1
|
Guglielmo A, Borghi A, Schettini N, Perillo M, Corazza M, Piraccini BM, Pileri A. Mycosis fungoides and IL-4/13 inhibitors: what is known and unmet needs. Expert Rev Clin Immunol 2025:1-7. [PMID: 40369852 DOI: 10.1080/1744666x.2025.2507332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/22/2025] [Accepted: 05/13/2025] [Indexed: 05/16/2025]
Abstract
INTRODUCTION Dupilumab is a monoclonal antibody that inhibits the IL-4 receptor alpha, preventing the binding of IL-4 and IL-13 and the subsequent signal transduction. Mycosis fungoides (MF) and Sézary syndrome (SS) are the most common form of cutaneous T-cell lymphoma (CTCL). Several cases of MF have been reported following the initiation of dupilumab in patients previously diagnosed with atopic dermatitis. AREAS COVERED This article is a narrative review of the current state of knowledge regarding the correlation between dupilumab and the development of CTCL. Clinical studies on this topic are reviewed, with a particular focus on the pathogenetic theories proposed to date. Finally, we present a new theory, previously undescribed, regarding the potential role of the cytokine microenvironment in triggering CTCL in patients treated with dupilumab. EXPERT OPINION Dupilumab could unmask CTCLs by inhibiting IL-4. In fact, a recent study observed that IL-4 plays a key role in maintaining the 'equilibrium phase' between tumor and microenvironment cells. Disruption of this balance could promote the escape of tumor cells and lead to the unmasking of CTCLs.
Collapse
Affiliation(s)
- Alba Guglielmo
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Borghi
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Natale Schettini
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Monica Corazza
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Bianca Maria Piraccini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Pileri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Ostaszewska A, Michalska Z, Dzierzynska M, Fularczyk M, Bielak K, Morawska A, Kosmala M, Kulig I, Morytz J, Trusiak H, Zimowska M, Rodziewicz-Motowidlo S, Ciemerych MA, Archacka K, Brzoska E. Beneficial but diverse influence of custom-designed hydrogels modified with IL-4 and SDF-1 peptides on selected populations of cells essential for skeletal muscle regeneration. Int J Biol Macromol 2025:144282. [PMID: 40381767 DOI: 10.1016/j.ijbiomac.2025.144282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/28/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Under specific circumstances, such as extensive injuries, the development of degenerative diseases, or aging, the naïve potential of skeletal muscle to regenerate may be limited. For this reason, different tools and approaches are being tested which could result in the improvement of skeletal muscle reconstruction. Among them are hydrogels, investigated also by us, additionally functionalized with fragments of proteins known to support skeletal muscle regeneration, i.e., stromal-derived factor 1 or interleukin 4. In the current study, we evaluated the impact of such custom-designed hydrogels on different human cells important for efficient muscle regeneration, i.e., myoblasts, crucial for myofiber reconstruction, fibroblasts, ensuring ECM formation, and endothelial cells, securing new vessel development in regenerated muscles. Our results indicate that hydrogels functionalized with SDF-1 and IL-4 peptides induce beneficial but diverse effects in analyzed cell types, influencing either their proliferation, migration, or differentiation. Most importantly, hydrogels tested by us do not harm analyzed cell types, indicating that in vivo skeletal muscle regeneration might be improved by them.
Collapse
Affiliation(s)
- Anna Ostaszewska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Zuzanna Michalska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Maria Dzierzynska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Kacper Bielak
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Agnieszka Morawska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Magdalena Kosmala
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Izabela Kulig
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Justyna Morytz
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Hanna Trusiak
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Małgorzata Zimowska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | | | - Maria A Ciemerych
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| |
Collapse
|
3
|
Permkam C, Roytrakul S, Phaonakrop N, Suriyaphol G, Tuntivanich N. Comparison of proteomic profiles and biological properties for corneal wound healing of canine amniotic membrane and its extracts. Vet Ophthalmol 2025; 28:593-604. [PMID: 39080879 DOI: 10.1111/vop.13255] [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: 01/03/2024] [Revised: 04/28/2024] [Accepted: 06/20/2024] [Indexed: 05/23/2025]
Abstract
OBJECTIVE The application of canine amniotic membrane (cAM) for corneal reconstruction is widely used in the veterinary field. However, the information on biological properties and alternative forms of cAM for corneal wound healing is limited. This study aimed to investigate the proteomic profiles and corneal wound healing properties of cAM, cAM extract (cAME), and lyophilized cAM extract (cAMX). ANIMAL STUDIED A total number of 14 cAMs were sterilely harvested from healthy full-term puppies and randomly divided into three different forms: cAM (n = 14), cAME (n = 14), and cAMX (n = 14). PROCEDURES Each form of cAMs was subjected to proteomic analysis using label-free liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), followed by bioinformatic analysis. The proteins were classified into properties by comparing them with the literature search on human amniotic membrane (hAM) properties and the effect on corneal wound healing when given topically. RESULTS The analyses identified 8136 proteins in cAM, 8211 proteins in cAME, and 7093 proteins in cAMX. A total number of 100 proteins were matched with proteins in hAM properties and were classified into anti-inflammatory, anti-fibrotic, anti-microbial, anti-angiogenic, promotion of epithelialization, analgesic, and support cell adhesion and growth properties. Furthermore, proteins with corneal wound healing effects were identified in cAME and cAMX. CONCLUSIONS cAM and its extracts contain numerous proteins, including proteins related to corneal wound healing properties. Additionally, cAME and cAMX showed proteins involved in corneal wound healing and their potential benefits for topical use in ophthalmology.
Collapse
Affiliation(s)
- Chompunut Permkam
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Narumon Phaonakrop
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Gunnaporn Suriyaphol
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Nalinee Tuntivanich
- Department of Veterinary Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
4
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
5
|
Pang Y, Li J, Hu H, Ung COL. Genetic associations of prostate cancer in China: a systematic review. BMC Cancer 2025; 25:604. [PMID: 40181298 PMCID: PMC11966891 DOI: 10.1186/s12885-025-13830-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVES In recent years, there has been a notable increase in the incidence and mortality rates of prostate cancer (PCa) in China, highlighting it as a significant public health issue. This study aimed to investigate the genetic association of PCa in China to better inform national disease management and medical resource allocation. METHODS A systematic literature review was conducted using 5 English databases (Web of Science, PubMed, Embase, Cochrane, Scopus) and 1 Chinese database (CNKI) to identify articles published from database inception to October 8, 2022, which reported the genetic associations of PCa in China. RESULTS Of the 11,195 articles retrieved, 41 were included in the review. A total of 116 different polymorphisms (including single nucleotide polymorphisms, deletions, insertions, and repeat lengths) in 58 genes were studied in Chinese populations. Among these, 37 out of 51 polymorphisms in 28 candidate genes such as BIRC5, C2orf43, COX-2, CYR61 (IGFBP10), DNMT1, DNMT3B, EXO1, FOXP4, and 7 unmapped SNPs were found to have either a positive or negative effect on PCa risk. However, 18 variants in 5 genes remain controversial across different studies. Additionally, 23 SNPs in 16 genes were reported to be associated with disease stage, Gleason score, PSA levels, PCa risk, and clinicopathological characteristics of PCa in China. CONCLUSION In Chinese populations, PCa risk and clinical features may result from individual genes, gene-gene interactions, and gene-environment interactions. These findings provide important insights into the relationship between genetic susceptibility and PCa risk in Chinese men.
Collapse
Affiliation(s)
- Yimin Pang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Junjun Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hao Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Carolina Oi Lam Ung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China.
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
6
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2025; 48:295-312. [PMID: 39325360 PMCID: PMC11996958 DOI: 10.1007/s13402-024-00992-0] [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] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
7
|
Wagner M, Nishikawa H, Koyasu S. Reinventing type 2 immunity in cancer. Nature 2025; 637:296-303. [PMID: 39780006 DOI: 10.1038/s41586-024-08194-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025]
Abstract
Our understanding of type 2 immunity has undergone a substantial transformation in recent years, revealing previously unknown functions. Beyond its canonical role in defence against parasitic helminth infections, type 2 immunity safeguards the host through additional mechanisms, including the suppression of excessive type 1 immune responses, regulation of tissue repair and maintenance of adipose tissue homeostasis. However, unlike type 1 immune responses, type 2 immunity is perceived as a potential promoter of tumorigenesis. Emerging evidence challenges this perspective, painting a more nuanced picture in which type 2 immunity might protect against or even actively suppress tumour growth and progression. In this Review, we explore discoveries that highlight the potential of type 2 immunity in reshaping the landscape of cancer immunotherapies.
Collapse
Affiliation(s)
- Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network-PORT Polish Center for Technology Development, Wrocław, Poland.
