1
|
Wang Y, Sen-Majumdar A, Li JM, Sarkar S, Passang T, Li Y, Cohen J, Chen Z, Chaudagar K, Das PK, Wang S, Bruk N, Papadantonakis N, Giver CR, Waller EK. Identification and characterization of vasoactive intestinal peptide receptor antagonists with high-affinity and potent anti-leukemia activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622716. [PMID: 39605448 PMCID: PMC11601233 DOI: 10.1101/2024.11.08.622716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Vasoactive intestinal peptide (VIP) is a neuropeptide involved in tumor growth and immune modulating functions. Previous research indicated that a VIP antagonist (VIPhyb) enhances T-cell activation and induces T-cell-dependent anti-leukemic activity in mice. We created a combinatorial library of VIPhyb C-terminal sequence variations to develop a more potent VIP-receptor (VIP-R) antagonist, hypothesizing that specific amino acid substitutions would improve receptor binding and plasma stability. In silico screening analyses identified sequences with improved docking scores predicting increased binding affinity to human VIP receptors VPAC1 and VPAC2. Fifteen peptides were synthesized and tested for their ability to potentiate activation of purified mouse and human T cells and enhance T cell-dependent anti-leukemia responses in murine models of acute myeloid leukemia. Treating C57Bl/6 mice engrafted with a C1498 myeloid leukemia cell line with daily subcutaneous injections of VIP-R antagonist peptides induced T cell activation resulting in specific anti-leukemia responses. Strikingly, the predicted binding affinity of the VIP-R antagonists to VIP receptors correlated positively with their ability to augment mouse T-cell proliferation and anti-leukemia activity. ANT308 and ANT195 emerged as top candidates due to their high predicted VIP-R binding, low EC 50 for in vitro T cell activation, and potent anti-leukemia activities. ANT308 decreased CREB phosphorylation, a downstream signaling pathway of the VIP receptor, and stimulated granzyme B and perforin expression in CD8+ T cells from AML patients. Combining in silico modeling, in vitro T cell activation properties, and in vivo anti-leukemia activity has identified promising VIP-R antagonist candidates for further development as novel immunotherapies for AML, especially for patients with relapsed disease.
Collapse
|
2
|
Zhang H, Passang T, Ravindranathan S, Bommireddy R, Jajja MR, Yang L, Selvaraj P, Paulos CM, Waller EK. The magic of small-molecule drugs during ex vivo expansion in adoptive cell therapy. Front Immunol 2023; 14:1154566. [PMID: 37153607 PMCID: PMC10160370 DOI: 10.3389/fimmu.2023.1154566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
In the past decades, advances in the use of adoptive cellular therapy to treat cancer have led to unprecedented responses in patients with relapsed/refractory or late-stage malignancies. However, cellular exhaustion and senescence limit the efficacy of FDA-approved T-cell therapies in patients with hematologic malignancies and the widespread application of this approach in treating patients with solid tumors. Investigators are addressing the current obstacles by focusing on the manufacturing process of effector T cells, including engineering approaches and ex vivo expansion strategies to regulate T-cell differentiation. Here we reviewed the current small-molecule strategies to enhance T-cell expansion, persistence, and functionality during ex vivo manufacturing. We further discussed the synergistic benefits of the dual-targeting approaches and proposed novel vasoactive intestinal peptide receptor antagonists (VIPR-ANT) peptides as emerging candidates to enhance cell-based immunotherapy.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Tenzin Passang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Mohammad Raheel Jajja
- Departmert of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, United States
| | - Lily Yang
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Chrystal M. Paulos
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Department of Microbiology and Immunology, Emory University of School of Medicine, Atlanta, GA, United States
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| |
Collapse
|
3
|
Gallino L, Hauk V, Castagnola L, Vota D, Pascuali N, Parborell F, May M, Fontana V, Merech F, Naguila Z, Waschek J, Leirós CP, Ramhorst R. Vasoactive intestinal peptide deficiency promotes ovarian dysfunction associated to a proinflammatory microenvironment reminiscent of premature aging. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166585. [PMID: 36423894 DOI: 10.1016/j.bbadis.2022.166585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 10/15/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022]
Abstract
Complex immune regulation during pregnancy is required to ensure a successful pregnancy outcome. Vasoactive intestinal peptide (VIP) has local immunoregulatory effects on the ovary, uterus and maternal-fetal interface that favor a tolerogenic maternal microenvironment. Since the VIP Knockout (KO) mice are subfertile, we investigated the mechanisms underlying the effects of VIP deficiency on ovarian physiology and immune homeostasis. Therefore, we studied VIP KO, deficient (HT) and wild type (WT) female mice in estrus at 3 or 8 months of age. Young KO mice showed abnormal cycle timing and regularity associated with dysfunctional ovaries. Ovaries presented higher number of atretic follicles and reduced number of corpora lutea leading to a lower ovulation rates. Part of the VIP KO mice (25 %) failed to ovulate or ovulated oocytes incompetent to be fertilized (50 %). In particular, ovaries of young KO mice exhibited features of premature aging accompanied by a pro-inflammatory milieu with increased levels of IL-1β. A unique macrophage subpopulation identified as "foamy macrophages" was found. On the other hand, aged VIP KO females did not gain body weight probably due to the sustained production of E2. Finally, the adoptive transfer of FOXP3+ cells to infertile VIP KO females resulted in their selective recruitment to the ovary. It increased FOXP3/RORγt and TGFβ/IL-6 ratio improving ovarian microenvironment and pregnancy rate. The present results suggest that VIP contributes to ovarian homeostatic mechanisms required for a successful pregnancy.
