-trans retinoic acid (atRA) as an inducer of TGF-β2 in intestinal epithelial cells (IECs) and to elucidate the involved signaling mechanisms. Methods AtRA effects on intestinal epithelium were investigated using IEC6 cells. TGF-β2 expression was measured using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blots. Signaling pathways were investigated using Western blots, transiently-transfected/transduced cells, kinase arrays, chromatin immunoprecipitation, and selective small molecule inhibitors. Results AtRA-treatment of IEC6 cells selectively increased TGF-β2 mRNA and protein expression in a time- and dose-dependent fashion, and increased the activity of the TGF-β2 promoter. AtRA effects were mediated via RhoA GTPase, Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1), p38α MAPK, and activating transcription factor (ATF)-2. AtRA increased phospho-ATF2 binding to the TGF-β2 promoter and increased histone H2B acetylation in the TGF-β2 nucleosome, which is typically associated with transcriptional activation. Conclusions AtRA induces TGF-β2 expression in IECs via RhoA- and p38α MAPK-mediated activation of the transcription factor ATF2. Further studies are needed to investigate the role of atRA as a protective/therapeutic agent in gut mucosal inflammation.
Collapse
|
43
|
Transforming growth factor β type II receptor as a marker in diffuse large B cell lymphoma. Tumour Biol 2015; 36:9903-8. [PMID: 26168957 DOI: 10.1007/s13277-015-3700-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 06/22/2015] [Indexed: 12/22/2022] Open
Abstract
The objective of this study was to investigate the expression and significance of the transforming growth factor β type II receptor (TGFβRII) in diffuse large B cell lymphoma. All patients were enrolled at the First Affiliated Hospital of Liaoning Medical University between 2001 and 2007. The median follow-up period was 53.3 months. Of the 338 patients studied, 131 (38.76 %) had TGFβRII positive expression on immunohistochemistry. The 5 year survival rate was significantly higher in patients with TGFβRII expression than in those without TGFβRII expression (40.3 vs. 31.6 %, P = 0.041). Multivariate analysis identified TGFβRII expression as an independent predictive parameter for survival, in addition to lactate dehydrogenase, clinical stage, and histologic subtype. TGFβRII expression may be considered a new prognostic factor of diffuse large B cell lymphoma.
Collapse
|
44
|
Bailey JC, Iyer AK, Renukaradhya GJ, Lin Y, Nguyen H, Brutkiewicz RR. Inhibition of CD1d-mediated antigen presentation by the transforming growth factor-β/Smad signalling pathway. Immunology 2015; 143:679-91. [PMID: 24990409 DOI: 10.1111/imm.12353] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 06/10/2014] [Accepted: 06/30/2014] [Indexed: 12/14/2022] Open
Abstract
CD1d-mediated lipid antigen presentation activates a subset of innate immune lymphocytes called invariant natural killer T (NKT) cells that, by virtue of their potent cytokine production, bridge the innate and adaptive immune systems. Transforming growth factor (TGF-β) is a known immune modulator that can activate the mitogen-activated protein kinase p38; we have previously shown that p38 is a negative regulator of CD1d-mediated antigen presentation. Several studies implicate a role for TGF-β in the activation of p38. Therefore, we hypothesized that TGF-β would impair antigen presentation by CD1d. Indeed, a dose-dependent decrease in CD1d-mediated antigen presentation and impairment of lipid antigen processing was observed in response to TGF-β treatment. However, it was found that this inhibition was not through p38 activation. Instead, Smads 2, 3 and 4, downstream elements of the TGF-β canonical signalling pathway, contributed to the observed effects. In marked contrast to that observed with CD1d, TGF-β was found to enhance MHC class II-mediated antigen presentation. Overall, these results suggest that the canonical TGF-β/Smad pathway negatively regulates an important arm of the host's innate immune responses - CD1d-mediated lipid antigen presentation to NKT cells.
Collapse
Affiliation(s)
- Jennifer C Bailey
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
45
|
Loomans HA, Andl CD. Intertwining of Activin A and TGFβ Signaling: Dual Roles in Cancer Progression and Cancer Cell Invasion. Cancers (Basel) 2014; 7:70-91. [PMID: 25560921 PMCID: PMC4381251 DOI: 10.3390/cancers7010070] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/23/2014] [Indexed: 12/22/2022] Open
Abstract
In recent years, a significant amount of research has examined the controversial role of activin A in cancer. Activin A, a member of the transforming growth factor β (TGFβ) superfamily, is best characterized for its function during embryogenesis in mesoderm cell fate differentiation and reproduction. During embryogenesis, TGFβ superfamily ligands, TGFβ, bone morphogenic proteins (BMPs) and activins, act as potent morphogens. Similar to TGFβs and BMPs, activin A is a protein that is highly systemically expressed during early embryogenesis; however, post-natal expression is overall reduced and remains under strict spatiotemporal regulation. Of importance, normal post-natal expression of activin A has been implicated in the migration and invasive properties of various immune cell types, as well as endometrial cells. Aberrant activin A signaling during development results in significant morphological defects and premature mortality. Interestingly, activin A has been found to have both oncogenic and tumor suppressor roles in cancer. Investigations into the role of activin A in prostate and breast cancer has demonstrated tumor suppressive effects, while in lung and head and neck squamous cell carcinoma, it has been consistently shown that activin A expression is correlated with increased proliferation, invasion and poor patient prognosis. Activin A signaling is highly context-dependent, which is demonstrated in studies of epithelial cell tumors and the microenvironment. This review discusses normal activin A signaling in comparison to TGFβ and highlights how its dysregulation contributes to cancer progression and cell invasion.