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Cancer Immunology, Research Institute/EPOC, National Cancer Center, Tokyo, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| |
Collapse
|
8
|
Mannan A, Dhiamn S, Garg N, Singh TG. Pharmacological modulation of Sonic Hedgehog signaling pathways in Angiogenesis: A mechanistic perspective. Dev Biol 2023; 504:58-74. [PMID: 37739118 DOI: 10.1016/j.ydbio.2023.09.009] [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] [Received: 06/01/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
The Sonic hedgehog (SHh) signaling pathway is an imperative operating network that helps in regulates the critical events during the development processes like multicellular embryo growth and patterning. Disruptions in SHh pathway regulation can have severe consequences, including congenital disabilities, stem cell renewal, tissue regeneration, and cancer/tumor growth. Activation of the SHh signal occurs when SHh binds to the receptor complex of Patch (Ptc)-mediated Smoothened (Smo) (Ptc-smo), initiating downstream signaling. This review explores how pharmacological modulation of the SHh pathway affects angiogenesis through canonical and non-canonical pathways. The canonical pathway for angiogenesis involves the activation of angiogenic cytokines such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), placental growth factor (PGF), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), stromal cell-derived factor 1α, transforming growth factor-β1 (TGF-β1), and angiopoietins (Ang-1 and Ang-2), which facilitate the process of angiogenesis. The Non-canonical pathway includes indirect activation of certain pathways like iNOS/Netrin-1/PKC, RhoA/Rock, ERK/MAPK, PI3K/Akt, Wnt/β-catenin, Notch signaling pathway, and so on. This review will provide a better grasp of the mechanistic approach of SHh in mediating angiogenesis, which can aid in the suppression of certain cancer and tumor growths.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sonia Dhiamn
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
9
|
Mieville V, Griffioen AW, Benamran D, Nowak-Sliwinska P. Advanced in vitro models for renal cell carcinoma therapy design. Biochim Biophys Acta Rev Cancer 2023; 1878:188942. [PMID: 37343729 DOI: 10.1016/j.bbcan.2023.188942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Renal cell carcinoma (RCC) and its principal subtype, clear cell RCC, are the most diagnosed kidney cancer. Despite substantial improvement over the last decades, current pharmacological intervention still fails to achieve long-term therapeutic success. RCC is characterized by a high intra- and inter-tumoral heterogeneity and is heavily influenced by the crosstalk of the cells composing the tumor microenvironment, such as cancer-associated fibroblasts, endothelial cells and immune cells. Moreover, multiple physicochemical properties such as pH, interstitial pressure or oxygenation may also play an important role. These elements are often poorly recapitulated in in vitro models used for drug development. This inadequate recapitulation of the tumor is partially responsible for the current lack of an effective and curative treatment. Therefore, there are needs for more complex in vitro or ex vivo drug screening models. In this review, we discuss the current state-of-the-art of RCC models and suggest strategies for their further development.
Collapse
Affiliation(s)
- Valentin Mieville
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland.
| |
Collapse
|
10
|
Li H, He C, Zhu R, Chen FM, Wang L, Leung FP, Tian XY, Tse G, Wong WT. Type 2 cytokines promote angiogenesis in ischemic muscle via endothelial IL-4Rα signaling. Cell Rep 2023; 42:112964. [PMID: 37556326 DOI: 10.1016/j.celrep.2023.112964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/21/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023] Open
Abstract
Peripheral arterial disease (PAD) is one of the leading causes of cardiovascular morbidity and mortality worldwide, yet current trials on therapeutic angiogenesis remain suboptimal. Type 2 immunity is critical for post-ischemic regeneration, but its regulatory role in revascularization is poorly characterized. Here, we show that type 2 cytokines, interleukin-4 (IL-4) and interleukin-13 (IL-13), are the key mediators in post-ischemic angiogenesis. IL-4/IL-13-deficient mice exhibit impaired reperfusion and muscle repair in an experimental model of PAD. We find that deletion of IL-4Rα in the endothelial compartment, rather than the myeloid compartment, leads to remarkable impairment in revascularization. Mechanistically, IL-4/IL-13 promote endothelial cell proliferation, migration, and tube formation via IL-4Rα/STAT6 signaling. Furthermore, attenuated IL-4/IL-13 expression is associated with the angiogenesis deficit in the setting of diabetic PAD, while IL-4/IL-13 treatment rescues this defective regeneration. Our findings reveal the therapeutic potential of type 2 cytokines in treating patients with muscle ischemia.
Collapse
Affiliation(s)
- Huixian Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Chufeng He
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Ruiwen Zhu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Francis M Chen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Lin Wang
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Fung Ping Leung
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Gary Tse
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong 999077, China; Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wing Tak Wong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
11
|
Resan M, Cvejic Z, Pancevski I, Thumann G, Kropp M, Guber I, Ristic D, Vojvodic D, Pajic B. Interleukin 12 in the Acute Phase of the Immune Response after Excimer Laser Treatment. J Clin Med 2023; 12:4472. [PMID: 37445506 DOI: 10.3390/jcm12134472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The aim of the research was to investigate the differences in the concentrations of IL-12, IL-4, IL-10, and IFN-γ in tears after LASIK and PRK procedures. MATERIALS AND METHODS The study included 68 myopic eyes up to -3.0 D refractive spherical equivalent, divided into two groups: Group 1 LASIK (n = 31) and Group 2 PRK (n = 37). Three tear samples were taken from each eye: immediately before the procedure (t0), 1 h after the procedure (t1), and 24 h after the procedure (t2). The concentrations of IL-12p70, IL-4, IL-10, and IFN-γ in the tear samples were determined by flow cytometry. Participants were not taking anti-inflammatory therapy 24 h after the procedure. RESULTS IL-4 levels 1 h after treatment did not differ between LASIK and PRK (p = 0.990), while 24 h after PRK there was a significant decrease in IL-4 levels (p < 0.05), but not after LASIK (p = 0.476). In both the LASIK (p < 0.05) and PRK (p < 0.05) groups, there is an increase in IL-10 concentrations 1 h after treatment, which persists 24 h after LASIK (p < 0.05) but not after PRK (p = 0.081). There is an increase in IL-12p70 concentration 1 h after treatment in both the LASIK (p < 0.001) and PRK groups (p < 0.001). There is also an increase in IL-12p70 concentration 24 h after PRK (p < 0.005), but not after LASIK (p = 0.775). CONCLUSIONS IL-4 concentration shows a significantly higher value in the LASIK group than in the PRK group after 24 h. IL-10 and IL-12p70 levels increase one hour after surgery in both groups. After 24 h, the IL-10 levels remain elevated in the LASIK group, and the IL-12p70 levels remain elevated in the PRK group. Thus, LASIK and PRK procedures show different inflammatory dynamics.
Collapse
Affiliation(s)
- Mirko Resan
- Eye Clinic, Military Medical Academy, 11000 Belgrade, Serbia
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
- Department of Physics, Faculty of Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Zeljka Cvejic
- Department of Physics, Faculty of Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Igor Pancevski
- Eye Clinic, Military Medical Academy, 11000 Belgrade, Serbia
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Gabriele Thumann
- Division of Ophthalmology, Department of Clinical Neurosciences, Geneva University Hospitals, 1205 Geneva, Switzerland
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Martina Kropp
- Division of Ophthalmology, Department of Clinical Neurosciences, Geneva University Hospitals, 1205 Geneva, Switzerland
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Ivo Guber
- Division of Ophthalmology, Department of Clinical Neurosciences, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Dragana Ristic
- Eye Clinic, Military Medical Academy, 11000 Belgrade, Serbia
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Danilo Vojvodic
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
- Department for Clinical and Experimental Immunology, Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia
| | - Bojan Pajic
- Faculty of Medicine of the Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
- Department of Physics, Faculty of Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
- Division of Ophthalmology, Department of Clinical Neurosciences, Geneva University Hospitals, 1205 Geneva, Switzerland
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
- Eye Clinic ORASIS, Swiss Eye Research Foundation, 5734 Reinach, Switzerland
| |
Collapse
|
12
|
Chiu FY, Kvadas RM, Mheidly Z, Shahbandi A, Jackson JG. Could senescence phenotypes strike the balance to promote tumor dormancy? Cancer Metastasis Rev 2023; 42:143-160. [PMID: 36735097 DOI: 10.1007/s10555-023-10089-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
After treatment and surgery, patient tumors can initially respond followed by a rapid relapse, or respond well and seemingly be cured, but then recur years or decades later. The state of surviving cancer cells during the long, undetected period is termed dormancy. By definition, the dormant tumor cells do not proliferate to create a mass that is detectable or symptomatic, but also never die. An intrinsic state and microenvironment that are inhospitable to the tumor would bias toward cell death and complete eradication, while conditions that favor the tumor would enable growth and relapse. In neither case would clinical dormancy be observed. Normal cells and tumor cells can enter a state of cellular senescence after stress such as that caused by cancer therapy. Senescence is characterized by a stable cell cycle arrest mediated by chromatin modifications that cause gene expression changes and a secretory phenotype involving many cytokines and chemokines. Senescent cell phenotypes have been shown to be both tumor promoting and tumor suppressive. The balance of these opposing forces presents an attractive model to explain tumor dormancy: phenotypes of stable arrest and immune suppression could promote survival, while reversible epigenetic programs combined with cytokines and growth factors that promote angiogenesis, survival, and proliferation could initiate the emergence from dormancy. In this review, we examine the phenotypes that have been characterized in different normal and cancer cells made senescent by various stresses and how these might explain the characteristics of tumor dormancy.
Collapse
Affiliation(s)
- Fang-Yen Chiu
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Raegan M Kvadas
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Zeinab Mheidly
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Ashkan Shahbandi
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - James G Jackson
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA.
| |
Collapse
|
13
|
GEINDREAU M, BRUCHARD M, VEGRAN F. Role of Cytokines and Chemokines in Angiogenesis in a Tumor Context. Cancers (Basel) 2022; 14:cancers14102446. [PMID: 35626056 PMCID: PMC9139472 DOI: 10.3390/cancers14102446] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Tumor growth in solid cancers requires adequate nutrient and oxygen supply, provided by blood vessels created by angiogenesis. Numerous studies have demonstrated that this mechanism plays a crucial role in cancer development and appears to be a well-defined hallmark of cancer. This process is carefully regulated, notably by cytokines with pro-angiogenic or anti-angiogenic features. In this review, we will discuss the role of cytokines in the modulation of angiogenesis. In addition, we will summarize the therapeutic approaches based on cytokine modulation and their clinical approval. Abstract During carcinogenesis, tumors set various mechanisms to help support their development. Angiogenesis is a crucial process for cancer development as it drives the creation of blood vessels within the tumor. These newly formed blood vessels insure the supply of oxygen and nutrients to the tumor, helping its growth. The main factors that regulate angiogenesis are the five members of the vascular endothelial growth factor (VEGF) family. Angiogenesis is a hallmark of cancer and has been the target of new therapies this past few years. However, angiogenesis is a complex phenomenon with many redundancy pathways that ensure its maintenance. In this review, we will first describe the consecutive steps forming angiogenesis, as well as its classical regulators. We will then discuss how the cytokines and chemokines present in the tumor microenvironment can induce or block angiogenesis. Finally, we will focus on the therapeutic arsenal targeting angiogenesis in cancer and the challenges they have to overcome.