Collapse
Affiliation(s)
- Lucila Gallino
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Vanesa Hauk
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Lara Castagnola
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Daiana Vota
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | | | | | - Maria May
- ININFA, Universidad de Buenos Aires, CONICET, Argentina
| | - Vanina Fontana
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Fatima Merech
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Zaira Naguila
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - James Waschek
- The David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Claudia Perez Leirós
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
4
|
Zhu J, Wang Y, Li J, Das PK, Zhang H, Passang T, Li JM, Nagy T, Gandhi K, Ravindranathan S, Giver CR, Hassan M, Li Y, Antonova AU, Wang S, Roback JD, Waller EK. Donor plasmacytoid dendritic cells limit graft-versus-host disease through vasoactive intestinal polypeptide expression. Blood 2022; 140:1431-1447. [PMID: 35443019 PMCID: PMC9507007 DOI: 10.1182/blood.2021012561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 03/21/2022] [Indexed: 11/20/2022] Open
Abstract
Vasoactive intestinal polypeptide (VIP), an anti-inflammatory neuropeptide with pleiotropic cardiovascular effects, induces differentiation of hematopoietic stem cells into regulatory dendritic cells that limit graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplant (HSCT) recipients. We have previously shown that donor plasmacytoid dendritic cells (pDCs) in bone marrow (BM) donor grafts limit the pathogenesis of GVHD. In this current study we show that murine and human pDCs express VIP, and that VIP-expressing pDCs limit T-cell activation and expansion using both in vivo and in vitro model systems. Using T cells or pDCs from transgenic luciferase+ donors in murine bone marrow transplantation (BMT), we show similar homing patterns of donor pDCs and T cells to the major sites for alloactivation of donor T cells: spleen and gut. Cotransplanting VIP-knockout (KO) pDCs with hematopoietic stem cells and T cells in major histocompatibility complex mismatched allogeneic BMT led to lower survival, higher GVHD scores, and more colon crypt cell apoptosis than transplanting wild-type pDCs. BMT recipients of VIP-KO pDCs had more T helper 1 polarized T cells, and higher plasma levels of granulocyte-macrophage colony-stimulating factor and tumor necrosis factor-α than recipients of wild-type pDCs. T cells from VIP-KO pDC recipients had increasing levels of bhlhe40 transcripts during the first 2 weeks posttransplant, and higher levels of CyclophilinA/Ppia transcripts at day 15 compared with T cells from recipients of wild-type pDCs. Collectively, these data indicate paracrine VIP synthesis by donor pDCs limits pathogenic T-cell inflammation, supporting a novel mechanism by which donor immune cells regulate T-cell activation and GVHD in allogeneic BMT.
Collapse
Affiliation(s)
- Jingru Zhu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
- Department of Oncology, Xiangya Hospital, Central South University (CSU), Changsha, People's Republic of China
| | - Yitong Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
- Department of Oncology, Xiangya Hospital, Central South University (CSU), Changsha, People's Republic of China
| | - Jingxia Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
- Department of Oncology, Xiangya Hospital, Central South University (CSU), Changsha, People's Republic of China
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pankoj Kumar Das
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Hanwen Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Tenzin Passang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Jian Ming Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Tamas Nagy
- Comparative Pathology Laboratory, Department of Pathology, College of Veterinary Medicine, University of Georgia, Atlanta, GA
| | - Khanjan Gandhi
- Bioinformatics & Systems Biology Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Cynthia R Giver
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | | | - Yiwen Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | | | - Shuhua Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - John D Roback
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
5
|
Ramhorst R, Grasso E, Vota D, Gori S, Hauk V, Paparini D, Calo G, Leirós CP. From decidualization to pregnancy progression: An overview of immune and metabolic effects of VIP. Am J Reprod Immunol 2022; 88:e13601. [DOI: 10.1111/aji.13601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/22/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Rosanna Ramhorst
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Esteban Grasso
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Daiana Vota
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Soledad Gori
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Vanesa Hauk
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Daniel Paparini
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Guillermina Calo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| | - Claudia Pérez Leirós
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN‐CONICET) Laboratorio de Inmunofarmacología Universidad de Buenos Aires (UBA) Buenos Aires Argentina
| |
Collapse
|
6
|
Unique molecular characteristics and microglial origin of Kv1.3 channel-positive brain myeloid cells in Alzheimer's disease. Proc Natl Acad Sci U S A 2021; 118:2013545118. [PMID: 33649184 DOI: 10.1073/pnas.2013545118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Kv1.3 potassium channels, expressed by proinflammatory central nervous system mononuclear phagocytes (CNS-MPs), are promising therapeutic targets for modulating neuroinflammation in Alzheimer's disease (AD). The molecular characteristics of Kv1.3-high CNS-MPs and their cellular origin from microglia or CNS-infiltrating monocytes are unclear. While Kv1.3 blockade reduces amyloid beta (Aβ) burden in mouse models, the downstream immune effects on molecular profiles of CNS-MPs remain unknown. We show that functional Kv1.3 channels are selectively expressed by a subset of CD11b+CD45+ CNS-MPs acutely isolated from an Aβ mouse model (5xFAD) as well as fresh postmortem human AD brain. Transcriptomic profiling of purified CD11b+Kv1.3+ CNS-MPs, CD11b+CD45int Kv1.3neg microglia, and peripheral monocytes from 5xFAD mice revealed that Kv1.3-high CNS-MPs highly express canonical microglial markers (Tmem119, P2ry12) and are distinct from peripheral Ly6chigh/Ly6clow monocytes. Unlike homeostatic microglia, Kv1.3-high CNS-MPs express relatively lower levels of homeostatic genes, higher levels of CD11c, and increased levels of glutamatergic transcripts, potentially representing phagocytic uptake of neuronal elements. Using irradiation bone marrow CD45.1/CD45.2 chimerism in 5xFAD mice, we show that Kv1.3+ CNS-MPs originate from microglia and not blood-derived monocytes. We show that Kv1.3 channels regulate membrane potential and early signaling events in microglia. Finally, in vivo blockade of Kv1.3 channels in 5xFAD mice by ShK-223 reduced Aβ burden, increased CD11c+ CNS-MPs, and expression of phagocytic genes while suppressing proinflammatory genes (IL1b). Our results confirm the microglial origin and identify unique molecular features of Kv1.3-expressing CNS-MPs. In addition, we provide evidence for CNS immunomodulation by Kv1.3 blockers in AD mouse models resulting in a prophagocytic phenotype.