Collapse
Affiliation(s)
- Holli A Loomans
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Claudia D Andl
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
46
|
Mion F, D'Incà F, Danelli L, Toffoletto B, Guarnotta C, Frossi B, Burocchi A, Rigoni A, Gerdes N, Lutgens E, Tripodo C, Colombo MP, Rivera J, Vitale G, Pucillo CE. Mast cells control the expansion and differentiation of IL-10-competent B cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:4568-79. [PMID: 25267976 DOI: 10.4049/jimmunol.1302593] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of B cell subsets with regulatory properties, dependent on IL-10 production, has expanded our view on the mechanisms that control inflammation. Regulatory B cells acquire the ability to produce IL-10 in a stepwise process: first, they become IL-10 competent, a poised state in which B cells are sensitive to trigger signals but do not actually express the Il-10 gene; then, when exposed to appropriate stimuli, they start producing IL-10. Even if the existence of IL-10-competent B cells is now well established, it is not yet known how different immune cell types cross talk with B cells and affect IL-10-competent B cell differentiation and expansion. Mast cells (MCs) contribute to the differentiation and influence the effector functions of various immune cells, including B lymphocytes. In this study, we explored whether MCs could play a role in the expansion of IL-10-competent B cells and addressed the in vivo relevance of MC deficiency on the generation of these cells. We show that MCs can expand IL-10-competent B cells, but they do not directly induce IL-10 production; moreover, the absence of MCs negatively affects IL-10-competent B cell differentiation. Noteworthy, our findings reveal that the CD40L/CD40 axis plays a significant role in MC-driven expansion of IL-10-competent B cells in vitro and highlight the importance of MC CD40L signaling in the colon.
Collapse
Affiliation(s)
- Francesca Mion
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Federica D'Incà
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Luca Danelli
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Barbara Toffoletto
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Carla Guarnotta
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, 90127 Palermo, Italy
| | - Barbara Frossi
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Alessia Burocchi
- Unità di Immunologia Molecolare, Dipartimento di Oncologia Sperimentale e Medicina Molecolare, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, 20133 Milan, Italy
| | - Alice Rigoni
- Unità di Immunologia Molecolare, Dipartimento di Oncologia Sperimentale e Medicina Molecolare, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, 20133 Milan, Italy
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany; Experimental Vascular Biology Laboratory, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, the Netherlands; and
| | - Claudio Tripodo
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, 90127 Palermo, Italy
| | - Mario P Colombo
- Unità di Immunologia Molecolare, Dipartimento di Oncologia Sperimentale e Medicina Molecolare, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, 20133 Milan, Italy
| | - Juan Rivera
- Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Gaetano Vitale
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
| | - Carlo E Pucillo
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy;
| |
Collapse
|
47
|
Caja F, Vannucci L. TGFβ: A player on multiple fronts in the tumor microenvironment. J Immunotoxicol 2014; 12:300-7. [DOI: 10.3109/1547691x.2014.945667] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
48
|
The role(s) of cytokines/chemokines in urinary bladder inflammation and dysfunction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:120525. [PMID: 24738044 PMCID: PMC3971501 DOI: 10.1155/2014/120525] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/26/2014] [Accepted: 02/05/2014] [Indexed: 12/17/2022]
Abstract
Bladder pain syndrome (BPS)/interstitial cystitis (IC) is a chronic pain syndrome characterized by pain, pressure, or discomfort perceived to be bladder related and with at least one urinary symptom. It was recently concluded that 3.3-7.9 million women (>18 years old) in the United States exhibit BPS/IC symptoms. The impact of BPS/IC on quality of life is enormous and the economic burden is significant. Although the etiology and pathogenesis of BPS/IC are unknown, numerous theories including infection, inflammation, autoimmune disorder, toxic urinary agents, urothelial dysfunction, and neurogenic causes have been proposed. Altered visceral sensations from the urinary bladder (i.e., pain at low or moderate bladder filling) that accompany BPS/IC may be mediated by many factors including changes in the properties of peripheral bladder afferent pathways such that bladder afferent neurons respond in an exaggerated manner to normally innocuous stimuli (allodynia). The goals for this review are to describe chemokine/receptor (CXCL12/CXCR4; CCL2/CCR2) signaling and cytokine/receptor (transforming growth factor (TGF-β)/TGF-β type 1 receptor) signaling that may be valuable LUT targets for pharmacologic therapy to improve urinary bladder function and reduce somatic sensitivity associated with urinary bladder inflammation.
