1
|
Christaki F, Ghasemi A, Pantham D, Abouali R, Provera A, Vecchio C, Sheferaw AN, Dianzani C, Sutti S, Rolla R, Sacchetti S, Giacomini L, Dianzani U, Stoppa I. Role of Balanced Involvement of the ICOS/ICOSL/Osteopontin Network in Cutaneous Wound Healing. Int J Mol Sci 2024; 25:12390. [PMID: 39596455 PMCID: PMC11594701 DOI: 10.3390/ijms252212390] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Inducible T-cell costimulator (ICOS, CD278) is a costimulatory receptor primarily expressed by activated T cells. It binds to ICOS ligand (ICOSL, CD275), which is expressed by various immune and non-immune cell types, particularly in inflamed tissues. ICOSL can also bind to osteopontin (OPN), a protein that functions both as a component of the extracellular matrix and as a soluble pro-inflammatory cytokine. Previous studies, including ours, have shown that ICOS and ICOSL play a role in skin wound healing, as mice deficient in either ICOS or ICOSL exhibit delayed healing. The aim of this study was to investigate the involvement of the ICOS/ICOSL/OPN network in skin wound healing by analyzing mice that are single knockouts for ICOS, ICOSL, or OPN, or double knockouts for ICOS/OPN or ICOSL/OPN. Our results showed that wound healing is impaired in all single knockout strains, but not in the two double knockout strains. Cellular and molecular analyses of the wound healing sites revealed that the healing defect in the single knockout strains is associated with reduced neutrophil infiltration and decreased expression of α-SMA (a marker of myofibroblasts), IL-6, TNFα, and VEGF. In contrast, the normalization of wound closure observed in the double knockout strains was primarily linked to increased vessel formation. A local treatment with recombinant ICOS-Fc improved healing in all mouse strains expressing ICOSL, but not in those lacking ICOSL, and led to a local increase in vessel formation and macrophage recruitment, predominantly of the M2 type.
Collapse
Affiliation(s)
- Foteini Christaki
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Amirabbas Ghasemi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Deepika Pantham
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Reza Abouali
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Alessia Provera
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Cristina Vecchio
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Anteneh Nigussie Sheferaw
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, 10124 Turin, Italy;
| | - Salvatore Sutti
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Sara Sacchetti
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Luca Giacomini
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| | - Ian Stoppa
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (F.C.); (A.G.); (D.P.); (R.A.); (A.P.); (C.V.); (A.N.S.); (S.S.); (R.R.); (S.S.); (L.G.); (I.S.)
| |
Collapse
|
2
|
Xu J, Gao Y, Ding Y, Feng Y, Chen J, Zhang S, Song X, Qiao S. Correlation between Tregs and ICOS-induced M2 macrophages polarization in colorectal cancer progression. Front Oncol 2024; 14:1373820. [PMID: 39104717 PMCID: PMC11298335 DOI: 10.3389/fonc.2024.1373820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Objective To explore the mechanism by which Tregs promote the progression of colorectal cancer by inducing tumor-associated macrophages to polarize into M2 type via ICOS. Methods Postoperative pathological tissues and clinical pathological data of 268 colorectal cancer patients who underwent initial surgery were collected. Immunohistochemistry (IHC) was used to detect the expression levels of ICOS, CD163 (a marker for M2 macrophages), and Foxp3 (a marker for Tregs) in cancerous, adjacent non-tumorous, and normal tissues. The relationship of ICOS, M2 macrophages, and Tregs in CRC with clinical pathological characteristics and pre-surgical tumor markers (such as CEA and CA199) was explored. Results The expression levels of M2 macrophages and Tregs increased with tumor progression, while ICOS expression showed a decreasing trend. Compared to adjacent and normal tissues, the expression levels of ICOS, M2 macrophages, and Tregs were higher in CRC tissues. The expression levels of M2 macrophages and Tregs were significantly positively correlated with tumor markers, while ICOS expression was significantly negatively correlated. Conclusion Tumor-associated m2 macrophages induced by Tregs and ICOS participate in the dynamic balance of the colorectal cancer tumor microenvironment, and their interaction affects colorectal carcinogenesis and progression. High levels of ICOS are associated with better long-term survival rates.
Collapse
Affiliation(s)
- Jiaxin Xu
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu Gao
- Computer Teaching and Research Section, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuting Ding
- Department of Ultrasound, Anshan Central Hospital, Anshan, Liaoning, China
| | - Yunpeng Feng
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jie Chen
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shenshen Zhang
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xiaoyu Song
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shifeng Qiao
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
3
|
Raineri D, Abreu H, Vilardo B, Kustrimovic N, Venegoni C, Cappellano G, Chiocchetti A. Deep Flow Cytometry Unveils Distinct Immune Cell Subsets in Inducible T Cell Co-Stimulator Ligand (ICOSL)- and ICOS-Knockout Mice during Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2024; 25:2509. [PMID: 38473756 DOI: 10.3390/ijms25052509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
The inducible T cell co-stimulator ligand (ICOSL), expressed by antigen presenting cells, binds to the inducible T cell co-stimulator (ICOS) on activated T cells. Improper function of the ICOS/ICOSL pathway has been implicated in several autoimmune diseases, including multiple sclerosis (MS). Previous studies showed that ICOS-knockout (KO) mice exhibit severe experimental autoimmune encephalomyelitis (EAE), the animal model of MS, but data on ICOSL deficiency are not available. In our study, we explored the impact of both ICOS and ICOSL deficiencies on MOG35-55 -induced EAE and its associated immune cell dynamics by employing ICOSL-KO and ICOS-KO mice with a C57BL/6J background. During EAE resolution, MOG-driven cytokine levels and the immunophenotype of splenocytes were evaluated by ELISA and multiparametric flow cytometry, respectively. We found that both KO mice exhibited an overlapping and more severe EAE compared to C57BL/6J mice, corroborated by a reduction in memory/regulatory T cell subsets and interleukin (IL-)17 levels. It is noteworthy that an unsupervised analysis showed that ICOSL deficiency modifies the immune response in an original way, by affecting T central and effector memory (TCM, TEM), long-lived CD4+ TEM cells, and macrophages, compared to ICOS-KO and C57BL/6J mice, suggesting a role for other binding partners to ICOSL in EAE development, which deserves further study.
Collapse
Affiliation(s)
- Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Beatrice Vilardo
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Natasa Kustrimovic
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Chiara Venegoni
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
4
|
Khazaei Monfared Y, Mahmoudian M, Zakeri-Milani P, Cecone C, Hayashi T, Ishii KJ, Conde J, Matencio A, Trotta F. Intratumoural Delivery of mRNA Loaded on a Cationic Hyper-Branched Cyclodextrin-Based Polymer Induced an Anti-Tumour Immunological Response in Melanoma. Cancers (Basel) 2023; 15:3748. [PMID: 37509409 PMCID: PMC10378402 DOI: 10.3390/cancers15143748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
mRNA technology has demonstrated potential for use as an effective cancer immunotherapy. However, inefficient in vivo mRNA delivery and the requirements for immune co-stimulation present major hurdles to achieving anti-tumour therapeutic efficacy. Therefore, we used a cationic hyper-branched cyclodextrin-based polymer to increase mRNA delivery in both in vitro and in vivo melanoma cancer. We found that the transfection efficacy of the mRNA-EGFP-loaded Ppoly system was significantly higher than that of lipofectamine and free mRNA in both 2D and 3D melanoma cancer cells; also, this delivery system did not show cytotoxicity. In addition, the biodistribution results revealed time-dependent and significantly higher mEGFP expression in complexes with Ppoly compared to free mRNA. We then checked the anti-tumour effect of intratumourally injected free mRNA-OVA, a foreign antigen, and loaded Ppoly; the results showed a considerable decrease in both tumour size and weight in the group treated with OVA-mRNA in loaded Ppoly compared to other formulations with an efficient adaptive immune response by dramatically increasing most leukocyte subtypes and OVA-specific CD8+ T cells in both the spleen and tumour tissues. Collectively, our findings suggest that the local delivery of cationic cyclodextrin-based polymer complexes containing foreign mRNA antigens might be a good and reliable concept for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Mohammad Mahmoudian
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran
| | - Parvin Zakeri-Milani
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran
| | - Claudio Cecone
- Department of Chemistry, University of Turin, 10125 Turin, Italy
| | - Tomoya Hayashi
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo 113-8654, Japan
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo 113-8654, Japan
| | - João Conde
- ToxOmics, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1099-085 Lisboa, Portugal
| | - Adrián Matencio
- Department of Chemistry, University of Turin, 10125 Turin, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Turin, 10125 Turin, Italy
| |
Collapse
|
5
|
Differential Modulation of Human M1 and M2 Macrophage Activity by ICOS-Mediated ICOSL Triggering. Int J Mol Sci 2023; 24:ijms24032953. [PMID: 36769276 PMCID: PMC9917690 DOI: 10.3390/ijms24032953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Activated T cells express the inducible T-cell co-stimulator (ICOS) that, upon binding to its ubiquitously expressed ligand (ICOSL), regulates the immune response and tissue repair. We sought to determine the effect of ICOS:ICOSL interaction on human M1 and M2 macrophages. M1 and M2 macrophages were polarized from monocyte-derived macrophages, and the effect of a soluble recombinant form of ICOS (ICOS-CH3) was assessed on cytokine production and cell migration. We show that ICOS-CH3 treatment increased the secretion of CCL3 and CCL4 in resting M1 and M2 cells. In LPS-treated M1 cells, ICOS-CH3 inhibited the secretion of TNF-α, IL-6, IL-10 and CCL4, while it increased that of IL-23. In contrast, M2 cells treated with LPS + IL4 displayed enhanced secretion of IL-6, IL-10, CCL3 and CCL4. In CCL7- or osteopontin-treated M1 cells, ICOS-CH3 boosted the migration rate of M1 cells while it decreased that of M2 cells. Finally, β-Pix expression was upregulated in M1 cells and downregulated in M2 cells by treatment with ICOS-CH3. These findings suggest that ICOSL activation modulates the activity of human M1 and M2 cells, thereby eliciting an overall anti-inflammatory effect consistent with its role in promoting tissue repair.