Collapse
|
7
|
Gallino L, Hauk V, Fernández L, Soczewski E, Gori S, Grasso E, Calo G, Saraco N, Berensztein E, Waschek JA, Pérez Leirós C, Ramhorst R. VIP Promotes Recruitment of Tregs to the Uterine-Placental Interface During the Peri-Implantation Period to Sustain a Tolerogenic Microenvironment. Front Immunol 2020; 10:2907. [PMID: 31969877 PMCID: PMC6960177 DOI: 10.3389/fimmu.2019.02907] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/26/2019] [Indexed: 01/02/2023] Open
Abstract
Uterine receptivity and embryo implantation are two main processes that need a finely regulated balance between pro-inflammatory and tolerogenic mediators to allow a successful pregnancy. The neuroimmune peptide vasoactive intestinal peptide (VIP) is a key regulator, and it is involved in the induction of regulatory T cells (Tregs), which are crucial in both processes. Here, we analyzed the ability of endogenous and exogenous VIP to sustain a tolerogenic microenvironment during the peri-implantation period, particularly focusing on Treg recruitment. Wild-type (WT) and VIP-deficient mice [heterozygous (HT, +/-), knockout (KO, -/-)], and FOXP3-knock-in-GFP mice either pregnant or in estrus were used. During the day of estrus, we found significant histological differences between the uterus of WT mice vs. VIP-deficient mice, with the latter exhibiting undetectable levels of FOXP3 expression, decreased expression of interleukin (IL)-10, and vascular endothelial growth factor (VEGF)c, and increased gene expression of the Th17 proinflammatory transcription factor RORγt. To study the implantation window, we mated WT and VIP (+/-) females with WT males and observed altered FOXP3, VEGFc, IL-10, and transforming growth factor (TGF)β gene expression at the implantation sites at day 5.5 (d5.5), demonstrating a more inflammatory environment in VIP (+/-) vs. VIP (+/+) females. A similar molecular profile was observed at implantation sites of WT × WT mice treated with VIP antagonist at d3.5. We then examined the ability GFP-sorted CD4+ cells from FOXP3-GFP females to migrate toward conditioned media (CM) obtained from d5.5 implantation sites cultured in the absence/presence of VIP or VIP antagonist. VIP treatment increased CD4+FOXP3+ and decreased CD4+ total cell migration towards implantation sites, and VIP antagonist prevented these effects. Finally, we performed adoptive cell transfer of Tregs (sorted from FOXP3-GFP females) in VIP-deficient-mice, and we observed that FOXP3-GFP cells were mainly recruited into the uterus/implantation sites compared to all other tested tissues. In addition, after Treg transfer, we found an increase in IL-10 expression and VEGFc in HT females and allowed embryo implantation in KO females. In conclusion, VIP contributes to a local tolerogenic response necessary for successful pregnancy, preventing the development of a hostile uterine microenvironment for implantation by the selective recruitment of Tregs during the peri-implantation period.
Collapse
Affiliation(s)
- Lucila Gallino
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanesa Hauk
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Fernández
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elizabeth Soczewski
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Soledad Gori
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Esteban Grasso
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermina Calo
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora Saraco
- Servicio de Endocrinología, Hospital Pediátrico Dr. Juan P. Garrahan, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Esperanza Berensztein
- Servicio de Endocrinología, Hospital Pediátrico Dr. Juan P. Garrahan, CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - James A Waschek
- Department of Psychiatry and Biobehavioral Sciences, The David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Claudia Pérez Leirós
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Laboratorio de Inmunofarmacología, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
8
|
Immunomodulatory Roles of PACAP and VIP: Lessons from Knockout Mice. J Mol Neurosci 2018; 66:102-113. [PMID: 30105629 DOI: 10.1007/s12031-018-1150-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023]
Abstract
A bidirectional cross-talk is established between the nervous and immune systems through common mediators including neuropeptides, neurotransmitters, and cytokines. Among these, PACAP and VIP are two highly related neuropeptides widely distributed in the organism with purported immunomodulatory actions. Due to their well-known anti-inflammatory properties, administration of these peptides has proven to be beneficial in models of acute and chronic inflammatory diseases. Nevertheless, the relevance of the endogenous source of these peptides in the modulation of immune responses remains to be elucidated. The development of transgenic mice with specific deletions in the genes coding for these neuropeptides (Vip and Adcyap1) or for their G-protein-coupled receptors VPAC1, VPAC2, and PAC1 (Vipr1, Vipr2, Adcyap1r1) has allowed to address this question, underscoring the complexity of the immunoregulatory properties of PACAP and VIP. The goal of this review is to integrate the existing information on the immune phenotypes of mice deficient for PACAP, VIP, or their receptors, to provide a global view on the roles of these endogenous neuropeptides during immunological health and disease.
Collapse
|
9
|
Forghani P, Petersen CT, Waller EK. Activation of VIP signaling enhances immunosuppressive effect of MDSCs on CMV-induced adaptive immunity. Oncotarget 2017; 8:81873-81879. [PMID: 29137229 PMCID: PMC5669855 DOI: 10.18632/oncotarget.20704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is recognized as a potent anti-inflammatory factor which affects both the innate and adaptive arms of the immune system. These effects include, but are not limited to, inhibition of T cell proliferation and disruption of immune homeostasis. Myeloid-derived suppressor cells (MDSC) are an immune regulatory cell type that has been described in settings of cancer and infectious disease._Here we demonstrate a reduced circulating monocytic MDSCs in the VIP -/-vs. wild type MCMV. VIP-/- MDSCs secretes less NO upon stimulation with LPS and interferon that relatively lose the ability to suppress T cells activation in vitro compared to wild type MDSCs._Considering the importance of VIP in immunomodulation, the possible effect of VIP in the suppressive function of MDSC populations following CMV infection remains unknown. We describe the possible role of VIP in the regulation of anti-CMV activity of T cells through the activation of MDSCs.
Collapse
Affiliation(s)
- Parvin Forghani
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Christopher T Petersen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Li JM, Petersen CT, Li JX, Panjwani R, Chandra DJ, Giver CR, Blazar BR, Waller EK. Modulation of Immune Checkpoints and Graft-versus-Leukemia in Allogeneic Transplants by Antagonizing Vasoactive Intestinal Peptide Signaling. Cancer Res 2016; 76:6802-6815. [PMID: 27671676 DOI: 10.1158/0008-5472.can-16-0427] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 08/16/2016] [Accepted: 09/11/2016] [Indexed: 12/23/2022]
Abstract
The goal of allogeneic bone marrow transplantation (allo-BMT) is elimination of leukemia cells through the graft-versus-leukemia (GvL) activity of donor cells, while limiting graft-versus-host disease (GvHD). Immune checkpoint pathways regulate GvL and GvHD activities, but blocking antibodies or genetic inactivation of these pathways can cause lethal GVHD. Vasoactive intestinal peptide (VIP) is an immunosuppressive neuropeptide that regulates coinhibitory pathways; its role in allo-BMT has not been studied. We found VIP transiently expressed in donor NK, NK-T, dendritic cells, and T cells after allo transplant, as well as host leukocytes. A peptide antagonist of VIP signaling (VIPhyb) increased T-cell proliferation in vitro and reduced IL10 expression in donor T cells. Treatment of allo-BMT recipients with VIPhyb, or transplanting donor grafts lacking VIP (VIP-KO), activated donor T-cells in lymphoid organs, reduced T-cell homing to GvHD target organs, and enhanced GvL without increasing GvHD in multiple allo-BMT models. Genetic or ex vivo depletion of donor NK cells or CD8+ T cells from allografts abrogated the VIPhyb-enhanced GvL activity. VIPhyb treatment led to downregulation of PD-1 and PD-L1 expression on donor immune cells, increased effector molecule expression, and expanded oligoclonal CD8+ T cells that protected secondary allo transplant recipients from leukemia. Blocking VIP signaling thus represents a novel pharmacologic approach to separate GvL from GvHD and enhance adaptive T-cell responses to leukemia-associated antigens in allo-BMT. Cancer Res; 76(23); 6802-15. ©2016 AACR.