Collapse
|
49
|
Abstract
Active, but dysfunctional, immune responses in patients with cancer have been studied in several tumour types, but owing to the heterogeneity of cancer theories of common reaction mechanisms seem to be obsolete. In this Review of published clinical studies of patients with cancer, expression and interplay of the following cytokines are examined: interleukin 2, interleukin 6, interleukin 8, interleukin 10, interleukin 12, interleukin 18, tumour necrosis factor α (TNFα), transforming growth factor β (TGFβ), interferon-γ, HLA-DR, macrophage migration inhibitory factor (MIF), and C-X-C motif chemokine receptor 4 (CXCR4). Clinical data were analysed in a non-quantitative descriptive manner and interpreted with regard to experimentally established physiological cytokine interactions. The clinical cytokine pattern that emerged suggests that simultaneous immunostimulation and immunosuppression occur in patients with cancer, with increased concentrations of the cytokines MIF, TNFα, interleukin 6, interleukin 8, interleukin 10, interleukin 18, and TGFβ. This specific cytokine pattern seems to have a prognostic effect, since high interleukin 6 or interleukin 10 serum concentrations are associated with negative prognoses in independent cancer types. Although immunostimulatory cytokines are involved in local cancer-associated inflammation, cancer cells seem to be protected from immunological eradication by cytokine-mediated local immunosuppression and a resulting defect of the interleukin 12-interferon-γ-HLA-DR axis. Cytokines produced by tumours might have a pivotal role in this defect. A working hypothesis is that the cancer-specific and histology-independent uniform cytokine cascade is one of the manifestations of the underlying paraneoplastic systemic disease, and this hypothesis links the stage of cancer with both the functional status of the immune system and the patient's prognosis. Neutralisation of this cytokine pattern could offer novel and so far unexploited treatment approaches for cancer.
Collapse
|
50
|
Namachivayam K, Blanco CL, Frost BL, Reeves AA, Jagadeeswaran R, MohanKumar K, Safarulla A, Mandal P, Garzon SA, Raj JU, Maheshwari A. Preterm human milk contains a large pool of latent TGF-β, which can be activated by exogenous neuraminidase. Am J Physiol Gastrointest Liver Physiol 2013; 304:G1055-65. [PMID: 23558011 PMCID: PMC3680715 DOI: 10.1152/ajpgi.00039.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human milk contains substantial amounts of transforming growth factor (TGF)-β, particularly the isoform TGF-β2. We previously showed in preclinical models that enterally administered TGF-β2 can protect against necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of premature infants. In this study we hypothesized that premature infants remain at higher risk of NEC than full-term infants, even when they receive their own mother's milk, because preterm human milk contains less bioactive TGF-β than full-term milk. Our objective was to compare TGF-β bioactivity in preterm vs. full-term milk and identify factors that activate milk-borne TGF-β. Mothers who delivered between 23 0/7 and 31 6/7 wk or at ≥37 wk of gestation provided milk samples at serial time points. TGF-β bioactivity and NF-κB signaling were measured using specific reporter cells and in murine intestinal tissue explants. TGF-β1, TGF-β2, TGF-β3, and various TGF-β activators were measured by real-time PCR, enzyme immunoassays, or established enzymatic activity assays. Preterm human milk showed minimal TGF-β bioactivity in the native state but contained a large pool of latent TGF-β. TGF-β2 was the predominant isoform of TGF-β in preterm milk. Using a combination of several in vitro and ex vivo models, we show that neuraminidase is a key regulator of TGF-β bioactivity in human milk. Finally, we show that addition of bacterial neuraminidase to preterm human milk increased TGF-β bioactivity. Preterm milk contains large quantities of TGF-β, but most of it is in an inactive state. Addition of neuraminidase can increase TGF-β bioactivity in preterm milk and enhance its anti-inflammatory effects.
Collapse
Affiliation(s)
- Kopperuncholan Namachivayam
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Cynthia L. Blanco
- 3Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Brandy L. Frost
- 4Department of Pathology, University of Illinois at Chicago, Chicago, Illinois; ,5Department of Pediatrics, NorthShore University Health System, Evanston, Illinois;
| | - Aaron A. Reeves
- 3Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas;
| | - Ramasamy Jagadeeswaran
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Krishnan MohanKumar
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Azif Safarulla
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Partha Mandal
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Steven A. Garzon
- 2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,6University of Chicago Pritzker School of Medicine, Chicago, Illinois; and
| | - J. Usha Raj
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois;
| | - Akhil Maheshwari
- 1Division of Neonatology, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,2Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; ,7Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|