Collapse
|
6
|
Nisin delivery by nanosponges increases its anticancer activity against in-vivo melanoma model. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
7
|
Ding S, Sun Z, Jiang J, Chang X, Shen Y, Gu Y, Liu C. Inducible costimulator ligand (ICOSL) on CD19 + B cells is involved in immunopathological damage of rheumatoid arthritis (RA). Front Immunol 2022; 13:1015831. [PMID: 36405702 PMCID: PMC9666393 DOI: 10.3389/fimmu.2022.1015831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/10/2022] [Indexed: 01/27/2025] Open
Abstract
Inducible costimulator (ICOS) and its ligand (ICOSL) are critical to regulate the immune response in autoimmune diseases. The participation of B lymphocytes exhibits pathogenic potential in the disease process of rheumatoid arthritis (RA). However, the precise role of ICOSL in RA remains unclear. In this study, we aimed to explore the regulatory effects of CD19+ICOSL+ B cells in the pathogenesis of RA. We demonstrated the increased expression of ICOS and ICOSL in patients with RA and collagen-induced arthritis (CIA) mice. The population of CD19+ICOSL+ B-cell subset was significantly correlated with clinicopathological characteristics of RA patients and CIA mice. Adoptive transfer of CD19+ICOSL+ B cells aggravated arthritic progression in CIA mice. Moreover, microarray analysis revealed that CD19+ICOSL+ cells could exert pivotal effect in pathological process of RA. Further blocking of ICOSL significantly inhibited proinflammatory responses and ameliorated arthritic progression. Therefore, CD19+ICOSL+ B-cell subset could be defined as a specific pathogenic cell subpopulation involved in immunopathological damage of RA. Blockade of ICOSL is promising to be a potential new approach for RA therapy.
Collapse
Affiliation(s)
- Sisi Ding
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyong Sun
- Departments of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Juean Jiang
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Chang
- Departments of Rheumatology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cuiping Liu
- Jiangsu Institute of Clinical Immunology & Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
8
|
Electrospun Collagen Scaffold Bio-Functionalized with Recombinant ICOS-Fc: An Advanced Approach to Promote Bone Remodelling. Polymers (Basel) 2022; 14:polym14183780. [PMID: 36145925 PMCID: PMC9503128 DOI: 10.3390/polym14183780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
The treatment of osteoporotic fractures is a severe clinical issue, especially in cases where low support is provided, e.g., pelvis. New treatments aim to stimulate bone formation in compromised scenarios by using multifunctional biomaterials combined with biofabrication techniques to produce 3D structures (scaffolds) that can support bone formation. Bone’s extracellular matrix (ECM) is mainly composed of type I collagen, making this material highly desirable in bone tissue engineering applications, and its bioactivity can be improved by incorporating specific biomolecules. In this work, type I collagen membranes were produced by electrospinning showing a fibre diameter below 200 nm. An optimized one-step strategy allowed to simultaneously crosslink the electrospun membranes and bind ICOS-Fc, a biomolecule able to reversibly inhibit osteoclast activity. The post-treatment did not alter the ECM-like nanostructure of the meshes and the physicochemical properties of collagen. UV-Vis and TGA analyses confirmed both crosslinking and grafting of ICOS-Fc onto the collagen fibres. The preservation of the biological activity of grafted ICOS-Fc was evidenced by the ability to affect the migratory activity of ICOSL-positive cells. The combination of ICOS-Fc with electrospun collagen represents a promising strategy to design multifunctional devices able to boost bone regeneration in osteoporotic fractures.
Collapse
|
9
|
Gigliotti CL, Boggio E, Favero F, Incarnato D, Santoro C, Oliviero S, Rojo JM, Zucchelli S, Persichetti F, Baldanzi G, Dianzani U, Corà D. Specific transcriptional programs differentiate ICOS from CD28 costimulatory signaling in human Naïve CD4+ T cells. Front Immunol 2022; 13:915963. [PMID: 36131938 PMCID: PMC9484324 DOI: 10.3389/fimmu.2022.915963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Costimulatory molecules of the CD28 family play a crucial role in the activation of immune responses in T lymphocytes, complementing and modulating signals originating from the T-cell receptor (TCR) complex. Although distinct functional roles have been demonstrated for each family member, the specific signaling pathways differentiating ICOS- from CD28-mediated costimulation during early T-cell activation are poorly characterized. In the present study, we have performed RNA-Seq-based global transcriptome profiling of anti-CD3-treated naïve CD4+ T cells upon costimulation through either inducible costimulator (ICOS) or CD28, revealing a set of signaling pathways specifically associated with each signal. In particular, we show that CD3/ICOS costimulation plays a major role in pathways related to STAT3 function and osteoarthritis (OA), whereas the CD3/CD28 axis mainly regulates p38 MAPK signaling. Furthermore, we report the activation of distinct immunometabolic pathways, with CD3/ICOS costimulation preferentially targeting glycosaminoglycans (GAGs) and CD3/CD28 regulating mitochondrial respiratory chain and cholesterol biosynthesis. These data suggest that ICOS and CD28 costimulatory signals play distinct roles during the activation of naïve T cells by modulating distinct sets of immunological and immunometabolic genes.
Collapse
Affiliation(s)
- Casimiro Luca Gigliotti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Francesco Favero
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Danny Incarnato
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, Netherlands
| | - Claudio Santoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Salvatore Oliviero
- Dipartimento di Scienze della Vita e Biologia dei Sistemi, Università di Torino, Torino, Italy
- Italian Institute for Genomic Medicine (IIGM), Torino, Italy
| | - Josè Maria Rojo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Silvia Zucchelli
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
| | - Francesca Persichetti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Gianluca Baldanzi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Biochemical Chemistry, “Maggiore della Carità” University Hospital, Novara, Italy
- *Correspondence: Umberto Dianzani,
| | - Davide Corà
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- CAAD - Center for Translational Research on Autoimmune and Allergic Disease, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
10
|
Alves GF, Stoppa I, Aimaretti E, Monge C, Mastrocola R, Porchietto E, Einaudi G, Collotta D, Bertocchi I, Boggio E, Gigliotti CL, Clemente N, Aragno M, Fernandes D, Cifani C, Thiemermann C, Dianzani C, Dianzani U, Collino M. ICOS-Fc as innovative immunomodulatory approach to counteract inflammation and organ injury in sepsis. Front Immunol 2022; 13:992614. [PMID: 36119089 PMCID: PMC9479331 DOI: 10.3389/fimmu.2022.992614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Inducible T cell co-stimulator (ICOS), an immune checkpoint protein expressed on activated T cells and its unique ligand, ICOSL, which is expressed on antigen-presenting cells and non-hematopoietic cells, have been extensively investigated in the immune response. Recent findings showed that a soluble recombinant form of ICOS (ICOS-Fc) can act as an innovative immunomodulatory drug as both antagonist of ICOS and agonist of ICOSL, modulating cytokine release and cell migration to inflamed tissues. Although the ICOS-ICOSL pathway has been poorly investigated in the septic context, a few studies have reported that septic patients have reduced ICOS expression in whole blood and increased serum levels of osteopontin (OPN), that is another ligand of ICOSL. Thus, we investigated the pathological role of the ICOS-ICOSL axis in the context of sepsis and the potential protective effects of its immunomodulation by administering ICOS-Fc in a murine model of sepsis. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in five-month-old male wild-type (WT) C57BL/6, ICOS-/-, ICOSL-/- and OPN-/- mice. One hour after the surgical procedure, either CLP or Sham (control) mice were randomly assigned to receive once ICOS-Fc, F119SICOS-Fc, a mutated form uncapable to bind ICOSL, or vehicle intravenously. Organs and plasma were collected 24 h after surgery for analyses. When compared to Sham mice, WT mice that underwent CLP developed within 24 h a higher clinical severity score, a reduced body temperature, an increase in plasma cytokines (TNF-α, IL-1β, IL-6, IFN-γ and IL-10), liver injury (AST and ALT) and kidney (creatinine and urea) dysfunction. Administration of ICOS-Fc to WT CLP mice reduced all of these abnormalities caused by sepsis. Similar beneficial effects were not seen in CLP-mice treated with F119SICOS-Fc. Treatment of CLP-mice with ICOS-Fc also attenuated the sepsis-induced local activation of FAK, P38 MAPK and NLRP3 inflammasome. ICOS-Fc seemed to act at both sides of the ICOS-ICOSL interaction, as the protective effect was lost in septic knockout mice for the ICOS or ICOSL genes, whereas it was maintained in OPN knockout mice. Collectively, our data show the beneficial effects of pharmacological modulation of the ICOS-ICOSL pathway in counteracting the sepsis-induced inflammation and organ dysfunction.