Collapse
Affiliation(s)
- Jian-Ming Li
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | | | - Jing-Xia Li
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Reema Panjwani
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Daniel J Chandra
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Cynthia R Giver
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Edmund K Waller
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.
| |
Collapse
|
11
|
Ramhorst R, Grasso E, Paparini D, Hauk V, Gallino L, Calo G, Vota D, Pérez Leirós C. Decoding the chemokine network that links leukocytes with decidual cells and the trophoblast during early implantation. Cell Adh Migr 2016; 10:197-207. [PMID: 26891097 DOI: 10.1080/19336918.2015.1135285] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chemokine network is central to the innate and adaptive immunity and entails a variety of proteins and membrane receptors that control physiological processes such as wound healing, angiogenesis, embryo growth and development. During early pregnancy, the chemokine network coordinates not only the recruitment of different leukocyte populations to generate the maternal-placental interface, but also constitutes an additional checkpoint for tissue homeostasis maintenance. The normal switch from a pro-inflammatory to an anti-inflammatory predominant microenvironment characteristic of the post-implantation stage requires redundant immune tolerance circuits triggered by key master regulators. In this review we will focus on the recruitment and conditioning of maternal immune cells to the uterus at the early implantation period with special interest on high plasticity macrophages and dendritic cells and their ability to induce regulatory T cells. We will also point to putative immunomodulatory polypeptides involved in immune homeostasis maintenance at the maternal-placental interface.
Collapse
Affiliation(s)
- Rosanna Ramhorst
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Esteban Grasso
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Daniel Paparini
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Vanesa Hauk
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Lucila Gallino
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Guillermina Calo
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Daiana Vota
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| | - Claudia Pérez Leirós
- a Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET , Buenos Aires , Argentina
| |
Collapse
|
12
|
Gorgoglione B, Carpio Y, Secombes CJ, Taylor NGH, Lugo JM, Estrada MP. Viral and bacterial septicaemic infections modulate the expression of PACAP splicing variants and VIP/PACAP receptors in brown trout immune organs. FISH & SHELLFISH IMMUNOLOGY 2015; 47:923-932. [PMID: 26481517 DOI: 10.1016/j.fsi.2015.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 06/05/2023]
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and PACAP-Related Peptide (PRP) are structurally similar peptides encoded in the same transcripts. Their transcription has been detected not only in the brain but also in a wide range of peripheral tissues, even including organs of the immune system. PACAP exerts pleiotropic activities through G-protein coupled membrane receptors: the PACAP-specific PAC-1 and the VPAC-1 and VPAC-2 receptors that exhibit similar affinities for the Vasoactive Intestinal Peptide (VIP) and PACAP. Recent findings added PACAP and its receptors to the growing list of mediators that allow cross-talk between the nervous, endocrine and immune systems in fish. In this study the expression of genes encoding for PACAP and PRP, as well as VIP/PACAP receptors was studied in laboratory-reared brown trout (Salmo trutta) after septicaemic infections. Respectively Viral Haemorrhagic Septicaemia Virus (VHSV-Ia) or the Gram-negative bacterium Yersinia ruckeri (ser. O1 - biot. 2) were used in infection challenges. Kidney and spleen, the teleost main lymphopoietic organs, were sampled during the first two weeks post-infection. RT-qPCR analysis assessed specific pathogens burden and gene expression levels. PACAP and PRP transcription in each organ was positively correlated to the respective pathogen burden, assessed targeting the VHSV-glycoprotein or Y. ruckeri 16S rRNA. Results showed as the transcription of PACAP splicing variants and VIP/PACAP receptors is modulated in these organs during an acute viral and bacterial septicaemic infections in brown trout. These gene expression results provide clues as to how the PACAP system is modulated in fish, confirming an involvement during active immune responses elicited by both viral and bacterial aetiological agents. However, further experimental evidence is still required to fully elucidate and characterize the role of PACAP and PRP for an efficient immune response against pathogens.
Collapse
Affiliation(s)
- Bartolomeo Gorgoglione
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Scotland, UK; CEFAS Weymouth Laboratory, Weymouth, England, UK
| | - Yamila Carpio
- Aquatic Biotechnology Project, Centre for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Scotland, UK
| | | | - Juana María Lugo
- Aquatic Biotechnology Project, Centre for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mario Pablo Estrada
- Aquatic Biotechnology Project, Centre for Genetic Engineering and Biotechnology, Havana, Cuba.
| |
Collapse
|
13
|
Fraccaroli L, Grasso E, Hauk V, Paparini D, Soczewski E, Mor G, Pérez Leirós C, Ramhorst R. VIP boosts regulatory T cell induction by trophoblast cells in an in vitro model of trophoblast-maternal leukocyte interaction. J Leukoc Biol 2015; 98:49-58. [PMID: 25877932 DOI: 10.1189/jlb.1a1014-492rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/28/2015] [Indexed: 12/17/2022] Open
Abstract
Inducible regulatory T cells (Tregs) exert a timely and efficient immunosuppressive action at the critical peri-implantation stage essential for maternal tolerance to the conceptus. Vasoactive intestinal peptide (VIP) promotes anti-inflammatory and tolerogenic profiles through binding to VIP receptors on immune cells. We evaluated whether VIP produced by trophoblast cells induces Tregs during the early interaction of maternal leukocytes with trophoblast cells, thus contributing to maternal tolerance. We used an in vitro model of maternal leukocyte-trophoblast cell interaction represented by cocultures of fertile women's PBMCs with a human trophoblast cell line (Swan-71) and evaluated the effect of VIP added exogenously and of the endogenous polypeptide. VIP increased the frequency of CD4(+)CD25(+)FoxP3(+) cells after coculture, and these cells were able to suppress the maternal alloresponse. VIP also increased the frequency of CD4(+)IL10(+) and CD4(+)TGFβ(+) cells, but it did not modulate IFN-γ or IL-17 production. Swan-71 secreted VIP, and their coculture with maternal PBMCs significantly increased the frequency of Tregs. This effect was even more pronounced if the trophoblast cells had been pretreated with VIP. In both situations, the VIP antagonist prevented the increase in the frequency of CD4(+)Foxp3(+) cells, reflecting a specific effect of the polypeptide after the interaction with Swan-71 cells. Finally, the increase in CD4(+)CD25(+)FoxP3(+) frequency was prevented by an anti-TGF-β Ab and a VIP antagonist. These results suggest that VIP could have an active role in the immunoregulatory processes operating in the maternal-placental interface by contributing to the induction of Tregs through a mechanism involving TGF-β1.