Collapse
Affiliation(s)
- Mannon GEINDREAU
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
| | - Mélanie BRUCHARD
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
- Centre Georges-François Leclerc, UNICANCER, 21000 Dijon, France
- LipSTIC Labex, 21000 Dijon, France
| | - Frédérique VEGRAN
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
- Centre Georges-François Leclerc, UNICANCER, 21000 Dijon, France
- LipSTIC Labex, 21000 Dijon, France
- Correspondence:
| |
Collapse
|
14
|
A review on inflammation and angiogenesis as key mechanisms involved in the pathogenesis of bovine cystic ovarian disease. Theriogenology 2022; 186:70-85. [DOI: 10.1016/j.theriogenology.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
|
15
|
Robinson I, Lucia GS, Li A, Oberholtzer N, Plante J, Quinn KM, Reuben D, Mehrotra S, Valdebran M. Eosinophils and melanoma: Implications for immunotherapy. Pigment Cell Melanoma Res 2022; 35:192-202. [PMID: 34927354 PMCID: PMC9012984 DOI: 10.1111/pcmr.13025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022]
Abstract
New therapies such as immune checkpoint blockers (ICB) have offered extended survival to patients affected by advanced melanoma. However, ICBs have demonstrated debilitating side effects on the joints, liver, lungs, skin, and gut. Several biomarkers have been identified for their role in predicting which patients better tolerate ICBs. Still, these biomarkers are limited by immunologic and genetic heterogeneity and the complexity of translation into clinical practice. Recent observational studies have suggested eosinophil counts, and serum levels of eosinophil cationic protein are significantly associated with prolonged survival in advanced-stage melanoma. It is likely that eosinophils thereby modulate treatment response through mechanisms yet to be explored. Here, we review the functionality of eosinophils, their oncogenic role in melanoma and discuss how these mechanisms may influence patient response to ICBs and their implications in clinical practice.
Collapse
Affiliation(s)
- India Robinson
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Gabriella Santa Lucia
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Andraia Li
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Nathaniel Oberholtzer
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - John Plante
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Kristen M Quinn
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel Reuben
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Manuel Valdebran
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
16
|
Peluzzo AM, Autieri MV. Challenging the Paradigm: Anti-Inflammatory Interleukins and Angiogenesis. Cells 2022; 11:cells11030587. [PMID: 35159396 PMCID: PMC8834461 DOI: 10.3390/cells11030587] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis is a vital biological process, and neovascularization is essential for the development, wound repair, and perfusion of ischemic tissue. Neovascularization and inflammation are independent biological processes that are linked in response to injury and ischemia. While clear that pro-inflammatory factors drive angiogenesis, the role of anti-inflammatory interleukins in angiogenesis remains less defined. An interleukin with anti-inflammatory yet pro-angiogenic effects would hold great promise as a therapeutic modality to treat many disease states where inflammation needs to be limited, but revascularization and reperfusion still need to be supported. As immune modulators, interleukins can polarize macrophages to a pro-angiogenic and reparative phenotype, which indirectly influences angiogenesis. Interleukins could also potentially directly induce angiogenesis by binding and activating its receptor on endothelial cells. Although a great deal of attention is given to the negative effects of pro-inflammatory interleukins, less is described concerning the potential protective effects of anti-inflammatory interleukins on various disease processes. To focus this review, we will consider IL-4, IL-10, IL-13, IL-19, and IL-33 to be anti-inflammatory interleukins, all of which have recognized immunomodulatory effects. This review will summarize current research concerning anti-inflammatory interleukins as potential drivers of direct and indirect angiogenesis, emphasizing their role in future therapeutics.
Collapse
|
17
|
Payazdan M, Khatami S, Galehdari H, Delfan N, Shafiei M, Heydaran S. The anti-inflammatory effects of sialic acid on the human glia cells by the upregulation of IL-4 and IL-10 genes' expressions. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
18
|
Bell M, Gottschalk S. Engineered Cytokine Signaling to Improve CAR T Cell Effector Function. Front Immunol 2021; 12:684642. [PMID: 34177932 PMCID: PMC8220823 DOI: 10.3389/fimmu.2021.684642] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Adoptive immunotherapy with T cells genetically modified to express chimeric antigen receptors (CARs) is a promising approach to improve outcomes for cancer patients. While CAR T cell therapy is effective for hematological malignancies, there is a need to improve the efficacy of this therapeutic approach for patients with solid tumors and brain tumors. At present, several approaches are being pursued to improve the antitumor activity of CAR T cells including i) targeting multiple antigens, ii) improving T cell expansion/persistence, iii) enhancing homing to tumor sites, and iv) rendering CAR T cells resistant to the immunosuppressive tumor microenvironment (TME). Augmenting signal 3 of T cell activation by transgenic expression of cytokines or engineered cytokine receptors has emerged as a promising strategy since it not only improves CAR T cell expansion/persistence but also their ability to function in the immunosuppressive TME. In this review, we will provide an overview of cytokine biology and highlight genetic approaches that are actively being pursued to augment cytokine signaling in CAR T cells.
Collapse
Affiliation(s)
- Matthew Bell
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
19
|
Lu KH, Goel S. Transforming growth factor β in breast cancer: another new trick for the old dog. Immunol Cell Biol 2020; 99:249-251. [PMID: 33280167 DOI: 10.1111/imcb.12421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 11/27/2022]
Abstract
Two recent papers from the laboratory of Professor Ming Li demonstrate that inhibition of transforming growth factor β specifically in CD4+ T cells can suppress tumor growth through an unanticipated mechanism.
Collapse
Affiliation(s)
- Kun-Hui Lu
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Youngblood R, Flesher CG, Delproposto J, Baker NA, Neeley CK, Li F, Lumeng CN, Shea LD, O'Rourke RW. Regulation of adipose tissue inflammation and systemic metabolism in murine obesity by polymer implants loaded with lentiviral vectors encoding human interleukin-4. Biotechnol Bioeng 2020; 117:3891-3901. [PMID: 32729936 PMCID: PMC8358590 DOI: 10.1002/bit.27523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 11/08/2022]
Abstract
Dysfunctional adipose tissue plays a central role in the pathogenesis of the obesity-related metabolic disease, including type 2 diabetes. Targeting adipose tissue using biopolymer implants is a novel therapeutic approach for metabolic disease. We transplanted porous poly(lactide-co-glycolide) (PLG) implants coated with human interleukin-4 (hIL-4)-expressing lentivirus into epididymal white adipose tissue (eWAT) of mice fed a high-fat diet. Tissue and systemic inflammation and metabolism were studied with flow cytometry, immunohistochemistry, quantitative real-time polymerase chain reaction, adipose tissue histology, and in vivo glucose tolerance testing at 2 and 10 weeks of a high-fat diet. PLG implants carrying hIL-4-expressing lentivirus implanted into epididymal white adipose tissue of mice-regulated adipose tissue inflammation, including increased CD3+ CD4+ T-cell frequency, increased eWAT adipocyte hypertrophy, and decreased FASN and ATGL expression, along with reduced fasting blood glucose levels. These effects were observed in early obesity but were not maintained in established obesity. Local delivery of bioimplants loaded with cytokine-expressing lentivirus vectors to adipose tissue influences tissue inflammation and systemic metabolism in early obesity. Further study will be required to show more durable metabolic effects. These data demonstrate that polymer biomaterials implanted into adipose tissue have the potential to modulate local tissue and systemic inflammation and metabolism.
Collapse
Affiliation(s)
- Richard Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Carmen G Flesher
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jennifer Delproposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nicki A Baker
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Christopher K Neeley
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Fanghua Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
- Graduate Program in Immunology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Robert W O'Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
21
|
Abstract
The immune system uses two distinct defence strategies against infections: microbe-directed pathogen destruction characterized by type 1 immunity1, and host-directed pathogen containment exemplified by type 2 immunity in induction of tissue repair2. Similar to infectious diseases, cancer progresses with self-propagating cancer cells inflicting host-tissue damage. The immunological mechanisms of cancer cell destruction are well defined3-5, but whether immune-mediated cancer cell containment can be induced remains poorly understood. Here we show that depletion of transforming growth factor-β receptor 2 (TGFBR2) in CD4+ T cells, but not CD8+ T cells, halts cancer progression as a result of tissue healing and remodelling of the blood vasculature, causing cancer cell hypoxia and death in distant avascular regions. Notably, the host-directed protective response is dependent on the T helper 2 cytokine interleukin-4 (IL-4), but not the T helper 1 cytokine interferon-γ (IFN-γ). Thus, type 2 immunity can be mobilized as an effective tissue-level defence mechanism against cancer.