Collapse
Affiliation(s)
| | - Ian Stoppa
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Eleonora Aimaretti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Chiara Monge
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Raffaella Mastrocola
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Elisa Porchietto
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Giacomo Einaudi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Debora Collotta
- Department of Neurosciences (Rita Levi Montalcini), University of Turin, Turin, Italy
| | - Ilaria Bertocchi
- Department of Neurosciences (Rita Levi Montalcini), University of Turin, Turin, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | | | - Nausicaa Clemente
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Daniel Fernandes
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Massimo Collino
- Department of Neurosciences (Rita Levi Montalcini), University of Turin, Turin, Italy
- *Correspondence: Massimo Collino,
| |
Collapse
|
11
|
Callaghan B, Twigg MS, Baccile N, Van Bogaert INA, Marchant R, Mitchell CA, Banat IM. Microbial sophorolipids inhibit colorectal tumour cell growth in vitro and restore haematocrit in Apc min+/- mice. Appl Microbiol Biotechnol 2022; 106:6003-6016. [PMID: 35965289 PMCID: PMC9467956 DOI: 10.1007/s00253-022-12115-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/28/2022]
Abstract
Abstract
Sophorolipids are glycolipid biosurfactants consisting of a carbohydrate sophorose head with a fatty acid tail and exist in either an acidic or lactonic form. Sophorolipids are gaining interest as potential cancer chemotherapeutics due to their inhibitory effects on a range of tumour cell lines. Currently, most anti-cancer studies reporting the effects of sophorolipids have focused on lactonic preparations with the effects of acidic sophorolipids yet to be elucidated. We produced a 94% pure acidic sophorolipid preparation which proved to be non-toxic to normal human colonic and lung cells. In contrast, we observed a dose-dependent reduction in viability of colorectal cancer lines treated with the same preparation. Acidic sophorolipids induced apoptosis and necrosis, reduced migration, and inhibited colony formation in all cancer cell lines tested. Furthermore, oral administration of 50 mg kg−1 acidic sophorolipids over 70 days to Apcmin+/− mice was well tolerated and resulted in an increased haematocrit, as well as reducing splenic size and red pulp area. Oral feeding did not affect tumour numbers or sizes in this model. This is the first study to show that acidic sophorolipids dose-dependently and specifically reduces colon cancer cell viability in addition to reducing tumour-associated bleeding in the Apcmin+/− mouse model. Key points • Acidic sophorolipids are produced by yeast species such as Starmerella bombicola. • Acidic sophorolipids selectively killed colorectal cells with no effect on healthy gut epithelia. • Acidic sophorolipids reduced tumour-associated gut bleed in a colorectal mouse model. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12115-6.
Collapse
Affiliation(s)
- Breedge Callaghan
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | - Matthew S Twigg
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | - Niki Baccile
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, Collège de France UMR 7574, Chimie de La Matière Condensée de Paris, UMR 7574, 75005, Paris, France
| | - Inge N A Van Bogaert
- Centre for Synthetic Biology, Department of Biotechnology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Roger Marchant
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | | | - Ibrahim M Banat
- School of Biomedical Sciences, Ulster University, Coleraine, BT52 1SA, UK.
| |
Collapse
|
12
|
Stoppa I, Gigliotti CL, Clemente N, Pantham D, Dianzani C, Monge C, Puricelli C, Rolla R, Sutti S, Renò F, Boldorini R, Boggio E, Dianzani U. ICOSL Stimulation by ICOS-Fc Accelerates Cutaneous Wound Healing In Vivo. Int J Mol Sci 2022; 23:ijms23137363. [PMID: 35806368 PMCID: PMC9266942 DOI: 10.3390/ijms23137363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Background: ICOS and its ligand ICOSL are immune receptors whose interaction triggers bidirectional signals that modulate the immune response and tissue repair. Aim: The aim of this study was to assess the in vivo effects of ICOSL triggering by ICOS-Fc, a recombinant soluble form of ICOS, on skin wound healing. Methods: The effect of human ICOS-Fc on wound healing was assessed, in vitro, and, in vivo, by skin wound healing assay using ICOS−/− and ICOSL−/− knockout (KO) mice and NOD-SCID-IL2R null (NSG) mice. Results: We show that, in wild type mice, treatment with ICOS-Fc improves wound healing, promotes angiogenesis, preceded by upregulation of IL-6 and VEGF expression; increases the number of fibroblasts and T cells, whereas it reduces that of neutrophils; and increases the number of M2 vs. M1 macrophages. Fittingly, ICOS-Fc enhanced M2 macrophage migration, while it hampered that of M1 macrophages. ICOS−/− and ICOSL−/− KO, and NSG mice showed delayed wound healing, and treatment with ICOS-Fc improved wound closure in ICOS−/− and NSG mice. Conclusion: These data show that the ICOS/ICOSL network cooperates in tissue repair, and that triggering of ICOSL by ICOS-Fc improves cutaneous wound healing by increasing angiogenesis and recruitment of reparative macrophages.
Collapse
Affiliation(s)
- Ian Stoppa
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- NOVAICOS srls, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- NOVAICOS srls, 28100 Novara, Italy
| | - Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
| | - Deepika Pantham
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- NOVAICOS srls, 28100 Novara, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, 10124 Turin, Italy; (C.D.); (C.M.)
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, 10124 Turin, Italy; (C.D.); (C.M.)
| | - Chiara Puricelli
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- Maggiore della Carità University Hospital, 28100 Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- Maggiore della Carità University Hospital, 28100 Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
| | - Filippo Renò
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
| | - Renzo Boldorini
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- Maggiore della Carità University Hospital, 28100 Novara, Italy
| | - Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- NOVAICOS srls, 28100 Novara, Italy
- Correspondence: ; Tel.: +39-0321660658
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, 28100 Novara, Italy; (I.S.); (C.L.G.); (N.C.); (D.P.); (C.P.); (R.R.); (S.S.); (F.R.); (R.B.); (U.D.)
- Maggiore della Carità University Hospital, 28100 Novara, Italy
| |
Collapse
|
13
|
Kuske M, Haist M, Jung T, Grabbe S, Bros M. Immunomodulatory Properties of Immune Checkpoint Inhibitors-More than Boosting T-Cell Responses? Cancers (Basel) 2022; 14:1710. [PMID: 35406483 PMCID: PMC8996886 DOI: 10.3390/cancers14071710] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
The approval of immune checkpoint inhibitors (ICI) that serve to enhance effector T-cell anti-tumor responses has strongly improved success rates in the treatment of metastatic melanoma and other tumor types. The currently approved ICI constitute monoclonal antibodies blocking cytotoxic T-lymphocyte-associated protein (CTLA)-4 and anti-programmed cell death (PD)-1. By this, the T-cell-inhibitory CTLA-4/CD80/86 and PD-1/PD-1L/2L signaling axes are inhibited. This leads to sustained effector T-cell activity and circumvents the immune evasion of tumor cells, which frequently upregulate PD-L1 expression and modulate immune checkpoint molecule expression on leukocytes. As a result, profound clinical responses are observed in 40-60% of metastatic melanoma patients. Despite the pivotal role of T effector cells for triggering anti-tumor immunity, mounting evidence indicates that ICI efficacy may also be attributable to other cell types than T effector cells. In particular, emerging research has shown that ICI also impacts innate immune cells, such as myeloid cells, natural killer cells and innate lymphoid cells, which may amplify tumoricidal functions beyond triggering T effector cells, and thus improves clinical efficacy. Effects of ICI on non-T cells may additionally explain, in part, the character and extent of adverse effects associated with treatment. Deeper knowledge of these effects is required to further develop ICI treatment in terms of responsiveness of patients to treatment, to overcome resistance to ICI and to alleviate adverse effects. In this review we give an overview into the currently known immunomodulatory effects of ICI treatment in immune cell types other than the T cell compartment.
Collapse
Affiliation(s)
| | | | | | | | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.K.); (M.H.); (T.J.); (S.G.)
| |
Collapse
|
14
|
Raineri D, Cappellano G, Vilardo B, Maione F, Clemente N, Canciani E, Boggio E, Gigliotti CL, Monge C, Dianzani C, Boldorini R, Dianzani U, Chiocchetti A. Inducible T-Cell Costimulator Ligand Plays a Dual Role in Melanoma Metastasis upon Binding to Osteopontin or Inducible T-Cell Costimulator. Biomedicines 2021; 10:biomedicines10010051. [PMID: 35052731 PMCID: PMC8772802 DOI: 10.3390/biomedicines10010051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/23/2022] Open
Abstract
Recently, we demonstrated that inducible T-cell costimulator (ICOS) shares its unique ligand (ICOSL) with osteopontin (OPN), and OPN/ICOSL binding promotes tumor metastasis and angiogenesis in the 4T1 breast cancer model. Literature showed that OPN promotes melanoma metastasis by suppressing T-cell activation and recruiting myeloid suppressor cells (MDSC). On the opposite, ICOS/ICOSL interaction usually sustains an antitumor response. Here, we engineered murine B16F10 melanoma cells, by transfecting or silencing ICOSL. In vitro data showed that loss of ICOSL favors anchorage-independent growth and induces more metastases in vivo, compared to ICOSL expressing cells. To dissect individual roles of the three molecules, we compared data from C57BL/6 with those from OPN-KO, ICOS-KO, and ICOSL-KO mice, missing one partner at a time. We found that OPN produced by the tumor microenvironment (TME) favors the metastasis by interacting with stromal ICOSL. This activity is dominantly inhibited by ICOS expressed on TME by promoting Treg expansion. Importantly, we also show that OPN and ICOSL highly interact in human melanoma metastases compared to primary tumors. Interfering with this binding may be explored in immunotherapy either for nonresponding or patients resistant to conventional therapies.