Collapse
Affiliation(s)
- Laura Fraccaroli
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Esteban Grasso
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Vanesa Hauk
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Daniel Paparini
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Elizabeth Soczewski
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Gil Mor
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- *Laboratory of Immunopharmacology, University of Buenos Aires School of Sciences, IQUIBICEN-CONICET (National Research Council), Buenos Aires, Argentina; Reproductive Immunology Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA; and University of Buenos Aires School of Medicine, Buenos Aires, Argentina
| |
Collapse
|
14
|
The effect of SCF and ouabain on small intestinal motility dysfunction induced by gastric cancer peritoneal metastasis. Clin Exp Metastasis 2015; 32:267-77. [PMID: 25689893 DOI: 10.1007/s10585-015-9702-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 01/26/2015] [Indexed: 12/13/2022]
Abstract
The interstitial cells of Cajal (ICCs) play an important role in maintaining the normal function of gastrointestinal dynamics. In our previous study, we reported that, in advanced gastric cancer, the frequency of bowel movement is always reduced, due in part to the decreased number of ICCs. To investigate the impact of ICCs in gastric cancer, we established a mouse model of gastric cancer peritoneal metastasis using SGC-7901 gastric adenocarcinoma cells and their supernatant. Then, stem cell factor (SCF) and ouabain were used as therapeutic agents to improve gut dynamics. Our data showed that, compared with the normal mice, treatment with SGC-7901 cells and their supernatant led to a significant reduction of the muscle layer thickness, a decreased number of ICCs, broadened gaps between ICCs and surrounding cells, degeneration and necrosis of smooth muscle cells (SMCs), and infiltration of inflammatory cells. In contrast to SGC-7901 cell and supernatant treatment, SCF intervention caused mild submucosal edema and mitochondrial proliferation in the ICCs and SMCs. Additionally, ouabain treatment led to inflammatory cells infiltration into the submucosa and a decreased volume of ICCs. In conclusion, our data illustrated that, under the condition of gastric cancer peritoneal metastasis, the dysfunction of intestinal peristalsis may be related to pathological changes in ICCs. Moreover, we demonstrated that SCF treatment may help to improve intestinal dynamics by regulating the number and function of ICCs.
Collapse
|
15
|
Li G, Wu K, Tao K, Lu X, Ma J, Mao Z, Li H, Shi L, Li J, Niu Y, Xiang F, Wang G. Vasoactive intestinal peptide induces CD14+HLA-DR‑/low myeloid-derived suppressor cells in gastric cancer. Mol Med Rep 2015; 12:760-8. [PMID: 25695487 DOI: 10.3892/mmr.2015.3374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 01/29/2015] [Indexed: 11/06/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of cells, which have been revealed to inhibit T-cell responses in tumor-bearing mice. In addition, a number of immune suppressive mechanisms have linked MDSCs and the development of human cancer. However, the role of MDSCs in human gastric cancer tissue remains to be elucidated as specific markers are lacking. Therefore, the aim of the present study was to investigate the frequency and immune suppressive function of MDSCs denoted in the present study as cluster of differentiation 14 (CD14)+human leukocyte antigen (HLA)-DR-/low in gastric cancer patients. In the present study, MDSCs were directly isolated and characterized from the tumor and adjacent normal tissue of gastric cancer patients. Functional analysis of the CD14+HLA-DR-/low MDSCs co-cultured with allogeneic CD4+ T cells were performed and compared with controls. In addition, the interferon-γ (IFN-γ) and interleukin (IL)-2 production was compared in order to investigate the capacity of vasoactive intestinal peptide (VIP) to induce CD14+HLA-DR(-/low) MDSC-mediated CD4+ T-cell dysfunction and whether IL-10 secretion is involved in this mechanism. As a result, the quantity of CD14+HLA-DR(-/low) cells in tumor tissue from gastric cancer patients was significantly higher than that in the adjacent normal tissue. In addition, CD14+HLA-DR-/low MDSCs isolated from tumor tissue were observed to inhibit the CD4+ T-cells' immune responses in comparison with those from the adjacent normal tissue. Furthermore, VIP was able to induce the differentiation of CD14+ mononuclear cells isolated from healthy donor peripheral blood mononuclear cells into activated MDSC cells. Of note, the immunosuppressive effect of VIP-induced CD14+HLA-DR(-/low) MDSCs on CD4+ T cells was mediated by IL-10 secretion, which was demonstrated in the subsequent decrease of IFN-γ and IL-2 production. In conclusion, CD14+HLA-DR(-/low) cells were significantly increased in gastric cancer tissue and were shown to have a critical role in CD4+T-cell immunosuppression. In addition, VIP as a novel cytokine may induce the differentiation of CD14+ mononuclear cells towards CD14+HLA-DR(-/low) MDSCs. An improved understanding of phenotypic heterogeneity and the mechanism of generation of MDSCs in gastric cancer patients is important in the design of effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Gang Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ke Wu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kaixiong Tao
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaoming Lu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jianhua Ma
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhengqiang Mao
- Department of Surgery Oncology, Central Hospital, Xinxiang, Henan 453000, P.R. China
| | - Hang Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Liang Shi
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Li
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yanfeng Niu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fan Xiang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guobin Wang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
16
|
Ganea D, Hooper KM, Kong W. The neuropeptide vasoactive intestinal peptide: direct effects on immune cells and involvement in inflammatory and autoimmune diseases. Acta Physiol (Oxf) 2015; 213:442-52. [PMID: 25422088 DOI: 10.1111/apha.12427] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/13/2014] [Accepted: 11/21/2014] [Indexed: 12/13/2022]
Abstract
Neuropeptides represent an important category of endogenous contributors to the establishment and maintenance of immune deviation in the immune-privileged organs such as the CNS and in the control of acute inflammation in the peripheral immune organs. Vasoactive intestinal peptide (VIP) is a major immunoregulatory neuropeptide widely distributed in the central and peripheral nervous system. In addition to neurones, VIP is synthesized by immune cells which also express VIP receptors. Here, we review the current information on VIP production and VIP-receptor-mediated effects in the immune system, the role of endogenous and exogenous VIP in inflammatory and autoimmune disorders and the present and future VIP therapeutic approaches.