Collapse
|
22
|
VanderVeen BN, Murphy EA, Carson JA. The Impact of Immune Cells on the Skeletal Muscle Microenvironment During Cancer Cachexia. Front Physiol 2020; 11:1037. [PMID: 32982782 PMCID: PMC7489038 DOI: 10.3389/fphys.2020.01037] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Progressive weight loss combined with skeletal muscle atrophy, termed cachexia, is a common comorbidity associated with cancer that results in adverse consequences for the patient related to decreased chemotherapy responsiveness and increased mortality. Cachexia's complexity has provided a barrier for developing successful therapies to prevent or treat the condition, since a large number of systemic disruptions that can regulate muscle mass are often present. Furthermore, considerable effort has focused on investigating how tumor derived factors and inflammatory mediators directly signal skeletal muscle to disrupt protein turnover regulation. Currently, there is developing appreciation for understanding how cancer alters skeletal muscle's complex microenvironment and the tightly regulated interactions between multiple cell types. Skeletal muscle microenvironment interactions have established functions in muscle response to regeneration from injury, growth, aging, overload-induced hypertrophy, and exercise. This review explores the growing body of evidence for immune cell modulation of the skeletal muscle microenvironment during cancer-induced muscle wasting. Emphasis is placed on the regulatory network that integrates physiological responses between immune cells with other muscle cell types including satellite cells, fibroblast cells, and endothelial cells to regulate myofiber size and plasticity. The overall goal of this review is to provide an understanding of how different cell types that constitute the muscle microenvironment and their signaling mediators contribute to cancer and chemotherapy-induced muscle wasting.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - James A. Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
23
|
Loh IP, Sherwin T. Is Keratoconus an Inflammatory Disease? The Implication of Inflammatory Pathways. Ocul Immunol Inflamm 2020; 30:246-255. [PMID: 32791016 DOI: 10.1080/09273948.2020.1780271] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Keratoconus is considered a non-inflammatory condition. Recently however, increased proinflammatory cytokines have been detected in the tears of keratoconic patients and clinical and immunohistochemical observations reported infiltration of matured dendritic cells and leukocytes. Our laboratory utilized cytokine antibody arrays to elucidate the inflammatory aspects of keratoconus. METHODS Protein was extracted from 42 corneal buttons (14 keratoconic and 28 non-keratoconic) and incubated with cytokine antibody arrays scanning 120 cytokines. Mann Whitney U test with a p-value of <0.05 was considered significant. RESULTS Pathways for wound healing, neuroprotection, angiogenesis, and inflammation were activated in keratoconic samples with 23 cytokines showing significant elevation. Fifteen were expressed only in keratoconus with 8 cytokines elevated 1.7-42-fold. CONCLUSION This study identified elevated inflammatory pathways covering immune responses in keratoconus. Our results support the evidence for inflammatory pathway activation in keratoconus and a possible redefinition of keratoconus as a chronic inflammatory corneal disease.
Collapse
Affiliation(s)
- I-Ping Loh
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Trevor Sherwin
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Shih HJ, Chen CL, Torng PL. IGFBP3 inhibits angiogenesis through intracellular regulation of THBS1 expression. Am J Cancer Res 2020; 10:1728-1744. [PMID: 32642286 PMCID: PMC7339270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP3) has been postulated to be a mediator of growth suppression signaling. It was shown to function as a suppressor of invasion in epithelial ovarian cancer (EOC). In this study, we identified an angiogenesis inhibitor, thrombospondin-1 (THBS1), which correlated with IGFBP3 expression in EOC cells. After restoring IGFBP3 expression in an EOC cell line using an inducible plasmid, the transfectants showed an increase in IGFBP3 associated with a parallel increase in THBS1. IGFBP3 decreased cell capillary tube formation in HUVECs, which was reversed after anti-THBS1 treatment. IGFBP3 also decreased blood vessel development in chick embryo chorioallantoic membrane (CAM) assay, which was reversed after THBS1 silencing using THBS1 siRNA. Heterotransplantation of IGFBP3 transfectants significantly decreased tumor growth and vascular formation. Luciferase promoter assay illustrated that THBS1 promoter was activated in the presence of both intracellular and extracellular IGFBP3. The signal was stronger in intracellular IGFBP3 expression than that in extracellular IGFBP3 neutralization. In conclusion, we have identified a novel association between IGFBP3 expression and THBS1 elevation, which consequently results in a decrease in angiogenesis. IGFBP3 could activate THBS1 through promoter regulation mainly via an intracellular signaling pathway. Such angiogenesis-regulating ability could be associated with tumor progression and may represent a major function of IGFBP3 as an onco-suppressor in the pathogenesis of ovarian cancer.
Collapse
Affiliation(s)
- Ho-Jun Shih
- Graduate Institute of Clinical Medicine College of Medicine, National Taiwan UniversityTaiwan
| | - Chi-Ling Chen
- Graduate Institute of Clinical Medicine College of Medicine, National Taiwan UniversityTaiwan
- Department of Internal Medicine and Hepatitis Research Center, National Taiwan University College of Medicine and HospitalTaipei, Taiwan
| | - Pao-Ling Torng
- Department of Obstetrics and Gynecology, National Taiwan University HospitalTaipei, Taiwan
- Department of Obstetrics and Gynecology, Hsin-Chu Branch, National Taiwan University HospitalHsin-Chu, Taiwan
| |
Collapse
|
25
|
Ferreira RC, Batista TM, Duarte SS, Silva DKF, Lisboa TMH, Cavalcanti RFP, Leite FC, Mangueira VM, Sousa TKGD, Abrantes RAD, Trindade EOD, Athayde-Filho PFD, Brandão MCR, Medeiros KCDP, Farias DF, Sobral MV. A novel piperine analogue exerts in vivo antitumor effect by inducing oxidative, antiangiogenic and immunomodulatory actions. Biomed Pharmacother 2020; 128:110247. [PMID: 32450524 DOI: 10.1016/j.biopha.2020.110247] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/01/2020] [Accepted: 05/10/2020] [Indexed: 02/08/2023] Open
Abstract
Structural diversity characterizes natural products as prototypes for design of lead compounds. The aim of this study was to synthetize, and to evaluate the toxicity and antitumor action of a new piperine analogue, the butyl 4-(4-nitrobenzoate)-piperinoate (DE-07). Toxicity was evaluated against zebrafish, and in mice (acute and micronucleus assays). To evaluate the DE-07 antitumor activity Ehrlich ascites carcinoma model was used in mice. Angiogenesis, Reactive Oxygen Species (ROS) production and cytokines levels were investigated. Ninety-six hours exposure to DE-07 did not cause morphological or developmental changes in zebrafish embryos and larvae, with estimated LC50 (lethal concentration 50%) higher than 100 μg/mL. On the acute toxicity assay in mice, LD50 (lethal dose 50%) was estimated at around 1000 mg/kg, intraperitoneally (i.p.). DE-07 (300 mg/kg, i.p.) did not induce increase in the number of micronucleated erythrocytes in mice, suggesting no genotoxicity. On Ehrlich tumor model, DE-07 (12.5, 25 or 50 mg/kg, i.p.) induced a significant decrease on cell viability. In addition, there was an increase on ROS production and a decrease in peritumoral microvessels density. Moreover, DE-07 induced an increase of cytokines levels involved in oxidative stress and antiangiogenic effect (IL-1β, TNF-α and IL-4). No significant clinical toxicological effects were recorded in Ehrlich tumor transplanted animals. These data provide evidence that DE-07 presents low toxicity, and antitumor effect via oxidative and antiangiogenic actions by inducing modulation of inflammatory response in the tumor microenvironment.
Collapse
Affiliation(s)
- Rafael Carlos Ferreira
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Tatianne Mota Batista
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Sâmia Sousa Duarte
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Daiana Karla Frade Silva
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Thaís Mangeon Honorato Lisboa
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Raquel Fragoso Pereira Cavalcanti
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Fagner Carvalho Leite
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Vivianne Mendes Mangueira
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Tatyanna Kélvia Gomes de Sousa
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Renata Albuquerque de Abrantes
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | | | | | | | - Karina Carla de Paula Medeiros
- Department of Morphology, Center of Biosciences, Federal University of Rio Grande Do Norte, 59078-970, Rio Grande do Norte, Brazil
| | - Davi Felipe Farias
- Department of Molecular Biology, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil
| | - Marianna Vieira Sobral
- Postgraduate Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, 58051-970, João Pessoa, Paraíba, Brazil.
| |
Collapse
|
26
|
Madu CO, Wang S, Madu CO, Lu Y. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J Cancer 2020; 11:4474-4494. [PMID: 32489466 PMCID: PMC7255381 DOI: 10.7150/jca.44313] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is a significant event in a wide range of healthy and diseased conditions. This process frequently involves vasodilation and an increase in vascular permeability. Numerous players referred to as angiogenic factors, work in tandem to facilitate the outgrowth of endothelial cells (EC) and the consequent vascularity. Conversely, angiogenic factors could also feature in pathological conditions. Angiogenesis is a critical factor in the development of tumors and metastases in numerous cancers. An increased level of angiogenesis is associated with decreased survival in breast cancer patients. Therefore, a good understanding of the angiogenic mechanism holds a promise of providing effective treatments for breast cancer progression, thereby enhancing patients' survival. Disrupting the initiation and progression of this process by targeting angiogenic factors such as vascular endothelial growth factor (Vegf)-one of the most potent member of the VEGF family- or by targeting transcription factors, such as Hypoxia-Inducible Factors (HIFs) that act as angiogenic regulators, have been considered potential treatment options for several types of cancers. The objective of this review is to highlight the mechanism of angiogenesis in diseases, specifically its role in the progression of malignancy in breast cancer, as well as to highlight the undergoing research in the development of angiogenesis-targeting therapies.