Collapse
Affiliation(s)
- Davide Raineri
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Beatrice Vilardo
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Federica Maione
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Nausicaa Clemente
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
| | - Elena Canciani
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| | - Elena Boggio
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
| | - Casimiro Luca Gigliotti
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
| | - Chiara Monge
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy; (C.M.); (C.D.)
| | - Chiara Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy; (C.M.); (C.D.)
| | - Renzo Boldorini
- Divisione di Anatomia Patologica, Dipartimento di Scienze della Salute, AOU Maggiore della Carità, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Umberto Dianzani
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Laboratorio di Biochimica Clinica, Dipartimento di Scienze della Salute, AOU Maggiore della Carità, Università del Piemonte Orientale, Corso Mazzini 18, 28100 Novara, Italy
- Correspondence:
| | - Annalisa Chiocchetti
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy; (D.R.); (G.C.); (B.V.); (F.M.); (N.C.); (E.C.); (E.B.); (C.L.G.); (A.C.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
15
|
Boggio E, Gigliotti CL, Moia R, Scotta A, Crespi I, Boggione P, De Paoli L, Deambrogi C, Garzaro M, Vidali M, Chiocchetti A, Stoppa I, Rolla R, Dianzani C, Monge C, Clemente N, Gaidano G, Dianzani U. Inducible T-cell co-stimulator (ICOS) and ICOS ligand are novel players in the multiple-myeloma microenvironment. Br J Haematol 2021; 196:1369-1380. [PMID: 34954822 DOI: 10.1111/bjh.17968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022]
Abstract
The inducible T-cell co-stimulator (ICOS) is a T-cell receptor that, once bound to ICOS ligand (ICOSL) expressed on several cell types including the B-cell lineage, plays a decisive role in adaptive immunity by regulating the interplay between B and T cells. In addition to its immunomodulatory functions, we have shown that ICOS/ICOSL signalling can inhibit the activity of osteoclasts, unveiling a novel mechanism of lymphocyte-bone cells interactions. ICOS and ICOSL can also be found as soluble forms, namely sICOS and sICOSL. Here we show that: (i) levels of sICOS and sICOSL are increased in multiple myeloma (MM) compared to monoclonal gammopathy of undetermined significance and smouldering MM; (ii) levels of sICOS and sICOSL variably correlate with several markers of tumour burden; and (iii) sICOS levels tend to be higher in Durie-Salmon stage II/III versus stage I MM and correlate with overall survival as an independent variable. Moreover, surface ICOS and ICOSL are expressed in both myeloma cells and normal plasma cells, where they probably regulate different functional stages. Finally, ICOSL triggering inhibits the migration of myeloma cell lines in vitro and the growth of ICOSL+ MOPC-21 myeloma cells in vivo. These results suggest that ICOS and ICOSL represent novel markers and therapeutic targets for MM.
Collapse
Affiliation(s)
- Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | | | - Ilaria Crespi
- Maggiore della Carità University Hospital, Novara, Italy
| | - Paola Boggione
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Lorenzo De Paoli
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Clara Deambrogi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Massimiliano Garzaro
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Matteo Vidali
- Maggiore della Carità University Hospital, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Ian Stoppa
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Turin, Italy
| | - Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale, Novara, Italy.,Maggiore della Carità University Hospital, Novara, Italy
| |
Collapse
|
16
|
Ramavath NN, Gadipudi LL, Provera A, Gigliotti LC, Boggio E, Bozzola C, Albano E, Dianzani U, Sutti S. Inducible T-Cell Costimulator Mediates Lymphocyte/Macrophage Interactions During Liver Repair. Front Immunol 2021; 12:786680. [PMID: 34925367 PMCID: PMC8678521 DOI: 10.3389/fimmu.2021.786680] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
The liver capacity to recover from acute liver injury is a critical factor in the development of acute liver failure (ALF) caused by viral infections, ischemia/reperfusion or drug toxicity. Liver healing requires the switching of pro-inflammatory monocyte-derived macrophages(MoMFs) to a reparative phenotype. However, the mechanisms involved are still incompletely characterized. In this study we investigated the contribution of T-lymphocyte/macrophage interaction through the co-stimulatory molecule Inducible T-cell co-stimulator (ICOS; CD278) and its ligand (ICOSL; CD275) in modulating liver repair. The role of ICOS/ICOSL dyad was investigated during the recovery from acute liver damage induced by a single dose of carbon tetrachloride (CCl4). Flow cytometry of non-parenchymal liver cells obtained from CCl4-treated wild-type mice revealed that the recovery from acute liver injury associated with a specific up-regulation of ICOS in CD8+ T-lymphocytes and with an increase in ICOSL expression involving CD11bhigh/F4-80+ hepatic MoMFs. Although ICOS deficiency did not influence the severity of liver damage and the evolution of inflammation, CCl4-treated ICOS knockout (ICOS-/-) mice showed delayed clearance of liver necrosis and increased mortality. These animals were also characterized by a significant reduction of hepatic reparative MoMFs due to an increased rate of cell apoptosis. An impaired liver healing and loss of reparative MoMFs was similarly evident in ICOSL-deficient mice or following CD8+ T-cells ablation in wild-type mice. The loss of reparative MoMFs was prevented by supplementing CCl4-treated ICOS-/- mice with recombinant ICOS (ICOS-Fc) which also stimulated full recovery from liver injury. These data demonstrated that CD8+ T-lymphocytes play a key role in supporting the survival of reparative MoMFs during liver healing trough ICOS/ICOSL-mediated signaling. These observations open the possibility of targeting ICOS/ICOSL dyad as a novel tool for promoting efficient healing following acute liver injury.
Collapse
Affiliation(s)
- Naresh Naik Ramavath
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Laila Lavanya Gadipudi
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Alessia Provera
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Luca C Gigliotti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Cristina Bozzola
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University of East Piedmont, Novara, Italy
| |
Collapse
|
17
|
Battaglia L, Scomparin A, Dianzani C, Milla P, Muntoni E, Arpicco S, Cavalli R. Nanotechnology Addressing Cutaneous Melanoma: The Italian Landscape. Pharmaceutics 2021; 13:1617. [PMID: 34683910 PMCID: PMC8540596 DOI: 10.3390/pharmaceutics13101617] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
Cutaneous melanoma is one of the most aggressive solid tumors, with a low survival for the metastatic stage. Currently, clinical melanoma treatments include surgery, chemotherapy, targeted therapy, immunotherapy and radiotherapy. Of note, innovative therapeutic regimens concern the administration of multitarget drugs in tandem, in order to improve therapeutic efficacy. However, also, if this drug combination is clinically relevant, the patient's response is not yet optimal. In this scenario, nanotechnology-based delivery systems can play a crucial role in the clinical treatment of advanced melanoma. In fact, their nano-features enable targeted drug delivery at a cellular level by overcoming biological barriers. Various nanomedicines have been proposed for the treatment of cutaneous melanoma, and a relevant number of them are undergoing clinical trials. In Italy, researchers are focusing on the pharmaceutical development of nanoformulations for malignant melanoma therapy. The present review reports an overview of the main melanoma-addressed nanomedicines currently under study in Italy, alongside the state of the art of melanoma therapy. Moreover, the latest Italian advances concerning the pre-clinical evaluation of nanomedicines for melanoma are described.
Collapse
Affiliation(s)
- Luigi Battaglia
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| | - Anna Scomparin
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
- . Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chiara Dianzani
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| | - Paola Milla
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| | - Elisabetta Muntoni
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| | - Silvia Arpicco
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| | - Roberta Cavalli
- . Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy; (L.B.); (A.S.); (C.D.); (P.M.); (E.M.); (S.A.)
| |
Collapse
|
18
|
Kanda Y, Okazaki T, Katakai T. Motility Dynamics of T Cells in Tumor-Draining Lymph Nodes: A Rational Indicator of Antitumor Response and Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:4616. [PMID: 34572844 PMCID: PMC8465463 DOI: 10.3390/cancers13184616] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 01/22/2023] Open
Abstract
The migration status of T cells within the densely packed tissue environment of lymph nodes reflects the ongoing activation state of adaptive immune responses. Upon encountering antigen-presenting dendritic cells, actively migrating T cells that are specific to cognate antigens slow down and are eventually arrested on dendritic cells to form immunological synapses. This dynamic transition of T cell motility is a fundamental strategy for the efficient scanning of antigens, followed by obtaining the adequate activation signals. After receiving antigenic stimuli, T cells begin to proliferate, and the expression of immunoregulatory receptors (such as CTLA-4 and PD-1) is induced on their surface. Recent findings have revealed that these 'immune checkpoint' molecules control the activation as well as motility of T cells in various situations. Therefore, the outcome of tumor immunotherapy using checkpoint inhibitors is assumed to be closely related to the alteration of T cell motility, particularly in tumor-draining lymph nodes (TDLNs). In this review, we discuss the migration dynamics of T cells during their activation in TDLNs, and the roles of checkpoint molecules in T cell motility, to provide some insight into the effect of tumor immunotherapy via checkpoint blockade, in terms of T cell dynamics and the importance of TDLNs.
Collapse
Affiliation(s)
- Yasuhiro Kanda
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-8510, Japan;
| | - Taku Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan;
| | - Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-8510, Japan;
| |
Collapse
|
19
|
Aragoneses-Fenoll L, Montes-Casado M, Ojeda G, García-Paredes L, Arimura Y, Yagi J, Dianzani U, Portolés P, Rojo JM. Role of endocytosis and trans-endocytosis in ICOS costimulator-induced downmodulation of the ICOS Ligand. J Leukoc Biol 2021; 110:867-884. [PMID: 33527556 PMCID: PMC8597029 DOI: 10.1002/jlb.2a0220-127r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/16/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
The interaction between the T-lymphocyte costimulatory molecule ICOS and its ligand (ICOS-L) is needed for efficient immune responses, but expression levels are tightly controlled, as altered expression of ICOS or ICOS-L may lead to immunodeficiency, or favor autoimmune diseases and tumor growth. Using cells of mouse B cell lymphoma (M12.C3) and melanoma (B16), or hamster CHO cells transfected with various forms of mouse ICOS-L, and ICOS+ T cell lines, we show that, within minutes, ICOS induces significant downmodulation of surface ICOS-L that is largely mediated by endocytosis and trans-endocytosis. So, after interaction with ICOS+ cells, ICOS-L was found inside permeabilized cells, or in cell lysates, with significant transfer of ICOS from ICOS+ T cells to ICOS-L-expressing cells, and simultaneous loss of surface ICOS by the T cells. Data from cells expressing ICOS-L mutants show that conserved, functionally important residues in the cytoplasmic domain of mouse ICOS-L (Arg300 , Ser307 and Tyr308 ), or removal of ICOS-L cytoplasmic tail have minor effect on its internalization. Internalization was dependent on temperature, and was partially dependent on actin polymerization, the GTPase dynamin, protein kinase C, or the integrity of lipid rafts. In fact, a fraction of ICOS-L was detected in lipid rafts. On the other hand, proteinase inhibitors had negligible effects on early modulation of ICOS-L from the cell surface. Our data add a new mechanism of control of ICOS-L expression to the regulation of ICOS-dependent responses.
