1
|
Chhabra A, Thakur ML. Theragnostic Radionuclide Pairs for Prostate Cancer Management: 64Cu/ 67Cu, Can Be a Budding Hot Duo. Biomedicines 2022; 10:2787. [PMID: 36359312 PMCID: PMC9687163 DOI: 10.3390/biomedicines10112787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 08/26/2023] Open
Abstract
Prostate cancer (PCa) is one of the preeminent causes of mortality in men worldwide. Theragnostic, a combination of therapy and diagnostic, using radionuclide pairs to diagnose and treat disease, has been shown to be a promising approach for combating PCa. In PCa patients, bone is one of the most common sites of metastases, and about 90% of patients develop bone metastases. This review focuses on (i) clinically translated theragnostic radionuclide pairs for the management of PCa, (ii) radionuclide therapy of bone metastases in PCa, and (iii) a special emphasis on emerging theragnostic radionuclide pair, Copper-64/Copper-67 (64Cu/67Cu) for managing the disease.
Collapse
Affiliation(s)
- Anupriya Chhabra
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mathew L. Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
2
|
Hawley E, Mia K, Yusuf M, Swanson KC, Doetkott C, Dorsam GP. Messenger RNA Gene Expression Screening of VIP and PACAP Neuropeptides and Their Endogenous Receptors in Ruminants. BIOLOGY 2022; 11:biology11101512. [PMID: 36290416 PMCID: PMC9598725 DOI: 10.3390/biology11101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022]
Abstract
Vasoactive Intestinal Peptide (VIP) and Pituitary Adenylate-Cyclase-Activating Peptide (PACAP) are anti-inflammatory neuropeptides that play important roles in human and rodent gut microbiota homeostasis and host immunity. Pharmacologically regulating these neuropeptides is expected to have significant health and feed efficiency benefits for agriculturally relevant animals. However, their expression profile in ruminant tissues is not well characterized. To this end, we screened for VIP and PACAP neuropeptides and their endogenous GPCRs using 15 different tissues from wethers and steers by RT-qPCR. Our results revealed relatively similar expression profiles for both VIP and PACAP neuropeptide ligands in the brain and intestinal tissue of both species. In contrast, the tissue expression profiles for VPAC1, VPAC2, and PAC1 were more widespread and disparate, with VPAC1 being the most diversely expressed receptor with mRNA detection in the brain and throughout the gastrointestinal tract. These data are an important first step to allow for future investigations regarding the VIP and PACAP signaling pathways in livestock ruminant species.
Collapse
Affiliation(s)
- Emma Hawley
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Kafi Mia
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Mustapha Yusuf
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Kendall C. Swanson
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Curt Doetkott
- Information Technology Services, North Dakota State University, Fargo, ND 58102, USA
| | - Glenn P. Dorsam
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND 58102, USA
- Correspondence:
| |
Collapse
|
3
|
Langer I, Jeandriens J, Couvineau A, Sanmukh S, Latek D. Signal Transduction by VIP and PACAP Receptors. Biomedicines 2022; 10:406. [PMID: 35203615 PMCID: PMC8962308 DOI: 10.3390/biomedicines10020406] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 02/05/2023] Open
Abstract
Homeostasis of the human immune system is regulated by many cellular components, including two neuropeptides, VIP and PACAP, primary stimuli for three class B G protein-coupled receptors, VPAC1, VPAC2, and PAC1. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) regulate intestinal motility and secretion and influence the functioning of the endocrine and immune systems. Inhibition of VIP and PACAP receptors is an emerging concept for new pharmacotherapies for chronic inflammation and cancer, while activation of their receptors provides neuroprotection. A small number of known active compounds for these receptors still impose limitations on their use in therapeutics. Recent cryo-EM structures of VPAC1 and PAC1 receptors in their agonist-bound active state have provided insights regarding their mechanism of activation. Here, we describe major molecular switches of VPAC1, VPAC2, and PAC1 that may act as triggers for receptor activation and compare them with similar non-covalent interactions changing upon activation that were observed for other GPCRs. Interhelical interactions in VIP and PACAP receptors that are important for agonist binding and/or activation provide a molecular basis for the design of novel selective drugs demonstrating anti-inflammatory, anti-cancer, and neuroprotective effects. The impact of genetic variants of VIP, PACAP, and their receptors on signalling mediated by endogenous agonists is also described. This sequence diversity resulting from gene splicing has a significant impact on agonist selectivity and potency as well as on the signalling properties of VIP and PACAP receptors.