Collapse
Affiliation(s)
- Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152. USA
| | - Stephanie Wang
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Chinua O. Madu
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Yi Lu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163. USA
| |
Collapse
|
27
|
Baba T, Miyazaki D, Inata K, Uotani R, Miyake H, Sasaki SI, Shimizu Y, Inoue Y, Nakamura K. Role of IL-4 in bone marrow driven dysregulated angiogenesis and age-related macular degeneration. eLife 2020; 9:54257. [PMID: 32366355 PMCID: PMC7200155 DOI: 10.7554/elife.54257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/19/2020] [Indexed: 12/15/2022] Open
Abstract
Age-associated sterile inflammation can cause dysregulated choroidal neovascularization (CNV) as age-related macular degeneration (AMD). Intraocular fluid screening of 234 AMD patients identified high levels of IL-4. The purpose of this study was to determine the functional role of IL-4 in CNV formation using murine CNV model. Our results indicate that the IL-4/IL-4 receptors (IL4Rs) controlled tube formation and global proangiogenic responses of bone marrow cells. CCR2+ bone marrow cells were recruited to form very early CNV lesions. IL-4 rapidly induces CCL2, which enhances recruitment of CCR2+ bone marrow cells. This in vivo communication, like quorum-sensing, was followed by the induction of IL-4 by the bone marrow cells during the formation of mature CNVs. For CNV development, IL-4 in bone marrow cells are critically required, and IL-4 directly promotes CNV formation mainly by IL-4R. The IL-4/IL-4Rα axis contributes to pathological angiogenesis through communications with bone marrow cells leading to retinal degeneration.
Collapse
Affiliation(s)
- Takashi Baba
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Dai Miyazaki
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kodai Inata
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryu Uotani
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hitomi Miyake
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Shin-Ichi Sasaki
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yumiko Shimizu
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yoshitsugu Inoue
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kazuomi Nakamura
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, Tottori, Japan
| |
Collapse
|
28
|
Biomarkers for the Noninvasive Diagnosis of Endometriosis: State of the Art and Future Perspectives. Int J Mol Sci 2020; 21:ijms21051750. [PMID: 32143439 PMCID: PMC7084761 DOI: 10.3390/ijms21051750] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Early and accurate diagnosis of endometriosis is crucial for the management of this benign, yet debilitating pathology. Despite the advances of modern medicine, there is no common ground regarding the pathophysiology of this disease as it continues to affect the quality of life of millions of women of reproductive age. The lack of specific symptoms often determines a belated diagnosis. The gold standard remains invasive, surgery followed by a histopathological exam. A biomarker or a panel of biomarkers is easy to measure, usually noninvasive, and could benefit the clinician in both diagnosing and monitoring the treatment response. Several studies have advanced the idea of biomarkers for endometriosis, thereby circumventing unnecessary invasive techniques. Our paper aims at harmonizing the results of these studies in the search of promising perspectives on early diagnosis. METHODS We selected the papers from Google Academic, PubMed, and CrossRef and reviewed recent articles from the literature, aiming to evaluate the effectiveness of various putative serum and urinary biomarkers for endometriosis. RESULTS The majority of studies focused on a panel of biomarkers, rather than a single biomarker and were unable to identify a single biomolecule or a panel of biomarkers with sufficient specificity and sensitivity in endometriosis. CONCLUSION Noninvasive biomarkers, proteomics, genomics, and miRNA microarray may aid the diagnosis, but further research on larger datasets along with a better understanding of the pathophysiologic mechanisms are needed.
Collapse
|
29
|
Follicular structures of cows with cystic ovarian disease present altered expression of cytokines. ZYGOTE 2019; 27:285-298. [PMID: 31412964 DOI: 10.1017/s0967199419000285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ovulation is considered an inflammatory, cytokine-mediated event. Cytokines, which are recognized as growth factors with immunoregulatory properties, are involved in many cellular processes at the ovarian level. In this sense, cytokines affect fertility and are involved in the development of different ovarian disorders such as bovine cystic ovarian disease (COD). Because it has been previously demonstrated that ovarian cells represent both sources and targets of cytokines, the aim of this study was to examine the expression of several cytokines, including IL-1β, IL-1RA, IL-1RI, IL-1RII, IL-4 and IL-8, in ovarian follicular structures from cows with spontaneous COD. The protein expression of these cytokines was evaluated by immunohistochemistry. Additionally, IL-1β, IL-4 and IL-8 concentrations in follicular fluid (FF) and serum were determined by enzyme-linked immunosorbent assay (ELISA). In granulosa and theca cells, IL-1RI, IL-1RII, IL-1RA and IL-4 expression levels were higher in cystic follicles than in the control dominant follicles. The serum and FF concentrations of IL-1β and IL-4 showed no differences between groups, whereas IL-8 concentration was detected only in FF of cysts from cows with COD. The FF and serum concentrations of IL-1β and IL-8 showed no significant differences, whereas IL-4 concentration was higher in FF than in serum in both the control and COD groups. These results evidenced an altered expression of cytokines in ovaries of cows with COD that could contribute to the pathogenesis of this disease.
Collapse
|
30
|
Ribatti D. Interleukins as modulators of angiogenesis and anti-angiogenesis in tumors. Cytokine 2019; 118:3-7. [DOI: 10.1016/j.cyto.2018.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/24/2022]
|
31
|
Alterations in IL-4, IL-10 and IFN-γ levels synergistically decrease lipid content and protein expression of FAS and mature SREBP-1 in human sebocytes. Arch Dermatol Res 2019; 311:563-571. [PMID: 31127384 DOI: 10.1007/s00403-019-01932-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 01/26/2023]
Abstract
When anti-acne alternatives from dietary and plant sources are ingested, systemic alterations of interleukin (IL)-4, IL-10, IL-12 and interferon (IFN)-γ, individually or simultaneously, are induced at a 0.1-10.0-fold (×) range of normal physiological concentrations (1×). However, little is known about the effects of these cytokines on excess sebum, a pathophysiological factor of acne development. In this study, human sebocytes were treated with 0.1-10.0× of IL-4, IL-10, IL-12 and IFN-γ for 3 or 5 days to elucidate the effects on lipid content. Treatment with individual cytokines decreased the lipid content at specific concentrations rather than in a concentration-dependent manner. Specifically, 5.0× of IL-4, 5.0× of IFN-γ (5.0IFN), and 0.5×, 5.0× and 10.0× of IL-10 for 3 days, and 0.5× of IL-4 (0.5IL4) for 5 days decreased lipid content to 87.6-93.0% of the control. Treatment with other concentrations of IL-4, IL-10 and IFN-γ, and 0.1-10.0× of IL-12 did not alter lipid content. Combined treatment with 0.5IL4, 5.0IFN and 0.5× of IL-10 for 3 or 5 days decreased the lipid content more than each individual treatment. However, this effect was more evident after 3 days, in parallel with decreased levels of triglycerides, cholesterol esters and free fatty acids, the major lipid compositions of sebocytes, and decreased protein expression of fatty acid synthase (FAS) and mature sterol response element-binding protein-1 (SREBP-1), the lipogenesis-related factors, without altered cell proliferation. We demonstrated that suppressed IL-4 and IL-10 with enhanced IFN-γ synergistically decreased lipid content and protein expression of FAS and mature SREBP-1 in human sebocytes.