Collapse
Affiliation(s)
- Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - María Montes-Casado
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Gloria Ojeda
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Lucía García-Paredes
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain.,Current address: Hospital 12 de Octubre, Departamento de Oncología Médica, Av. de Córdoba, s/n, Madrid, 28041, Spain
| | - Yutaka Arimura
- Host Defense for Animals, Nippon Veterinary and Life Science University, 1-7-1 Kyonan, Musashino, Tokyo, 180-8602, Japan
| | - Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, 108-8639, Japan
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, 28100, Italy
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain.,Presidencia, Consejo Superior de Investigaciones Científicas, Madrid, 28006, Spain
| | - José M Rojo
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| |
Collapse
|
20
|
Fiorilli S, Pagani M, Boggio E, Gigliotti CL, Dianzani C, Gauthier R, Pontremoli C, Montalbano G, Dianzani U, Vitale-Brovarone C. Sr-Containing Mesoporous Bioactive Glasses Bio-Functionalized with Recombinant ICOS-Fc: An In Vitro Study. NANOMATERIALS 2021; 11:nano11020321. [PMID: 33513769 PMCID: PMC7911784 DOI: 10.3390/nano11020321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022]
Abstract
Osteoporotic bone fractures represent a critical clinical issue and require personalized and specific treatments in order to stimulate compromised bone tissue regeneration. In this clinical context, the development of smart nano-biomaterials able to synergistically combine chemical and biological cues to exert specific therapeutic effects (i.e., pro-osteogenic, anti-clastogenic) can allow the design of effective medical solutions. With this aim, in this work, strontium-containing mesoporous bioactive glasses (MBGs) were bio-functionalized with ICOS-Fc, a molecule able to reversibly inhibit osteoclast activity by binding the respective ligand (ICOS-L) and to induce a decrease of bone resorption activity. N2 adsorption analysis and FT-IR spectroscopy were used to assess the successful grafting of ICOS-Fc on the surface of Sr-containing MBGs, which were also proved to retain the peculiar ability to release osteogenic strontium ions and an excellent bioactivity after functionalization. An ELISA-like assay allowed to confirm that grafted ICOS-Fc molecules were able to bind ICOS-L (the ICOS binding ligand) and to investigate the stability of the amide binding to hydrolysis in aqueous environment up to 21 days. In analogy to the free form of the molecule, the inhibitory effect of grafted ICOS-Fc on cell migratory activity was demonstrated by using ICOSL positive cell lines and the ability to inhibit osteoclast differentiation and function was confirmed by monitoring the differentiation of monocyte-derived osteoclasts (MDOCs), which revealed a strong inhibitory effect, also proven by the downregulation of osteoclast differentiation genes. The obtained results showed that the combination of ICOS-Fc with the intrinsic properties of Sr-containing MBGs represents a very promising approach to design personalized solutions for patients affected by compromised bone remodeling (i.e., osteoporosis fractures).
Collapse
Affiliation(s)
- Sonia Fiorilli
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
- Correspondence:
| | - Mattia Pagani
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
| | - Elena Boggio
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy; (E.B.); (C.L.G.)
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy;
| | - Casimiro Luca Gigliotti
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy; (E.B.); (C.L.G.)
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy;
| | - Chiara Dianzani
- Department of Drug Science and Technology, Università di Torino, Via Pietro Giuria 9, 10125 Torino, Italy;
| | - Rémy Gauthier
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
| | - Carlotta Pontremoli
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
| | - Giorgia Montalbano
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy;
| | - Chiara Vitale-Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.P.); (R.G.); (C.P.); (G.M.); (C.V.-B.)
| |
Collapse
|
21
|
Iwata R, Hyoung Lee J, Hayashi M, Dianzani U, Ofune K, Maruyama M, Oe S, Ito T, Hashiba T, Yoshimura K, Nonaka M, Nakano Y, Norian L, Nakano I, Asai A. ICOSLG-mediated regulatory T-cell expansion and IL-10 production promote progression of glioblastoma. Neuro Oncol 2021; 22:333-344. [PMID: 31634400 DOI: 10.1093/neuonc/noz204] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Targeting immune checkpoint proteins has recently gained substantial attention due to the dramatic success of this strategy in clinical trials for some cancers. Inducible T-cell co-stimulator ligand (ICOSLG) is a member of the B7 family of immune regulatory ligands, expression of which in cancer is implicated in disease progression due to regulation of antitumor adaptive immunity. Although aberrant ICOSLG expression has been reported in glioma cells, the underlying mechanisms that promote glioblastoma (GBM) progression remain elusive. METHODS Here, we investigated a causal role for ICOSLG in GBM progression by analyzing ICOSLG expression in both human glioma tissues and patient-derived GBM sphere cells (GSCs). We further examined its immune modulatory effects and the underlying molecular mechanisms. RESULTS Bioinformatics analysis and GBM tissue microarray showed that upregulation of ICOSLG expression was associated with poor prognosis in patients with GBM. ICOSLG expression was upregulated preferentially in mesenchymal GSCs but not in proneural GSCs in a tumor necrosis factor-α/nuclear factor-kappaB-dependent manner. Furthermore, ICOSLG expression by mesenchymal GSCs promoted expansion of T cells that produced interleukin-10. Knockdown of the gene encoding ICOSLG markedly reduced GBM tumor growth in immune competent mice, with a concomitant downregulation of interleukin-10 levels in the tumor microenvironment. CONCLUSIONS Inhibition of the ICOSLG-inducible co-stimulator axis in GBM may provide a promising immunotherapeutic approach for suppressing a subset of GBM with an elevated mesenchymal signature.
Collapse
Affiliation(s)
- Ryoichi Iwata
- Department of Neurosurgery, Kansai Medical University, Hirakata, Japan
| | - Joo Hyoung Lee
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mikio Hayashi
- Department of Physiology, Kansai Medical University, Hirakata, Japan
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases, Department of Health Sciences, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | - Kohei Ofune
- Department of Neurosurgery, Kansai Medical University, Hirakata, Japan
| | - Masato Maruyama
- Department of Anatomy and Brain Science, Kansai Medical University, Hirakata, Japan
| | - Souichi Oe
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Tetsuo Hashiba
- Department of Neurosurgery, Kansai Medical University, Hirakata, Japan
| | | | - Masahiro Nonaka
- Department of Neurosurgery, Kansai Medical University, Hirakata, Japan
| | - Yosuke Nakano
- Department of Anatomy and Brain Science, Kansai Medical University, Hirakata, Japan
| | - Lyse Norian
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ichiro Nakano
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Akio Asai
- Department of Neurosurgery, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
22
|
Abstract
ICOSL/ICOS are costimulatory molecules pertaining to immune checkpoints; their binding transduces signals having anti-tumor activity. Osteopontin (OPN) is here identified as a ligand for ICOSL. OPN binds a different domain from that used by ICOS, and the binding induces a conformational change in OPN, exposing domains that are relevant for its functions. Here we show that in vitro, ICOSL triggering by OPN induces cell migration, while inhibiting anchorage-independent cell growth. The mouse 4T1 breast cancer model confirms these data. In vivo, OPN-triggering of ICOSL increases angiogenesis and tumor metastatization. The findings shed new light on ICOSL function and indicate that another partner beside ICOS may be involved; they also provide a rationale for developing alternative therapeutic approaches targeting this molecular trio. Davide Raineri, Chiara Dianzani et al. show that osteopontin binds ICOSL at a different domain than the one used by ICOS. Activation of ICOSL by osteopontin induces cell migration in vitro and tumor metastatization in a 4T1 breast cancer mouse model; highlighting the functional role of this interaction in cancer progression.
Collapse
|
23
|
Tong M, Fang X, Yang J, Wu P, Guo Y, Sun J. Abnormal membrane-bound and soluble programmed death ligand 2 (PD-L2) expression in systemic lupus erythematosus is associated with disease activity. Immunol Lett 2020; 227:96-101. [PMID: 32891685 DOI: 10.1016/j.imlet.2020.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Programmed death ligand (PD-L) 2 and PD-L1 are the second and first ligands, respectively, for programmed cell death-1 protein (PD-1), which is one of the key factors responsible for inhibitory T cell signaling, mediating mechanisms of tolerance and providing immune homeostasis. Studies have shown that PD-1 and its ligand PD-L1 are abnormally expressed in autoimmune diseases, such as rheumatoid arthritis and autoimmune hepatitis, but its other ligand, PD-L2, has rarely been studied. This study analyzed the changes in membrane-bound PD-L2 expression in peripheral blood mononuclear cells and soluble PD-L2 (sPD-L2) levels in the serum of patients with systemic lupus erythematosus (SLE) to explore the relationship between PD-L2 expression with disease activity and related test parameters. Our results showed that membrane-bound PD-L2 expression on monocytes was significantly higher and the sPD-L2 levels were significantly lower in SLE patients than in healthy subjects. Patients with active SLE accompanied by lupus nephritis, joint pain, and clinical manifestations of oral ulcers had relatively low secretion of sPD-L2. In addition, this secretion level was significantly and positively correlated with complement components 3 and 4 (C3/C4). These results suggest that PD-L2 may be a promising biomarker associated with the pathogenesis of SLE.