Collapse
Affiliation(s)
- D. Ganea
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| | - K. M. Hooper
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| | - W. Kong
- Department of Microbiology and Immunology; Temple University School of Medicine; Philadelphia PA USA
| |
Collapse
|
17
|
A 2.5-kilobase deletion containing a cluster of nine microRNAs in the latency-associated-transcript locus of the pseudorabies virus affects the host response of porcine trigeminal ganglia during established latency. J Virol 2014; 89:428-42. [PMID: 25320324 DOI: 10.1128/jvi.02181-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED The alphaherpesvirus pseudorabies virus (PrV) establishes latency primarily in neurons of trigeminal ganglia when only the transcription of the latency-associated transcript (LAT) locus is detected. Eleven microRNAs (miRNAs) cluster within the LAT, suggesting a role in establishment and/or maintenance of latency. We generated a mutant (M) PrV deleted of nine miRNA genes which displayed properties that were almost identical to those of the parental PrV wild type (WT) during propagation in vitro. Fifteen pigs were experimentally infected with either WT or M virus or were mock infected. Similar levels of virus excretion and host antibody response were observed in all infected animals. At 62 days postinfection, trigeminal ganglia were excised and profiled by deep sequencing and quantitative RT-PCR. Latency was established in all infected animals without evidence of viral reactivation, demonstrating that miRNAs are not essential for this process. Lower levels of the large latency transcript (LLT) were found in ganglia infected by M PrV than in those infected by WT PrV. All PrV miRNAs were expressed, with highest expression observed for prv-miR-LLT1, prv-miR-LLT2 (in WT ganglia), and prv-miR-LLT10 (in both WT and M ganglia). No evidence of differentially expressed porcine miRNAs was found. Fifty-four porcine genes were differentially expressed between WT, M, and control ganglia. Both viruses triggered a strong host immune response, but in M ganglia gene upregulation was prevalent. Pathway analyses indicated that several biofunctions, including those related to cell-mediated immune response and the migration of dendritic cells, were impaired in M ganglia. These findings are consistent with a function of the LAT locus in the modulation of host response for maintaining a latent state. IMPORTANCE This study provides a thorough reference on the establishment of latency by PrV in its natural host, the pig. Our results corroborate the evidence obtained from the study of several LAT mutants of other alphaherpesviruses encoding miRNAs from their LAT regions. Neither PrV miRNA expression nor high LLT expression levels are essential to achieve latency in trigeminal ganglia. Once latency is established by PrV, the only remarkable differences are found in the pattern of host response. This indicates that, as in herpes simplex virus, LAT functions as an immune evasion locus.
Collapse
|
18
|
Waschek JA. VIP and PACAP: neuropeptide modulators of CNS inflammation, injury, and repair. Br J Pharmacol 2014; 169:512-23. [PMID: 23517078 DOI: 10.1111/bph.12181] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/26/2013] [Accepted: 03/08/2013] [Indexed: 01/14/2023] Open
Abstract
Inflammatory processes play both regenerative and destructive roles in multiple sclerosis, stroke, CNS trauma, amyotrophic lateral sclerosis and aging-related neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's. Endogenous defence mechanisms against these pathologies include those that are directly neuroprotective, and those that modulate the expression of inflammatory mediators in microglia, astrocytes, and invading inflammatory cells. While a number of mechanisms and molecules have been identified that can directly promote neuronal survival, less is known about how the brain protects itself from harmful inflammation, and further, how it co-opts the healing function of the immune system to promote CNS repair. The two closely related neuroprotective peptides, vasoactive intestinal peptide (VIP) and pituitary adenylyl cyclase-activating peptide (PACAP), which are up-regulated in neurons and immune cells after injury and/or inflammation, are known to protect neurons, but also exert powerful in vivo immunomodulatory actions, which are primarily anti-inflammatory. These peptide actions are mediated by high-affinity receptors expressed not only on neurons, but also astrocytes, microglia and peripheral inflammatory cells. Well-established immunomodulatory actions of these peptides are to inhibit macrophage and microglia production and release of inflammatory mediators such as TNF-α and IFN-γ, and polarization of T-cell responses away from Th1 and Th17, and towards a Th2 phenotype. More recent studies have revealed that these peptides can also promote the production of both natural and inducible subsets of regulatory T-cells. The neuroprotective and immunomodulatory actions of VIP and PACAP suggest that receptors for these peptides may be therapeutic targets for neurodegenerative and neuroinflammatory diseases and other forms of CNS injury.
Collapse
Affiliation(s)
- J A Waschek
- Department of Psychiatry and Semel Institute, University of California at Los Angeles, Los Angeles, CA 90095-7332, USA.
| |
Collapse
|
19
|
Hossain MS, Ramachandiran S, Gewirtz AT, Waller EK. Recombinant TLR5 agonist CBLB502 promotes NK cell-mediated anti-CMV immunity in mice. PLoS One 2014; 9:e96165. [PMID: 24879439 PMCID: PMC4039429 DOI: 10.1371/journal.pone.0096165] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/04/2014] [Indexed: 12/02/2022] Open
Abstract
Prior work using allogeneic bone marrow transplantation (allo-BMT) models showed that peritransplant administration of flagellin, a toll-like receptor 5 (TLR5) agonist protected murine allo-BMT recipients from CMV infection while limiting graft-vs-host disease (GvHD). However, the mechanism by which flagellin-TLR5 interaction promotes anti-CMV immunity was not defined. Here, we investigated the anti-CMV immunity of NK cells in C57BL/6 (B6) mice treated with a highly purified cGMP grade recombinant flagellin variant CBLB502 (rflagellin) followed by murine CMV (mCMV) infection. A single dose of rflagellin administered to mice between 48 to 72 hours prior to MCMV infection resulted in optimal protection from mCMV lethality. Anti-mCMV immunity in rflagellin-treated mice correlated with a significantly reduced liver viral load and increased numbers of Ly49H+ and Ly49D+ activated cytotoxic NK cells. Additionally, the increased anti-mCMV immunity of NK cells was directly correlated with increased numbers of IFN-γ, granzyme B- and CD107a producing NK cells following mCMV infection. rFlagellin-induced anti-mCMV immunity was TLR5-dependent as rflagellin-treated TLR5 KO mice had ∼10-fold increased liver viral load compared with rflagellin-treated WT B6 mice. However, the increased anti-mCMV immunity of NK cells in rflagellin-treated mice is regulated indirectly as mouse NK cells do not express TLR5. Collectively, these data suggest that rflagellin treatment indirectly leads to activation of NK cells, which may be an important adjunct benefit of administering rflagellin in allo-BMT recipients.