Collapse
|
32
|
Dwyer CJ, Knochelmann HM, Smith AS, Wyatt MM, Rangel Rivera GO, Arhontoulis DC, Bartee E, Li Z, Rubinstein MP, Paulos CM. Fueling Cancer Immunotherapy With Common Gamma Chain Cytokines. Front Immunol 2019; 10:263. [PMID: 30842774 PMCID: PMC6391336 DOI: 10.3389/fimmu.2019.00263] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
Adoptive T cell transfer therapy (ACT) using tumor infiltrating lymphocytes or lymphocytes redirected with antigen receptors (CAR or TCR) has revolutionized the field of cancer immunotherapy. Although CAR T cell therapy mediates robust responses in patients with hematological malignancies, this approach has been less effective for treating patients with solid tumors. Additionally, toxicities post T cell infusion highlight the need for safer ACT protocols. Current protocols traditionally expand T lymphocytes isolated from patient tumors or from peripheral blood to large magnitudes in the presence of high dose IL-2 prior to infusion. Unfortunately, this expansion protocol differentiates T cells to a full effector or terminal phenotype in vitro, consequently reducing their long-term survival and antitumor effectiveness in vivo. Post-infusion, T cells face further obstacles limiting their persistence and function within the suppressive tumor microenvironment. Therapeutic manipulation of T cells with common γ chain cytokines, which are critical growth factors for T cells, may be the key to bypass such immunological hurdles. Herein, we discuss the primary functions of the common γ chain cytokines impacting T cell survival and memory and then elaborate on how these distinct cytokines have been used to augment T cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Guillermo O Rangel Rivera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Dimitrios C Arhontoulis
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Eric Bartee
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
33
|
Knochelmann HM, Smith AS, Dwyer CJ, Wyatt MM, Mehrotra S, Paulos CM. CAR T Cells in Solid Tumors: Blueprints for Building Effective Therapies. Front Immunol 2018; 9:1740. [PMID: 30140266 PMCID: PMC6094980 DOI: 10.3389/fimmu.2018.01740] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/13/2018] [Indexed: 01/06/2023] Open
Abstract
Genetic redirection of T lymphocytes with chimeric antigen receptors (CARs) has soared from treating cancers preclinically to FDA approval for hematologic malignancies and commercial-grade production scale in under 30 years. To date, solid tumors are less susceptible to CAR therapies and instead have been treated more successfully with immune checkpoint blockade or tumor-infiltrating lymphocyte therapy. Here, we discuss the current challenges in treating solid tumors with CAR T cells, and the obstacles within the host and tumor microenvironment hindering their efficacy. We present a novel three-pronged approach for enhancing the efficacy of CAR T cells whereby a single infusion product can synergize the power of an optimal CAR construct, a highly potent T cell subset, and rejuvenate the endogenous immune response to conquer therapeutically-resistant solid tumors.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Shikhar Mehrotra
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
34
|
Altered expression of IL-1β, IL-1RI, IL-1RII, IL-1RA and IL-4 could contribute to anovulation and follicular persistence in cattle. Theriogenology 2018; 110:61-73. [DOI: 10.1016/j.theriogenology.2017.12.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/19/2017] [Accepted: 12/31/2017] [Indexed: 12/13/2022]
|
35
|
Germline variants in IL4, MGMT and AKT1 are associated with prostate cancer-specific mortality: An analysis of 12,082 prostate cancer cases. Prostate Cancer Prostatic Dis 2018; 21:228-237. [PMID: 29298992 PMCID: PMC6026113 DOI: 10.1038/s41391-017-0029-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 11/09/2017] [Accepted: 11/20/2017] [Indexed: 02/08/2023]
Abstract
Background Prostate cancer (PCa) is a leading cause of mortality and genetic factors can influence tumour aggressiveness. Several germline variants have been associated with PCa-specific mortality (PCSM), but further replication evidence is needed. Methods Twenty-two previously identified PCSM-associated genetic variants were genotyped in seven PCa cohorts (12,082 patients; 1544 PCa deaths). For each cohort, Cox proportional hazards models were used to calculate hazard ratios and 95% confidence intervals for risk of PCSM associated with each variant. Data were then combined using a meta-analysis approach. Results Fifteen SNPs were associated with PCSM in at least one of the seven cohorts. In the meta-analysis, after adjustment for clinicopathological factors, variants in the MGMT (rs2308327; HR 0.90; p-value = 3.5 × 10−2) and IL4 (rs2070874; HR 1.22; p-value = 1.1 × 10−3) genes were confirmed to be associated with risk of PCSM. In analyses limited to men diagnosed with local or regional stage disease, a variant in AKT1, rs2494750, was also confirmed to be associated with PCSM risk (HR 0.81; p-value = 3.6 × 10−2). Conclusions This meta-analysis confirms the association of three genetic variants with risk of PCSM, providing further evidence that genetic background plays a role in PCa-specific survival. While these variants alone are not sufficient as prognostic biomarkers, these results may provide insights into the biological pathways modulating tumour aggressiveness.
Collapse
|
36
|
Periodically Pulsed Immunotherapy in a Mathematical Model of Tumor, CD4 + T Cells, and Antitumor Cytokine Interactions. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2017; 2017:2906282. [PMID: 29250133 PMCID: PMC5700558 DOI: 10.1155/2017/2906282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 09/15/2017] [Accepted: 10/12/2017] [Indexed: 11/26/2022]
Abstract
Immunotherapy is one of the most recent approaches for controlling and curing malignant tumors. In this paper, we consider a mathematical model of periodically pulsed immunotherapy using CD4+ T cells and an antitumor cytokine. Mathematical analyses are performed to determine the threshold of a successful treatment. The interindividual variability is explored by one-, two-, and three-parameter bifurcation diagrams for a nontreatment case. Numerical simulation conducted in this paper shows that (i) the tumor can be regulated by administering CD4+ T cells alone in a patient with a strong immune system or who has been diagnosed at an early stage, (ii) immunotherapy with a large amount of an antitumor cytokine can boost the immune system to remit or even to suppress tumor cells completely, and (iii) through polytherapy the tumor can be kept at a smaller size with reduced dosages.
Collapse
|
37
|
Garash R, Bajpai A, Marcinkiewicz BM, Spiller KL. Drug delivery strategies to control macrophages for tissue repair and regeneration. Exp Biol Med (Maywood) 2016; 241:1054-63. [PMID: 27190256 PMCID: PMC4950366 DOI: 10.1177/1535370216649444] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue repair and regeneration is a complex process. Our bodies have an excellent capacity to regenerate damaged tissues in many situations. However, tissue healing is impaired in injuries that exceed a critical size or are exacerbated by chronic inflammatory diseases like diabetes. In these instances, biomaterials and drug delivery strategies are often required to facilitate tissue regeneration by providing physical and biochemical cues. Inflammation is the body's response to injury. It is critical for wound healing and biomaterial integration and vascularization, as long as the timing is well controlled. For example, chronic inflammation is well known to impair healing in chronic wounds. In this review, we highlight the importance of a well-controlled inflammatory response, primarily mediated by macrophages in tissue repair and regeneration and discuss various strategies designed to promote regeneration by controlling macrophage behavior. These strategies include temporally controlled delivery of anti-inflammatory drugs, delivery of macrophages as cellular therapy, controlled release of cytokines that modulate macrophage phenotype, and the design of nanoparticles that exploit the inherent phagocytic behavior of macrophages. A clear outcome of this review is that a deeper understanding of the role and timing of complex macrophage phenotypes or activation states is required to fully harness their abilities with drug delivery strategies.
Collapse
Affiliation(s)
- Reham Garash
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Anamika Bajpai
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Brandon M Marcinkiewicz
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Kara L Spiller
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
38
|
Xing Y, Tian Y, Kurosawa T, Matsui S, Touma M, Wu Q, Sugimoto K. Inhibition of blood vessel formation in tumors by IL-18-polarized M1 macrophages. Genes Cells 2016; 21:287-95. [PMID: 26791003 DOI: 10.1111/gtc.12329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/29/2015] [Indexed: 12/16/2022]
Abstract
We previously showed that interleukin (IL)-18 produced by NFSA cells induced the M1 type of macrophages in NFSA tumors, caused the destruction of endothelial cells in vitro and may have resulted in the necrosis of NFSA tumors by enhancing macrophage phagocytosis and cytotoxicity. However, the effect of IL-18 on blood vessel formation in vivo has not been elucidated. MS-K cells do not express il-18, and they form tumors with well-developed blood vessels. Here, we established IL-18-over-expressing MS-K cell clones (MS-K-IL-18) to address the roles of IL-18 in angiogenesis. The over-expression of IL-18 inhibited the proliferation rate of the MS-K-IL-18 cells in vitro and blood vessel formation in the MS-K-IL-18 tumors. Interestingly, CD14-positive cells from the MS-K-IL-18 tumor had up-regulated expression of the M1-type macrophage marker il-6 and down-regulated expression of interferon (ifn)-γ. Furthermore, FACS analysis showed more accumulation of CD11b+/CD80+ M1 macrophages in the MS-K-IL-18 tumors than in the parental MS-K tumor. Moreover, an in vitro coculture assay showed that MS-K-IL-18-conditioned medium (CM) stimulated macrophages to induce the apoptosis of endothelial cells. Cumulatively, our data showed that IL-18 inhibited tumor blood vessel formation in vivo.
Collapse
Affiliation(s)
- Yanjiang Xing
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| | - Yijun Tian
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| | - Takamasa Kurosawa
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| | - Sayaka Matsui
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| | - Maki Touma
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| | - Qiong Wu
- State Key Laboratory of Urban Water Resource and Environment, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Kenkichi Sugimoto
- Department of Cell Science, Faculty of Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan
| |
Collapse
|
39
|
Chen J, Ying XM, Huang XM, Huang P, Yan SC. Association between polymorphisms in selected inflammatory response genes and the risk of prostate cancer. Onco Targets Ther 2016; 9:223-9. [PMID: 26834482 PMCID: PMC4716763 DOI: 10.2147/ott.s91420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation represents an important event which facilitates prostate carcinogenesis. Genetic variations in inflammatory response genes could affect the level and function of the protein products, resulting in the differential prostate cancer risk among carriers of different variants. This study attempted to investigate the association of IL-4 rs2243250, IL-6 rs10499563, IL-8 rs4073, as well as NFKBIA rs2233406 and rs3138053 polymorphisms with prostate cancer risk in the Chinese population. Genotyping of the polymorphisms was performed by using polymerase chain reaction-restriction fragment length polymorphism technique on 439 prostate cancer patients and 524 controls, and the association of each polymorphic genotype with prostate cancer risk was evaluated by using logistic regression analysis based on allele, heterozygous, and homozygous comparison models, with adjustment to age and smoking status. We showed that the C allele of IL-4 rs2243250 polymorphism could increase prostate cancer risk (heterozygous comparison model: odds ratio [OR] =1.434, 95% confidence interval [CI] =1.092–1.881, P=0.009; homozygous comparison model: OR =2.301, 95% CI =1.402–3.775, P=0.001; allele comparison model: OR =1.509, 95% CI =1.228–1.853, P<0.001). On the other hand, the C allele of rs10499563 polymorphism could decrease prostate cancer risk (heterozygous comparison model: OR =0.694, 95% CI =0.525–0.918, P=0.010; homozygous comparison model: OR =0.499, 95% CI =0.269–0.926, P=0.028; allele comparison model: OR =0.692, 95% CI =0.553–0.867, P=0.001). No association was observed for the other polymorphisms. In conclusion, IL-4 rs2243250 and IL-6 rs10499563 polymorphisms could serve as potential predictive biomarkers for prostate cancer risk in the Chinese population.