Collapse
Affiliation(s)
- Min Tong
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Xiaohui Fang
- Laboratory, Suzhou Hospital of Traditional Chinese Medicine, Jiangsu, Suzhou, 215009, PR China
| | - Jie Yang
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Pingping Wu
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Yundi Guo
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Jing Sun
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China.
| |
Collapse
|
24
|
Guillermo Espinoza-Contreras J, Idalia Torres-Ruiz M, Ariel Waller-González L, De Jesús Ramírez-García J, Torres-López J, Ventura-Juárez J, Verónica Moreno-Córdova E, Ernesto López-Ramos J, Humberto Muñoz-Ortega M, Eugenia Vargas-Camaño M, González-Segovia R. Immunological markers and Helicobacter pylori in patients with stomach cancer: Expression and correlation. Biomed Rep 2020; 12:233-243. [PMID: 32257186 PMCID: PMC7100142 DOI: 10.3892/br.2020.1285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 01/17/2020] [Indexed: 01/22/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) and ICOS-L (also referred to as B7 homolog 1 and 2, respectively) modulate the immune inflammatory response. The aim of the present study was to examine the expression levels of these inflammatory mediators in two groups of patients with an Helicobacter pylori (H. pylori) infection; patients with and without gastric cancer. The association between bacterial virulence factors, CagA and VacA, was also examined, as well as their correlation with the inflammatory profile. Endoscopy analysis indicated that 18 patients suffered from cancer and 28 patients suffered from other gastric pathologies. PCR and reverse transcription-quantitative PCR were used to analyze gastric biopsies and determine the expression levels of the inflammatory modulators PD-L1 and ICOS-L, transcription factors, cytokines and other genes associated with inflammation and pathogenicity. All 46 patients were determined positive for markers of H. pylori. Patients with stomach cancer had lower levels of ICOS-L (P<0.05) and GATA3 (P<0.01), a negative correlation between CagA and IL-17 (P<0.05), a positive correlation between CagA and IL-10 (P<0.05), a negative correlation between vacA-m1 and retinoid orphan receptor γt (RORγt) (P<0.001), and a positive correlation between RORγt and ICOS-L (P<0.001). The reduced levels of ICOS-L and GATA3 along with the negative correlation between CagA and IL-17, and between vacA-m1 and RORγt were all associated with an increased risk of gastric cancer in the present cohort.
Collapse
Affiliation(s)
| | - Miriam Idalia Torres-Ruiz
- Departamento de Endoscopia, Centro Médico Nacional 20 de Noviembre ISSSTE, Ciudad de México, CP 03229, México
| | - Luis Ariel Waller-González
- Departamento de Endoscopia, Centro Médico Nacional 20 de Noviembre ISSSTE, Ciudad de México, CP 03229, México
| | | | - Javier Torres-López
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias UMAE, Hospital de Pediatría, IMSS, Ciudad de México, CP 06720, México
| | - Javier Ventura-Juárez
- Departamento de Morfología, Universidad Autónoma de Aguascalientes, Aguascalientes Ags, CP 20131, México
| | | | - Juan Ernesto López-Ramos
- Departamento de Microbiología, Universidad Autónoma de Aguascalientes, Aguascalientes Ags, CP 20131, México
| | | | - María Eugenia Vargas-Camaño
- Servicio de Inmunología Clínica y Alergía, Centro Médico Nacional 20 Noviembre ISSSTE, Ciudad de México, CP 03229, México
| | - Rodolfo González-Segovia
- Departamento de Microbiología, Universidad Autónoma de Aguascalientes, Aguascalientes Ags, CP 20131, México
| |
Collapse
|
25
|
The Cancer Stem Cell in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12030684. [PMID: 32183251 PMCID: PMC7140091 DOI: 10.3390/cancers12030684] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
The recognition of intra-tumoral cellular heterogeneity has given way to the concept of the cancer stem cell (CSC). According to this concept, CSCs are able to self-renew and differentiate into all of the cancer cell lineages present within the tumor, placing the CSC at the top of a hierarchical tree. The observation that these cells—in contrast to bulk tumor cells—are able to exclusively initiate new tumors, initiate metastatic spread and resist chemotherapy implies that CSCs are solely responsible for tumor recurrence and should be therapeutically targeted. Toward this end, dissecting and understanding the biology of CSCs should translate into new clinical therapeutic approaches. In this article, we review the CSC concept in cancer, with a special focus on hepatocellular carcinoma.
Collapse
|
26
|
Clemente N, Boggio E, Gigliotti LC, Raineri D, Ferrara B, Miglio G, Argenziano M, Chiocchetti A, Cappellano G, Trotta F, Caldera F, Capucchio MT, Yagi J, Rojo MJ, Renò F, Cavalli R, Dianzani C, Dianzani U. Immunotherapy of experimental melanoma with ICOS-Fc loaded in biocompatible and biodegradable nanoparticles. J Control Release 2020; 320:112-124. [PMID: 31962094 DOI: 10.1016/j.jconrel.2020.01.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 01/18/2023]
Abstract
Inducible T-cell costimulator (ICOS) upon binding to its ligand (ICOSL) mediates adaptive immunity and antitumor response. Thus, antitumor therapies targeting the ICOS/ICOSL pathway hold great promise for cancer treatment. In this regard, ICOSL triggering by a soluble recombinant form of ICOS (ICOS-Fc) hampered adhesiveness and migration of dendritic, endothelial, and tumor cells in vitro. Furthermore, in vivo treatment with ICOS-Fc previously showed the capability to inhibit lung metastatization of ICOSL+ B16-F10 melanoma cells when injected intravenously in mice, but it failed to block the growth of established subcutaneous B16-F10 murine tumors. Thus, we asked whether passive targeting of solid tumors with ICOS-Fc-loaded biocompatible and biodegradable nanoparticles (NPs) could instead prove effectiveness in reducing tumor growth. Here, ICOS-Fc was loaded in two types of polymer nanoparticles, i.e. cross-linked β-cyclodextrin nanosponges (CDNS) and poly(lactic-co-glycolic acid) (PLGA) NPs and in vitro characterized. In vivo experiments showed that treatment of C57BL6/J mice with ICOS-Fc loaded into the two nanoformulations inhibits the growth of established subcutaneous B16-F10 tumors. This anticancer activity appears to involve both anti-angiogenic and immunoregulatory effects, as shown by decreased tumor vascularization and downmodulation of IL-10 and Foxp3, two markers of regulatory T cells (Tregs). Overall, the substantial in vivo anticancer activity of ICOS-Fc-loaded CDNS and PLGA NPs against different components of the tumor microenvironment makes these nanoformulations attractive candidates for future combination cancer therapy.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Luca Casimiro Gigliotti
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Davide Raineri
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Benedetta Ferrara
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Gianluca Miglio
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Monica Argenziano
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Torino, 10125 Torino, Italy
| | - Fabrizio Caldera
- Department of Chemistry, University of Torino, 10125 Torino, Italy
| | | | - Junji Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 108-8639, Japan
| | - Maria Josè Rojo
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientıficas, 28040 Madrid, Spain
| | - Filippo Renò
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy
| | - Roberta Cavalli
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy.
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Torino, 10125 Torino, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Inter Interdisciplinary Research Center of Autoimmune Diseases, UPO, 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
27
|
Cucci MA, Compagnone A, Daga M, Grattarola M, Ullio C, Roetto A, Palmieri A, Rosa AC, Argenziano M, Cavalli R, Simile MM, Pascale RM, Dianzani C, Barrera G, Pizzimenti S. Post-translational inhibition of YAP oncogene expression by 4-hydroxynonenal in bladder cancer cells. Free Radic Biol Med 2019; 141:205-219. [PMID: 31207288 DOI: 10.1016/j.freeradbiomed.2019.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/15/2019] [Accepted: 06/07/2019] [Indexed: 12/23/2022]
Abstract
The transcriptional regulator YAP plays an important role in cancer progression and is negatively controlled by the Hippo pathway. YAP is frequently overexpressed in human cancers, including bladder cancer. Interestingly, YAP expression and activity can be inhibited by pro-oxidant conditions; moreover, YAP itself can also affect the cellular redox status through multiple mechanisms. 4-Hydroxynonenal (HNE), the most intensively studied end product of lipid peroxidation, is a pro-oxidant agent able to deplete GSH and has an anti-tumoral effect by affecting multiple signal pathways, including the down-regulation of oncogene expressions. These observations prompted us to investigate the effect of HNE on YAP expression and activity. We demonstrated that HNE inhibited YAP expression and its target genes in bladder cancer cells through a redox-dependent mechanism. Moreover, the YAP down-regulation was accompanied by an inhibition of proliferation, migration, invasion, and angiogenesis, as well as by an accumulation of cells in the G2/M phase of cell cycle and by an induction of apoptosis. We also established the YAP role in inhibiting cell viability and inducing apoptosis in HNE-treated cells by using an expression vector for YAP. Furthermore, we identified a post-translational mechanism for the HNE-induced YAP expression inhibition, involving an increase of YAP phosphorylation and ubiquitination, leading to proteasomal degradation. Our data established that HNE can post-translationally down-regulate YAP through a redox-dependent mechanism and that this modulation can contribute to determining the specific anti-cancer effects of HNE.