Collapse
Affiliation(s)
- Mohammad S. Hossain
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Sampath Ramachandiran
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Andrew T. Gewirtz
- Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Division of Stem Cell and Bone Marrow Transplantation, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
20
|
Fraccaroli L, Grasso E, Hauk V, Cortelezzi M, Pérez Leirós C, Ramhorst R. Contribution of vasoactive intestinal peptide to immune homeostasis in trophoblast-maternal leukocyte interaction under LPS stimulation. Neuroimmunomodulation 2014; 21:21-30. [PMID: 24135863 DOI: 10.1159/000355039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/09/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The maternal-fetal interface is a unique immunological site that generates an adequate microenvironment during pregnancy, recognizing and eliminating infections and tolerating the trophoblast/placenta unit. For that purpose, trophoblast cells display several tolerogenic mechanisms to allow fetal survival, such as production of the neuropeptide vasoactive intestinal peptide (VIP). Here we investigated the contribution of VIP to maintain homeostasis at the maternal-placental interface under lipopolysaccharide (LPS) stimulation. METHODS We performed cocultures between trophoblast cells (Swan-71 cell line) and maternal leukocytes obtained from fertile women as an in vitro model of maternal-placental interaction, and we focused on the effects of LPS on the modulation of VIP and their receptors (VPAC1 and VPAC2). RESULTS VIP could prevent the upregulation of IL-6, MCP-1, and nitrite production and maintain the production of IL-10 and TGF-β under LPS (10 µg/ml) stimulation after 48 h of coculture. To gain deeper insight into the mechanisms of how VIP could contribute to a tolerogenic microenvironment even in the presence of LPS, we investigated VIP production by maternal leukocytes and observed a significant increase in the frequency of CD4+VIP+ cells after interaction with Swan-71 cells in the presence of LPS. LPS increased VIP and inducible receptor VPAC2 expression directly on trophoblast cells in a dose- and time-dependent manner. CONCLUSIONS The present results suggest that VIP might act as an additional homeostatic mechanism during early stages at the maternal-placental interface to control exacerbated inflammatory responses such as the ones observed in intrauterine infections.
Collapse
Affiliation(s)
- Laura Fraccaroli
- Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires and IQUIBICEN-CONICET (National Research Council of Science and Technology, CONICET), Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
21
|
Nerve-derived transmitters including peptides influence cutaneous immunology. Brain Behav Immun 2013; 34:1-10. [PMID: 23517710 PMCID: PMC3750093 DOI: 10.1016/j.bbi.2013.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/27/2013] [Accepted: 03/10/2013] [Indexed: 01/01/2023] Open
Abstract
Clinical observations suggest that the nervous and immune systems are closely related. For example, inflammatory skin disorders; such as psoriasis, atopic dermatitis, rosacea and acne; are widely believed to be exacerbated by stress. A growing body of research now suggests that neuropeptides and neurotransmitters serve as a link between these two systems. Neuropeptides and neurotransmitters are released by nerves innervating the skin to influence important actors of the immune system, such as Langerhans cells and mast cells, which are located within close anatomic proximity. Catecholamines and other sympathetic transmitters that are released in response to activation of the sympathetic nervous system are also able to reach the skin and affect immune cells. Neuropeptides appear to direct the outcome of Langerhans cell antigen presentation with regard to the subtypes of Th cells generated and neuropeptides induce the degranulation of mast cells, among other effects. Additionally, endothelial cells, which release many inflammatory mediators and express cell surface molecules that allow leukocytes to exit the bloodstream, appear to be regulated by certain neuropeptides and transmitters. This review focuses on the evidence that products of nerves have important regulatory activities on antigen presentation, mast cell function and endothelial cell biology. These activities are highly likely to have clinical and therapeutic relevance.
Collapse
|
22
|
VIPhyb, an antagonist of vasoactive intestinal peptide receptor, enhances cellular antiviral immunity in murine cytomegalovirus infected mice. PLoS One 2013; 8:e63381. [PMID: 23723978 PMCID: PMC3664580 DOI: 10.1371/journal.pone.0063381] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/03/2013] [Indexed: 12/22/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is a neuropeptide hormone that suppresses Th1-mediated cellular immunity. We previously reported that VIP-knockout (VIP-KO) mice have enhanced cellular immune responses and increased survival following murine cytomegalovirus (mCMV) infection in C57BL/6 mice. In this study, we tested whether treatment with a VIP receptor antagonistic peptide protects C57BL/6 and BALB/c mice from mCMV-infection. One week of daily subcutaneous injections of VIPhyb was non-toxic and did not alter frequencies of immune cell subsets in non-infected mice. VIPhyb administration to mCMV-infected C57BL/6 and BALB/c mice markedly enhanced survival, viral clearance, and reduced liver and lung pathology compared with saline-treated controls. The numbers of effector/memory CD8+ T-cells and mature NK cells were increased in VIPhyb-treated mice compared with PBS-treated groups. Pharmacological blockade of VIP-receptor binding or genetic blockade of VIP-signaling prevented the up-regulation of PD-L1 and PD-1 expression on DC and activated CD8+ T-cells, respectively, in mCMV-infected mice, and enhanced CD80, CD86, and MHC-II expression on conventional and plasmacytoid DC. VIPhyb-treatment increased type-I IFN synthesis, numbers of IFN-γ- and TNF-α-expressing NK cells and T-cells, and the numbers of mCMV-M45 epitope-peptide-MHC-I tetramer CD8+ T-cells following mCMV infection. VIP-treatment lowered the percentage of Treg cells in spleens compared with PBS-treated WT mice following mCMV infection, while significantly decreasing levels of serum VEGF induced by mCMV-infection. The mice in all treated groups exhibited similar levels of anti-mCMV antibody titers. Short-term administration of a VIP-receptor antagonist represents a novel approach to enhance innate and adaptive cellular immunity in a murine model of CMV infection.
Collapse
|
23
|
Pérez Leirós C, Ramhorst R. Tolerance induction at the early maternal-placental interface through selective cell recruitment and targeting by immune polypeptides. Am J Reprod Immunol 2013; 69:359-68. [PMID: 23405982 DOI: 10.1111/aji.12087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/07/2013] [Indexed: 12/12/2022] Open
Abstract
Pregnancy challenges immune cells and immunomodulatory circuits of the mother and the developing fetus to dynamically adapt to each other in an homeostatic and tolerant environment for fetal growth. This entails the coordination of multiple cellular processes all devoted to accommodate and nourish the fetus while protecting the mother from endogenous and exogenous threatens. From the earliest stages of pregnancy, several strategies to efficiently communicate immune and trophoblast cells within the interface or at a distance were identified and chemokines might act at on different targets through direct or indirect mechanisms. Here, we briefly review some mechanisms of T regulatory cell recruitment to the early maternal-placental interfaces to accomplish immunotolerance and homeostatic control and we discuss evidence on two locally released polypeptides, RANTES (regulated on activation, normal, T-cell expressed, and secreted) and vasoactive intestinal peptide (VIP), as novel contributors to the multiplicity of immune tolerant responses and uterine quiescence requirements.