Collapse
Affiliation(s)
- Jun Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xue-Ming Ying
- Department of Oncology, Jingdezhen City People's Hospital, Jingdezhen, People's Republic of China
| | - Xue-Ming Huang
- Department of Urology, Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Peng Huang
- The Medical School of Nanchang University, School of Public Health, Nanchang, People's Republic of China
| | - Shao-Cong Yan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
40
|
Aziz S, Ahmed SS, Ali A, Khan FA, Zulfiqar G, Iqbal J, Khan AA, Shoaib M. Salivary Immunosuppressive Cytokines IL-10 and IL-13 Are Significantly Elevated in Oral Squamous Cell Carcinoma Patients. Cancer Invest 2015; 33:318-28. [PMID: 26046681 DOI: 10.3109/07357907.2015.1041642] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is considered to be one of the most fatal diseases worldwide, owing to its late diagnosis and lack of availability of established reliable biomarkers. The aim of this study was to highlight the significance of immunosuppressive cytokines as potential biomarkers in OSCC. Whole unstimulated saliva was collected from each individual (30 OSCC patients and 33 age- and gender-matched healthy controls). Immunosuppressive cytokines, including IL-4, IL-10, IL-13, and IL-1RA, were evaluated in each sample using Luminex multianalyte profiling (xMAP) technology on BioPlex instrument. Our results showed that all the studied salivary cytokines were raised in OSCC patients as compared to controls, where IL-10 and IL-13 salivary levels showed statistically significant difference (p = .004 and p = .010, respectively). Mean levels of salivary cytokines in three histologically defined OSCC categories, compared employing one-way ANOVA, showed that salivary levels of IL-1RA were highest in patients having poorly differentiated OSCC tumors as compared to those having moderately and well-differentiated tumors (p = .000 and p = .002, respectively). Among OSCC individuals, duration of smokeless tobacco correlated positively with IL-1RA (p = .036). We conclude that salivary levels of immunosuppressive cytokines, IL-4, IL-10, IL-13, and IL-1RA, could prove to be potential biomarkers of OSCC and can be further investigated as markers of early detection and disease progression.
Collapse
Affiliation(s)
- Salman Aziz
- College of Dentistry, Aljouf University , Saudi Arabia , 1
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Shin K, Kataru RP, Park HJ, Kwon BI, Kim TW, Hong YK, Lee SH. TH2 cells and their cytokines regulate formation and function of lymphatic vessels. Nat Commun 2015; 6:6196. [PMID: 25648335 DOI: 10.1038/ncomms7196] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 01/05/2015] [Indexed: 12/17/2022] Open
Abstract
Lymphatic vessels (LVs) are critical for immune surveillance and involved in the pathogenesis of diverse diseases. LV density is increased during inflammation; however, little is known about how the resolution of LVs is controlled in different inflammatory conditions. Here we show the negative effects of T helper type 2 (TH2) cells and their cytokines on LV formation. IL-4 and IL-13 downregulate essential transcription factors of lymphatic endothelial cells (LECs) and inhibit tube formation. Co-culture of LECs with TH2 cells also inhibits tube formation, but this effect is fully reversed by interleukin (IL)-4 and/or IL-13 neutralization. Furthermore, the in vivo blockade of IL-4 and/or IL-13 in an asthma model not only increases the density but also enhances the function of lung LVs. These results demonstrate an anti-lymphangiogenic function of TH2 cells and their cytokines, suggesting a potential usefulness of IL-4 and/or IL-13 antagonist as therapeutic agents for allergic asthma through expanding LV mediated-enhanced antigen clearance from the inflammatory sites.
Collapse
Affiliation(s)
- Kihyuk Shin
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Raghu P Kataru
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Hyeung Ju Park
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Bo-In Kwon
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Tae Woo Kim
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Young Kwon Hong
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90033, USA
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| |
Collapse
|
42
|
Gallium maltolate inhibits human cutaneous T-cell lymphoma tumor development in mice. J Invest Dermatol 2014; 135:877-884. [PMID: 25371972 DOI: 10.1038/jid.2014.476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 09/22/2014] [Accepted: 10/20/2014] [Indexed: 01/07/2023]
Abstract
Cutaneous T-cell lymphomas (CTCLs) represent a heterogeneous group of non-Hodgkin's lymphoma characterized by an accumulation of malignant CD4 T cells in the skin. The group IIIa metal salt, gallium nitrate, is known to have antineoplastic activity against B-cell lymphoma in humans, but its activity in CTCLs has not been elaborated in detail. Herein, we examined the antineoplastic efficacy of a gallium compound, gallium maltolate (GaM), in vitro and in vivo with murine models of CTCLs. GaM inhibited cell growth and induced apoptosis of cultured CTCL cells. In human CTCL xenograft models, peritumoral injection of GaM limited the growth of CTCL cells, shown by fewer tumor formations, smaller tumor sizes, and decreased neovascularization in tumor microenvironment. To identify key signaling pathways that have a role in GaM-mediated reduction of tumor growth, we analyzed inflammatory cytokines, as well as signal transduction pathways in CTCL cells treated by GaM. IFN-γ-induced chemokines and IL-13 were found to be notably increased in GaM-treated CTCL cells. However, immunosuppressive cytokines, such as IL-10, were decreased with GaM treatment. Interestingly, both oxidative stress and p53 pathways were involved in GaM-induced cytotoxicity. These results warrant further investigation of GaM as a therapeutic agent for CTCLs.
Collapse
|
43
|
Antibody-mediated delivery of interleukin 4 to the neo-vasculature reduces chronic skin inflammation. J Dermatol Sci 2014; 76:96-103. [PMID: 25190364 DOI: 10.1016/j.jdermsci.2014.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/01/2014] [Accepted: 07/27/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND The antibody-mediated delivery of cytokines ("immunocytokines") to sites of pathological angiogenesis represents an attractive strategy for the development of innovative biopharmaceuticals, capable of modulating the activity of the immune system in cancer and in chronic inflammatory conditions. OBJECTIVE Recombinant IL4 has previously been shown to be therapeutically active in patients with psoriasis. The antibody-mediated delivery of this cytokine to sites of chronic skin inflammatory conditions should lead to an improved potency and selectivity, compared to non-targeted IL4. METHODS The therapeutic activity of F8-IL4, a fusion protein of the F8 antibody (specific to the alternatively-spliced EDA domain of fibronectin) with murine IL4, was investigated in three immunocompetent mouse models of skin inflammation: two induced by the TLR7/8 ligand imiquimod (in Balb/c and C57BL/6) and one mediated by the over-expression of VEGF-A. RESULTS The EDA domain of fibronectin, a marker for angiogenesis, is expressed in the inflamed skin in all three models and F8-IL4 selectively localized to inflamed skin lesions following intravenous administration. The F8-IL4 fusion protein mediated a therapeutic benefit, which was superior to the one of a non-targeted version of IL4 and led to increased levels of key regulatory cytokines (including IL5, IL10, IL13, and IL27) in the inflamed skin, while IL2 levels were not affected in all treatment groups. A murine version of etanercept and a murine anti-IL17 antibody were used as positive control in the therapy experiments. CONCLUSION Skin inflammatory lesions can be selectively targeted using anti-EDA antibody-cytokine fusion proteins and the pharmacodelivery of IL4 confers a therapeutic benefit by shifting the cytokine balance.
Collapse
|
44
|
Jiménez-Massa AE, Alonso-Sardón M, Menacho-Miguel JA, Mirón-Canelo JA, González-Sarmiento R. [Genetic polymorphisms and lung cancer risk: a case-control study]. Med Clin (Barc) 2014; 143:97-103. [PMID: 24529400 DOI: 10.1016/j.medcli.2013.07.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 07/18/2013] [Accepted: 07/25/2013] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND OBJECTIVE The smoke fume, principal factor in the development of lung cancer, causes the expression of certain cytokines, including interleukin 4, 6, 8 and 10, which may act by inhibiting apoptosis and interfere cellular repair mechanisms and angiogenesis. To determine the possible relationship between gene polymorphisms of these cytokines and lung cancer. PATIENTS AND METHOD To achieve this objective we designed a case-control study, which included 400 patients who had come to the consultation for rapid diagnosis of lung cancer at the Pneumology Department, University Hospital of Salamanca, and whose main criterion exclusion was the lack of active contact with smoke fume. Patients were divided into 2 groups, each consisting of 200 patients: cases (patients diagnosed with lung cancer) and controls (patients without lung cancer). RESULTS A percentage of 62.8 of men were former smokers at diagnosis compared with 55.5% of women, although the former still had a greater cumulative consumption. Squamous cell carcinoma predominated in diagnosis (48.9% of patients) and more than half were in advanced stages (28.5% in stage iiiB and 25.5% in stage iv). No statistical significance was observed by linking the existence of tumor to the prevalence of any of the analyzed polymorphisms. CONCLUSIONS Polymorphisms in the study did not modify the risk of developing lung cancer.