Collapse
Affiliation(s)
- Marie Angele Cucci
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Alessandra Compagnone
- Department of Oncology, University of Turin, Via Michelangelo 27, 10125, Turin, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Margherita Grattarola
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Chiara Ullio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Antonietta Palmieri
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Arianna Carolina Rosa
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Monica Argenziano
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Roberta Cavalli
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Maria Maddalena Simile
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Rosa Maria Pascale
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Regione Gonzole 10, 10043, Orbassano, Turin, Italy.
| |
Collapse
|
28
|
Chiocchetti A, Cappellano G, Dianzani U. To each his own: a personalized vaccine for metastatic melanoma. Gland Surg 2019; 8:329-333. [PMID: 31538056 PMCID: PMC6723019 DOI: 10.21037/gs.2017.11.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| | - Giuseppe Cappellano
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
29
|
Clemente N, Argenziano M, Gigliotti CL, Ferrara B, Boggio E, Chiocchetti A, Caldera F, Trotta F, Benetti E, Annaratone L, Ribero S, Pizzimenti S, Barrera G, Dianzani U, Cavalli R, Dianzani C. Paclitaxel-Loaded Nanosponges Inhibit Growth and Angiogenesis in Melanoma Cell Models. Front Pharmacol 2019; 10:776. [PMID: 31354491 PMCID: PMC6639435 DOI: 10.3389/fphar.2019.00776] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/17/2019] [Indexed: 12/25/2022] Open
Abstract
This study investigated the effects of free paclitaxel (PTX) and PTX-loaded in pyromellitic nanosponges (PTX-PNS) in reducing in vitro and in vivo melanoma cell growth and invasivity, and in inhibiting angiogenesis. To test the response of cells to the two PTX formulations, the cell viability was evaluated by MTT assay in seven continuous cell lines, in primary melanoma cells, both in 2D and 3D cultures, and in human umbilical vein endothelial cells (HUVECs) after exposure to different concentrations of PTX or PTX-PNS. Cell motility was assessed by a scratch assay or Boyden chamber assay, evaluating cell migration in presence or absence of diverse concentrations of PTX or PTX-PNS. The effect of PTX and PTX-PNS on angiogenesis was evaluated as endothelial tube formation assay, a test able to estimate the formation of three-dimensional vessels in vitro. To assess the anticancer effect of PTX and PTX-PNS in in vivo experiments, the two drug formulations were tested in a melanoma mouse model obtained by B16-BL6 cell implantation in C57/BL6 mice. Results obtained were as follows: 1) MTT analysis revealed that cell proliferation was more affected by PTX-PNS than by PTX in all tested cell lines, in both 2D and 3D cultures; 2) the analysis of the cell migration showed that PTX-PNS acted at very lower concentrations than PTX; 3) tube formation assay showed that PTX-PNS were more effective in inhibiting tube formation than free PTX; and 4) in vivo experiments demonstrated that tumor weights, volumes, and growth were significantly reduced by PTX-PNS treatment with respect to PTX; the angiogenesis and the cell proliferation, detected in the tumor samples with CD31 and Ki-67 antibodies, respectively, indicated that, in the PTX-PNS-treated tumors, the tube formation was inhibited, and a low amount of proliferating cells was present. Taken together, our data demonstrated that our new PTX nanoformulation can respond to some important issues related to PTX treatment, lowering the anti-tumor effective doses and increasing the effectiveness in inhibiting melanoma growth in vivo.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), UPO, Novara, Italy
| | - Monica Argenziano
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Torino, Torino, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), UPO, Novara, Italy
| | - Benedetta Ferrara
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Torino, Torino, Italy
| | - Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), UPO, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), UPO, Novara, Italy
| | | | | | - Elisa Benetti
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Torino, Torino, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Simone Ribero
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), UPO, Novara, Italy
| | - Roberta Cavalli
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Torino, Torino, Italy
| | - Chiara Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco, University of Torino, Torino, Italy
| |
Collapse
|
30
|
Bessone F, Argenziano M, Grillo G, Ferrara B, Pizzimenti S, Barrera G, Cravotto G, Guiot C, Stura I, Cavalli R, Dianzani C. Low-dose curcuminoid-loaded in dextran nanobubbles can prevent metastatic spreading in prostate cancer cells. NANOTECHNOLOGY 2019; 30:214004. [PMID: 30654342 DOI: 10.1088/1361-6528/aaff96] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Preventing recurrences and metastasis of prostate cancer after prostatectomy by administering adjuvant therapies is quite a controversial issue. In addition to effectiveness, absence of side effects and long term toxicity are mandatory. Curcuminoids (Curc) extracted with innovative techniques and effectively loaded by polymeric nanobubbles (Curc-NBs) satisfy such requirements. Curc-NBs showed stable over 30 d, were effectively internalized by tumor cells and were able to slowly release Curc in a sustained way. Significant biological effects were detected in PC-3 and DU-145 cell lines where Curc-NBs were able to inhibit adhesion and migration, to promote cell apoptosis and to affect cell viability and colony-forming capacity in a dose-dependent manner. Since the favourable effects are already detectable at very low doses, which can be reached at a clinical level, the actual drug concentration can be visualized and monitored by US or MRI, Curc-NBs can be proposed as an effective adjuvant theranostic tool.
Collapse
Affiliation(s)
- F Bessone
- Department of Drug Science & Technology, University of Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ruiu R, Tarone L, Rolih V, Barutello G, Bolli E, Riccardo F, Cavallo F, Conti L. Cancer stem cell immunology and immunotherapy: Harnessing the immune system against cancer's source. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:119-188. [PMID: 31383404 DOI: 10.1016/bs.pmbts.2019.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite recent advances in diagnosis and therapy having improved cancer outcome, many patients still do not respond to treatments, resulting in the progression or relapse of the disease, eventually impairing survival expectations. The limited efficacy of therapy is often attributable to its inability to affect cancer stem cells (CSCs), a small population of cells resistant to current radio- and chemo-therapies. CSCs are characterized by self-renewal and tumor-initiating capabilities, and function as a reservoir for the local and distant recurrence of the disease. Therefore, new therapeutic approaches able to effectively target and deplete CSCs are urgently needed. Immunotherapy is facing a renewed interest for its potential in cancer treatment, and the possibility of harnessing the immune system to target CSCs is being addressed by a new exciting research field. In this chapter, we discuss the cancer stem cell model and illustrate CSC biological and molecular properties, critically addressing theoretical and practical issues linked with their definition and study. We then review the existing literature regarding the immunological properties of CSCs and the complex interplay occurring between CSCs and immune cells. Finally, we present up-to-date studies on CSC immunotargeting and its potential future perspective. In conclusion, understanding the interplay between CSC biology and tumor immunology will provide a deeper understanding of the mechanisms that regulate CSC immunological properties. This will contribute to the design of new CSC-directed immunotherapeutic strategies with the potential of strongly improving cancer outcomes.
Collapse
Affiliation(s)
- Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
32
|
Daga M, Pizzimenti S, Dianzani C, Cucci MA, Cavalli R, Grattarola M, Ferrara B, Scariot V, Trotta F, Barrera G. Ailanthone inhibits cell growth and migration of cisplatin resistant bladder cancer cells through down-regulation of Nrf2, YAP, and c-Myc expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 56:156-164. [PMID: 30668336 DOI: 10.1016/j.phymed.2018.10.034] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/22/2018] [Accepted: 10/27/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Ailanthone (Aila) is a natural active compound isolated from the Ailanthus altissima, which has been shown to possess an "in vitro" growth-inhibitory effect against several cancer cell lines. Advanced bladder cancer is a common disease characterized by a frequent onset of resistance to cisplatin-based therapy. The cisplatin (CDDP) resistance is accompanied by an increase in Nrf2 protein expression which contributes to conferring resistance. Recently, we demonstrated a cross-talk between Nrf2 and YAP. YAP has also been demonstrated to play an important role in chemoresistance of bladder cancer. PURPOSE We analyzed the antitumor effect of Aila in sensitive and CDDP-resistant bladder cancer cells and the molecular mechanisms involved in Aila activity. STUDY DESIGN Sensitive and CDDP-resistant 253J B-V and 253J bladder cancer cells, intrinsically CDDP-resistant T24 bladder cancer cells and HK-2 human renal cortex cells were used. Cells were treated with diverse concentrations of Aila and proliferation, cell cycle, apoptosis and gene expressions were determined. METHODS Aila toxicity and proliferation were determined by MTT and colony forming methods, respectively. Cell cycle was determined by cytofluorimetric analysis through PI staining method. Apoptosis was detected using Annexin V and PI double staining followed by quantitative flow cytometry. Expressions of Nrf2, Yap, c-Myc, and house-keeping genes were determined by western blot with specific antibodies. Cell migration was detected by wound healing and Boyden chamber analysis. RESULTS Aila inhibited the growth of sensitive and CDDP-resistant bladder cancer cells with the same effectiveness. On the contrary, the growth of HK-2 cells was only slightly reduced by Aila. Cell cycle analysis revealed an accumulation of Aila-treated bladder cancer cells in the G0/G1 phase. Interestingly, Aila strongly reduced Nrf2 expression in these cell lines. Moreover, Aila significantly reduced YAP, and c-Myc protein expression. The random and the oriented migration of bladder cancer cells were strongly inhibited by Aila treatment, in particular in CDDP-resistant cells. CONCLUSION Aila inhibited proliferation and invasiveness of bladder cancer cells. Its high effectiveness in CDDP resistant cells could be related to the inhibition of Nrf2, YAP, and c-Myc expressions. Aila could represent a new tool to treating CDDP-resistant bladder cancers.