Collapse
Affiliation(s)
- Claudia Pérez Leirós
- Immunopharmacology Laboratory, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET, Buenos Aires, Argentina
| | | |
Collapse
|
24
|
Pharmacological inhibition of VIP signaling enhances antiviral immunity and improves survival in murine cytomegalovirus-infected allogeneic bone marrow transplant recipients. Blood 2013; 121:2347-51. [PMID: 23325838 DOI: 10.1182/blood-2012-06-437640] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cytomegalovirus (CMV) infection following allogeneic bone marrow transplant (allo-BMT) is controlled by donor-derived cellular immunity. Vasoactive intestinal peptide (VIP) suppresses Th1 immunity. We hypothesized that blocking VIP-signaling would enhance anti-CMV immunity in murine recipients of allo-BMT. Recipients were transplanted with bone marrow (BM) and T-cells from major histocompatibility complex (MHC)-mismatched VIP-knockout (KO) or wild-type donors, and treated with 7 daily subcutaneous injections of VIPhyb (peptidic VIP-antagonist) or phosphate-buffered saline (PBS). Genetic and pharmacological blockade of VIP-signaling protected allo-BMT recipients from lethal murine CMV (mCMV) infection, improving survival without increasing graft-versus-host disease. Mice treated with VIPhyb or transplanted with VIP-KO allografts had significantly lower viral loads, increased numbers of mCMV-M45-peptide-MHC-tetramer(+) CD8(+) T-cells, with lower PD-1 expression, and enhanced primary and secondary cellular immune responses after mCMV infection than did PBS-treated mice. These results demonstrate that administration of a VIP antagonist after allo-BMT is a promising safely therapeutic approach to enhance antiviral cellular immunity.
Collapse
|
25
|
Delgado M. Immunoregulatory Neuropeptides. HANDBOOK OF BIOLOGICALLY ACTIVE PEPTIDES 2013:640-648. [DOI: 10.1016/b978-0-12-385095-9.00087-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
26
|
VIP deficient mice exhibit resistance to lipopolysaccharide induced endotoxemia with an intrinsic defect in proinflammatory cellular responses. PLoS One 2012; 7:e36922. [PMID: 22615845 PMCID: PMC3355097 DOI: 10.1371/journal.pone.0036922] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 04/17/2012] [Indexed: 12/20/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide with immunomodulatory properties. The administration of this peptide has been shown to have beneficial effects in murine models of inflammatory diseases including septic shock, rheumatoid arthritis, multiple sclerosis (MS) and Crohn's disease. However, the role of the endogenous peptide in inflammatory disease remains obscure because VIP-deficient mice were recently found to exhibit profound resistance in a model of MS. In the present study, we analyzed the response of female VIP deficient (KO) mice to intraperitoneal lipopolysaccharide (LPS) administration. We observed significant resistance to LPS in VIP KO mice, as evidenced by lower mortality and reduced tissue damage. The increased survival was associated with decreased levels of proinflammatory cytokines (TNFα, IL-6 and IL-12) in sera and peritoneal suspensions of these mice. Moreover, the expression of TNFα and IL-6 mRNA was reduced in peritoneal cells, spleens and lungs from LPS-treated VIP KO vs. WT mice, suggesting that the resistance might be mediated by an intrinsic defect in the responsiveness of immune cells to endotoxin. In agreement with this hypothesis, peritoneal cells isolated from VIP KO naive mice produced lower levels of proinflammatory cytokines in response to LPS in vitro. Finally, decreased NF-κB pathway activity in peritoneal cells was observed both in vivo and in vitro, as determined by assay of phosphorylated I-κB. The results demonstrate that female VIP KO mice exhibit resistance to LPS-induced shock, explainable in part by the presence of an intrinsic defect in the responsiveness of inflammatory cells to endotoxin.
Collapse
|
27
|
Delgado M, Ganea D. Vasoactive intestinal peptide: a neuropeptide with pleiotropic immune functions. Amino Acids 2011; 45:25-39. [PMID: 22139413 DOI: 10.1007/s00726-011-1184-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/22/2011] [Indexed: 01/07/2023]
Abstract
Vasoactive intestinal peptide (VIP), a 28-amino acid neuropeptide/neurotransmitter, is widely distributed in both the central and peripheral nervous system. VIP is released by both neurons and immune cells. Various cell types, including immune cells, express VIP receptors. VIP has pleiotropic effects as a neurotransmitter, immune regulator, vasodilator and secretagogue. This review is focused on VIP production and effects on immune cells, VIP receptor signaling as related to immune functions, and the involvement of VIP in inflammatory and autoimmune disorders. The review addresses present clinical use of VIP and future therapeutic directions.
Collapse
Affiliation(s)
- Mario Delgado
- Instituto de Parasitologia y Biomedicina, IPBLN-CSIC, Granada, Spain
| | | |
Collapse
|
28
|
Szema AM, Hamidi SA, Golightly MG, Rueb TP, Chen JJ. VIP Regulates the Development & Proliferation of Treg in vivo in spleen. Allergy Asthma Clin Immunol 2011; 7:19. [PMID: 22126441 PMCID: PMC3286388 DOI: 10.1186/1710-1492-7-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 11/29/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mounting evidence supports a key role for VIP as an anti-inflammatory agent and promoter of immune tolerance. It suppresses TNF-α and other inflammatory cytokines and chemokines, upregulates anti-inflammatory IL-10, and promotes immune tolerant cells called T regulatory (Treg) cells. VIP KO mice have recently been demonstrated to have spontaneous airway and pulmonary perivascular inflammatory responses, as part of asthma-like and pulmonary hypertension phenotypes, respectively. Both inflammatory responses are correctable with VIP. Focusing on this model, we have now investigated the influence of VIP not only on inflammatory cells but also on Treg cells. METHODS Using flow cytometric analysis, we examined the relative preponderance of CD25+CD4+ cells and anti-inflammatory Treg cells, in extracts of thymus and spleen from VIP KO mice (5 VIP KO; 5 VIP KO+ VIP; 10 wild-type). This method allowed antibody-based flow cytometric identification of Treg cells using surface markers CD25 and CD4, along with the: 1) intracellular activation marker FoxP3; and 2) Helios, which distinguishes cells of thymic versus splenic derivation. CONCLUSIONS Deletion of the VIP gene results in: 1) CD25+CD4- cell accumulation in the thymus, which is corrected by VIP treatment; 2) more Treg in thymus lacking Foxp3 expression, suggesting VIP is necessary for immune tolerance; and, 3) a tendency towards deficiency of Treg cells in the spleen, which is normalized by VIP treatment. Treg lacking Helios are induced by VIP intrasplenically rather than by migration from the thymus. These results confirm the dual role of VIP as an anti-inflammatory and immune tolerance-promoting agent.
Collapse
Affiliation(s)
- Anthony M Szema
- Stony Brook University School of Medicine, Department of Medicine, 100 Nicolls Road, Stony Brook, NY 11794, USA.
| | | | | | | | | |
Collapse
|