Collapse
Affiliation(s)
- Ana E Jiménez-Massa
- Servicio de Neumología, Hospital Universitario Río Hortega, Valladolid, España.
| | - Montserrat Alonso-Sardón
- Departamento de Medicina Preventiva, Salud Pública y Microbiología Clínica, Facultad de Medicina, Universidad de Salamanca, Salamanca, España
| | | | - José Antonio Mirón-Canelo
- Departamento de Medicina Preventiva, Salud Pública y Microbiología Clínica, Facultad de Medicina, Universidad de Salamanca, Salamanca, España
| | | |
Collapse
|
45
|
Aderhold C, Grobschmidt GM, Sauter A, Faber A, Hörmann K, Schultz JD. Interleukin 4, interleukin 6 and osteopontin-serological markers of head and neck malignancy in primary diagnostics: A pilot study. Oncol Lett 2014; 8:1112-1118. [PMID: 25120668 PMCID: PMC4114600 DOI: 10.3892/ol.2014.2312] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 04/01/2014] [Indexed: 12/16/2022] Open
Abstract
The progression of head and neck squamous cell carcinoma (HNSCC) is stimulated by various angiogenic peptides and growth factors. A correlation between tumor progression and the secretion of various serological mediators in patients with malignant tumors of the head and neck is of major interest for tumor diagnostics, evaluation of the therapy response and it may predict prognosis by specifying the individual tumor biology. Established chemotherapeutic regimes for head and neck tumors usually consist of platinum-based chemotherapeutic drugs and 5-fluorouracil (5-FU). The present pilot study sought to assess the eligibility of seven serological factors as biomarkers for malignant tumors of the head and neck: Platelet-derived growth factor, vascular endothelial growth factor, epidermal growth factor receptor, osteopontin, granulocyte-colony stimulating factor, interleukin-4 (IL-4) and IL-6. The serum levels of each factor in 20 patients receiving concomitant radiochemotherapy with cisplatin or carboplatin and 5-FU with curative intent were determined prior and subsequent to chemotherapy and were compared with 40 healthy controls. Another aim of the pilot study was to investigate whether the serum of patients showed significant differences in the concentrations of the analyzed factors at the start of concomitant radiochemotherapy compared with the controls, whether those markers indicated a neoplastic process and whether concomitant radiochemotherapy with cisplatin or carboplatin and 5-FU induced significant alterations of concentration compared with pre-therapeutic levels. The included patients were histopathologically diagnosed with HNSCC and the average age was 62.3 years. The serum samples of the patients were obtained during the course of regular pre- and post-chemotherapeutic blood draws one week prior to the start of radiochemotherapy and one week following the completion of chemotherapy. The healthy controls were collected from patients of the Sleep Laboratory of the Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital (Mannheim, Germany) without clinical evidence or laboratory signs of inflammation or history of a malignant disease. The average age was 50.3 years. The serological level of each factor was ascertained by enzyme-linked immunosorbent assay in duplicate. Serum levels of IL-4, IL-6 and osteopontin were significantly increased in patients with HNSCC compared with those in chemotherapy-naive healthy controls. IL-4 and osteopontin showed no significant therapy-associated alterations. Notably, IL-6 levels significantly increased post-therapeutically. Using logistic regression with osteopontin and IL-4, an individual risk-profile for random samples was calculated. IL-4, IL-6 and osteopontin appear to be suitable indicators of the neoplastic process as they are significantly increased in HNSCC patients compared with the control group. With the exception of IL-6, whose levels were in fact increased following therapy, a significant therapy-associated alteration of these factors was missing. Therefore, these serological markers failed to predict the therapy response, but they may be valuable as a screening instrument in primary diagnostics.
Collapse
Affiliation(s)
- Christoph Aderhold
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| | - Guido Manuel Grobschmidt
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| | - Alexander Sauter
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| | - Anne Faber
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| | - Karl Hörmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| | - Johannes David Schultz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty of Mannheim, University of Heidelberg, Mannheim D-68167, Germany
| |
Collapse
|
46
|
Xie M, Hu A, Luo Y, Sun W, Hu X, Tang S. Interleukin-4 and melatonin ameliorate high glucose and interleukin-1β stimulated inflammatory reaction in human retinal endothelial cells and retinal pigment epithelial cells. Mol Vis 2014; 20:921-8. [PMID: 24991184 PMCID: PMC4077596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 07/26/2014] [Indexed: 11/05/2022] Open
Abstract
PURPOSE We aimed to evaluate the effects of two immune regulatory factors, interleukin-4 (IL-4) and melatonin, on several inflammatory mediators that are involved in inflammation and angiogenesis in diabetic retinopathy (DR), in high glucose or interleukin-1β (IL-1β) induced primary human retinal endothelial cells (RECs) and human retinal pigment epithelial (RPE) cells. METHODS Human RECs and RPE cells were cultured in 30 mM D-glucose or 10 ng/ml IL-1β, with or without the presence of 40 ng/ml IL-4 or 100 μM melatonin. The mRNA and protein levels of vascular endothelial growth factor (VEGF), intercellular cell adhesion molecule-1 (ICAM-1), matrix metalloproteinases 2 (MMP2), and matrix metalloproteinases 9 (MMP9) were measured using real-time PCR and enzyme-linked immunosorbent assay (ELISA), respectively. RESULTS High glucose and IL-1β induced the expression of VEGF, ICAM-1, MMP2, and MMP9 in human RECs and RPE cells. IL-4 and melatonin downregulated the expression of VEGF, ICAM-1, MMP2, and MMP9 induced by high glucose and IL-1β. CONCLUSIONS Our results demonstrated that IL-4 and melatonin inhibited inflammation and angiogenesis triggered by high glucose and IL-1β, which suggests that these immune regulatory factors may be of potential therapeutic value in DR.
Collapse
Affiliation(s)
- Manyun Xie
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Andina Hu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yan Luo
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xinqian Hu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, China,Aier Eye Hospital Group, Changsha, China,Aier Research Institute of Ophthalmology, Changsha, China
| |
Collapse
|
47
|
The IL-1RN and IL-4 gene polymorphisms are potential genetic markers of susceptibility to bladder cancer: a case-control study. World J Urol 2014; 33:389-95. [PMID: 24850227 DOI: 10.1007/s00345-014-1323-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 05/12/2014] [Indexed: 10/25/2022] Open
Abstract
PURPOSE We investigated the relationship between the distribution of the IL-1RN, TNF-β and IL-4 polymorphism and the clinical features of bladder cancer. MATERIALS AND METHODS A total of 100 patients with bladder carcinoma and 102 healthy control subjects were enrolled in the study. The IL-1RN, IL-4 and TNF-β gene polymorphisms were identified by PCR restriction fragment length polymorphism-based analysis. Allelic frequencies were compared between patient and the controls. Tumor stage, histopathological grade, tumor size/number and smoking condition were evaluated with IL-1RN, IL-4 and TNF-β gene polymorphisms. RESULTS Allele distribution frequencies of IL-1RN and IL-4 gene polymorphisms were significantly different between patients and control groups. However, allele distribution of TNF-β gene was not statistically significant. There was no difference in allele distribution of the three genes in both groups regarding stage, tumor size, number of tumors and smoking condition. Although allele distribution of IL-4 gene showed significant difference considering histopathological grades in both smoking and total patients group, allele distribution of IL-1RN and TNF-β was not different. CONCLUSION The present research suggests that the IL-1RN and IL-4 gene polymorphisms are potential genetic markers of susceptibility to bladder cancer. In the future, clinical improvements on diagnosis, treatment and prognosis of bladder carcinoma are expected owing to development of more sensitive and specific tests for genetic polymorphisms of cytokines that are effective on inflammation.
Collapse
|
48
|
Abstract
Corneal wound healing is a complex process: its mechanisms and the underlying genetic control are not fully understood. It involves the integrated actions of multiple growth factors, cytokines and proteases produced by epithelial cells, stromal keratocytes, inflammatory cells and lacrimal gland cells. Following an epithelial insult, multiple cytokines are released triggering a cascade of events that leads to repair the epithelial defect and remodelling of the stroma to minimize the loss of transparency and function. In this review, we examine the literature surrounding the genomics of corneal wound healing with respect to the following topics: epithelial and stromal wound healing (including inhibition); corneal neovascularisation; the role of corneal nerves in wound healing; the endothelium; the role of aquaporins and aptamers. We also examine the effect of ectasia on corneal wound healing with regard to keratoconus and following corneal surgery. A better understanding of the cellular and molecular changes that occur during repair of corneal wounds will provide the opportunity to design treatments that selectively modulate key phases of the healing process resulting in scars that more closely resemble normal corneal architecture.
Collapse
Affiliation(s)
- Nick J R Maycock
- Eye Department, Queen Alexandra Hospital, Portsmouth, UKNational Institute for Health Research (NIHR), Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | | |
Collapse
|
49
|
Talaat RM, Salem TA, El-Masry S, Imbarek A, Mokhles M, Abdel-Aziz A. Circulating pro- and anti-angiogenic mediators in patients infected with hepatitis C at different stages of hepatocellular carcinoma. J Med Virol 2014; 86:1120-9. [DOI: 10.1002/jmv.23932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Roba M. Talaat
- Molecular Biology Department; Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University; Sadat City Egypt
| | - Tarek A. Salem
- Molecular Biology Department; Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University; Sadat City Egypt
| | - Samir El-Masry
- Molecular Biology Department; Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University; Sadat City Egypt
| | - Arafat Imbarek
- Molecular Biology Department; Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University; Sadat City Egypt
| | - Mohamed Mokhles
- Medical Biochemistry Department; Medical Division, National Research Center (NRC); Sadat City Egypt
| | - Amal Abdel-Aziz
- Molecular Biology Department; Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University; Sadat City Egypt
| |
Collapse
|
50
|
Şalva E, Turan SO, Kabasakal L, Alan S, Özkan N, Eren F, Akbuğa J. Investigation of the Therapeutic Efficacy of Codelivery of psiRNA–Vascular Endothelial Growth Factor and pIL-4 into Chitosan Nanoparticles in the Breast Tumor Model. J Pharm Sci 2014; 103:785-95. [DOI: 10.1002/jps.23815] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 11/13/2013] [Accepted: 11/20/2013] [Indexed: 01/16/2023]
|