Collapse
Affiliation(s)
- Martina Daga
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy.
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Marie Angele Cucci
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy
| | - Benedetta Ferrara
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Valentina Scariot
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco (TO), Italy
| | - Francesco Trotta
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125 Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy
| |
Collapse
|
33
|
Ciamporcero E, Daga M, Pizzimenti S, Roetto A, Dianzani C, Compagnone A, Palmieri A, Ullio C, Cangemi L, Pili R, Barrera G. Crosstalk between Nrf2 and YAP contributes to maintaining the antioxidant potential and chemoresistance in bladder cancer. Free Radic Biol Med 2018; 115:447-457. [PMID: 29248722 DOI: 10.1016/j.freeradbiomed.2017.12.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/25/2022]
Abstract
Redox adaptation plays an important role in cancer cells drug resistance. The antioxidant response is principally mediated by the transcription factor Nrf2, that induces the transcriptional activation of several genes involved in GSH synthesis, chemoresistance, and cytoprotection. YAP is emerging as a key mediator of chemoresistance in a variety of cancers, but its role in controlling the antioxidant status of the cells is yet elusive. Here, we show that impairing YAP protein expression reduced GSH content and Nrf2 protein and mRNA expression in bladder cancer cells. Moreover, in YAP knocked down cells the expression of FOXM1, a transcription factor involved in Nrf2 transcription, was down-regulated and the silencing of FOXM1 reduced Nrf2 expression. On the other hand, the silencing of Nrf2, as well as the depletion of GSH by BSO treatment, inhibited YAP expression, suggesting that cross-talk exists between YAP and Nrf2 proteins. Importantly, we found that silencing either YAP or Nrf2 enhanced sensitivity of bladder cancer cells to cytotoxic agents and reduced their migration. Furthermore, the inhibition of both YAP and Nrf2 expressions significantly increased cytotoxic drug sensitivity and synergistically reduced the migration of chemoresistant bladder cancer cells. These findings provide a rationale for targeting these transcriptional regulators in patients with chemoresistant bladder cancer, expressing high YAP and bearing a proficient antioxidant system.
Collapse
Affiliation(s)
- Eric Ciamporcero
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy.
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Alessandra Compagnone
- Department of Oncology, University of Turin, Via Michelangelo 27, 10125 Turin, Italy
| | - Antonietta Palmieri
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Chiara Ullio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Luigi Cangemi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Roberto Pili
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA; Genitourinary Program, Indiana University-Simon Cancer Center, Hematology/Oncology980 W. Walnut Street R3 C516, Indianapolis, IN 46202, USA
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
34
|
Reduced sB7-H3 Expression in the Peripheral Blood of Systemic Lupus Erythematosus Patients. J Immunol Res 2017; 2017:5728512. [PMID: 29423417 PMCID: PMC5750499 DOI: 10.1155/2017/5728512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/21/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022] Open
Abstract
Both membrane-bound and soluble forms of costimulatory molecules play important roles in immune-regulatory networks. B7-H3, a member of the B7 family, has been found with aberrant expression in tumors and infectious disease. However, the significance of sB7-H3 expression in systemic lupus erythematosus (SLE) has not been investigated. Using the peripheral blood of 78 SLE patients, we established a comprehensive database containing clinical data and relevant laboratory tests. We found that sB7-H3 expression in SLE patients was significantly lower compared with the healthy individuals. In addition, sB7-H3 levels in the patients were positively correlated with the disease activity as indicated by SLE disease activity index score, rashes, fever, and inflammatory cytokines. Moreover, sB7-H3 was associated with the counts of red blood cells and hemoglobin. Our findings suggest that sB7-H3 might counteract the aberrant immune response and potentially serve as a monitoring indicator of disease progression and therapeutic target in SLE treatment.
Collapse
|
35
|
Gigliotti CL, Ferrara B, Occhipinti S, Boggio E, Barrera G, Pizzimenti S, Giovarelli M, Fantozzi R, Chiocchetti A, Argenziano M, Clemente N, Trotta F, Marchiò C, Annaratone L, Boldorini R, Dianzani U, Cavalli R, Dianzani C. Enhanced cytotoxic effect of camptothecin nanosponges in anaplastic thyroid cancer cells in vitro and in vivo on orthotopic xenograft tumors. Drug Deliv 2017; 24:670-680. [PMID: 28368209 PMCID: PMC8241155 DOI: 10.1080/10717544.2017.1303856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/02/2017] [Accepted: 03/05/2017] [Indexed: 12/30/2022] Open
Abstract
Anaplastic carcinoma of the thyroid (ATC) is a lethal human malignant cancer with median survival of 6 months. To date, no treatment has substantially changed its course, which makes urgent need for the development of novel drugs or novel formulations for drug delivery. Nanomedicine has enormous potential to improve the accuracy of cancer therapy by enhancing availability and stability, decreasing effective doses and reducing side effects of drugs. Camptothecin (CPT) is an inhibitor of DNA topoisomerase-I with several anticancer properties but has poor solubility and a high degradation rate. Previously, we reported that CPT encapsulated in β-cyclodextrin-nanosponges (CN-CPT) increased solubility, was protected from degradation and inhibited the growth of prostate tumor cells both in vitro and in vivo. The aim of this study was to extend that work by assessing the CN-CPT effectiveness on ATC both in vitro and in vivo. Results showed that CN-CPT significantly inhibited viability, clonogenic capacity and cell-cycle progression of ATC cell lines showing a faster and enhanced effect compared to free CPT. Moreover, CN-CPT inhibited tumor cell adhesion to vascular endothelial cells, migration, secretion of pro-angiogenic factors (IL-8 and VEGF-α), expression of β-PIX, belonging to the Rho family activators, and phosphorylation of the Erk1/2 MAPK. Finally, CN-CPT significantly inhibited the growth, the metastatization and the vascularization of orthotopic ATC xenografts in SCID/beige mice without apparent toxic effects in vivo. This work extends the previous insight showing that β-cyclodextrin-nanosponges are a promising tool for the treatment of ATC.
Collapse
Affiliation(s)
- Casimiro Luca Gigliotti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Benedetta Ferrara
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Sergio Occhipinti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Boggio
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Mirella Giovarelli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Roberto Fantozzi
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Nausicaa Clemente
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Torino, Torino, Italy, and
| | - Caterina Marchiò
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Renzo Boldorini
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, UPO, Novara, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| |
Collapse
|
36
|
Gigliotti CL, Boggio E, Clemente N, Shivakumar Y, Toth E, Sblattero D, D’Amelio P, Isaia GC, Dianzani C, Yagi J, Rojo JM, Chiocchetti A, Boldorini R, Bosetti M, Dianzani U. ICOS-Ligand Triggering Impairs Osteoclast Differentiation and Function In Vitro and In Vivo. THE JOURNAL OF IMMUNOLOGY 2016; 197:3905-3916. [DOI: 10.4049/jimmunol.1600424] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/20/2016] [Indexed: 02/02/2023]
|
37
|
Hu X, Wu J, An J, Hu Y, Shen Y, Liu C, Zhang X. Development of a novel monoclonal antibody to human inducible co-stimulator ligand (ICOSL): Biological characteristics and application for enzyme-linked immunosorbent assay. Int Immunopharmacol 2016; 36:151-157. [PMID: 27138044 DOI: 10.1016/j.intimp.2016.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/28/2022]
Abstract
ICOSL (B7-H2, CD275), a co-stimulatory molecule of the B7 superfamily, functions as a positive signal in immune response. To investigate whether ICOSL could be released into sera and the possible biological function of soluble ICOS (sICOSL), we generated and characterized a functional anti-human ICOSL monoclonal antibody (mAb), 20B10, and developed a novel enzyme-linked immunosorbent assay (ELISA) based on two anti-human ICOSL antibodies with different epitope specificities. Using the ELISA system, we found that sICOSL in the serum of healthy donors increases in an age-dependent manner and that the matrix metalloproteinase inhibitor (MMPI) could suppress sICOSL production. Together, these data demonstrate that the existence of circulating sICOSL in human serum might play an important role in immunoregulation.
Collapse
Affiliation(s)
- Xiaohan Hu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Jian Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jingnan An
- Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Yumin Hu
- Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Cuiping Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Immunology, Medical College of Soochow University, Suzhou 215006, China.
| |
Collapse
|
38
|
Hu X, Liu C, An J, Shen Y, Hu Y, Jiang J, Wu J, Zhang X. Development of a Novel Functional Monoclonal Antibody to Human CD275: Characterization and Biological Activity. Monoclon Antib Immunodiagn Immunother 2016; 35:18-24. [PMID: 26788906 DOI: 10.1089/mab.2015.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CD275 (B7-H2, ICOSL), a co-stimulatory molecule of the B7 superfamily, plays a critical role in immune response. In this report, a novel mouse anti-human CD275 monoclonal antibody (MAb) was prepared using hybridoma technology, and immunological characteristics of the MAb were determined. The results showed that the MAb (clone 13D11) was IgG2(κ) and bound specifically to human CD275. By mutual competition, we found that the antibody recognized different epitopes of CD275 antigen compared with commercial antibodies and could block ICOS-CD275 interaction. Crosslinking of CD275 with MAb 13D11 markedly blocked ICOS positive signal and inhibited T cell proliferation and cytokine production. In addition, the 13D11 MAb was suitable for indirect ELISA detection. Thus, the MAb against human CD275 with high specificity and different activity would be useful for further study of this molecule.
Collapse
Affiliation(s)
- Xiaohan Hu
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Cuiping Liu
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Jingnan An
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Yu Shen
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Yumin Hu
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Juean Jiang
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Jian Wu
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Xueguang Zhang
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China .,2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| |
Collapse
|