Collapse
Affiliation(s)
- Ingrid Langer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles, B-1070 Brussels, Belgium; (I.L.); (J.J.)
| | - Jérôme Jeandriens
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université libre de Bruxelles, B-1070 Brussels, Belgium; (I.L.); (J.J.)
| | - Alain Couvineau
- UMR 1149 Inserm, Centre de Recherche sur l’Inflammation (CRI), Université de Paris, 75018 Paris, France;
| | - Swapnil Sanmukh
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland;
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland;
| |
Collapse
|
4
|
Ericsson AC, Bains M, McAdams Z, Daniels J, Busi SB, Waschek JA, Dorsam GP. The G Protein-Coupled Receptor, VPAC1, Mediates Vasoactive Intestinal Peptide-Dependent Functional Homeostasis of the Gut Microbiota. GASTRO HEP ADVANCES 2022; 1:253-264. [PMID: 36910129 PMCID: PMC9997614 DOI: 10.1016/j.gastha.2021.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Vasoactive intestinal peptide (VIP) is a neuropeptide involved in the regulation of feeding behavior and circadian rhythms, metabolism, and immunity. Previous studies revealed the homeostatic effects of VIP signaling on the gut microbiota. VIP-deficient mice demonstrate a gut microbiota dysbiosis characterized by reduced α-diversity and decreased relative abundance (RA) of Gram-positive Firmicutes. However, the mechanism by which VIP signaling affects changes in the microbiota is unknown. METHODS To investigate the role of the 2 cognate G protein-coupled receptors for VIP (VPAC1 and VPAC2) in VIP-mediated homeostasis of the microbiota, fecal samples from VPAC1- and VPAC2-deficient, heterozygous, and wild-type littermate mice were assessed via targeted amplicon sequencing. Their microbiota profiles were additionally compared with microbiota from VIP-deficient, heterozygous, and wild-type littermates, where genotype-dependent changes in the composition and predicted function of each cohort were compared. RESULTS While wild-type mice in each line differed in α-diversity and β-diversity, consistent changes in both metrics were observed in VIP-deficient and VPAC1-deficient mice. This includes a dramatic reduction in α-diversity, increased RA of Proteobacteria and Bacteroidetes, and decreased RA of Lachnospiraceae, Ruminococcaceae, Muribaculaceae, and Rikenellaceae. Specific amplicon sequence variants and predicted functions found to differ significantly based on VIP or VPAC1 genotype were concordant in their directions of change. Multiplatform predicted functional profiling suggested a defective VIP-VPAC1 axis was associated with reduced amino acid degradation along with reduced quinol and quinone biosynthesis. Furthermore, alterations in predicted functions include increased sugar degradation, nitrate reduction, and fatty acid biosynthetic pathways, among other changes. CONCLUSION We conclude that VIP signaling through VPAC1 is critical for the maintenance of normal function of the gut microbiota.
Collapse
Affiliation(s)
- Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri Metagenomics Center, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Manpreet Bains
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota
| | - Zachary McAdams
- Department of Veterinary Pathobiology, University of Missouri Metagenomics Center, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Justin Daniels
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota
| | - Susheel B. Busi
- Department of Systems Ecology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - James A. Waschek
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, University of California, Los Angeles, Los Angeles, California
| | - Glenn P. Dorsam
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
5
|
Bains M, Laney C, Wolfe AE, Orr M, Waschek JA, Ericsson AC, Dorsam GP. Vasoactive Intestinal Peptide Deficiency Is Associated With Altered Gut Microbiota Communities in Male and Female C57BL/6 Mice. Front Microbiol 2019; 10:2689. [PMID: 31849864 PMCID: PMC6900961 DOI: 10.3389/fmicb.2019.02689] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is crucial for gastrointestinal tract (GIT) health. VIP sustains GIT homeostasis through maintenance of the intestinal epithelial barrier and acts as a potent anti-inflammatory mediator that contributes to gut bacterial tolerance. Based on these biological functions by VIP, we hypothesized that its deficiency would alter gut microbial ecology. To this end, fecal samples from male and female VIP+/+, VIP+/-, and VIP-/- littermates (n = 47) were collected and 16S rRNA sequencing was conducted. Our data revealed significant changes in bacterial composition, biodiversity, and weight loss from VIP-/- mice compared to VIP+/+ and VIP+/- littermates, irrespective of sex. The gut bacteria compositional changes observed in VIP-/- mice was consistent with gut microbial structure changes reported for certain inflammatory and autoimmune disorders. Moreover, predicted functional changes by PICRUSt software suggested an energy surplus within the altered microbiota from VIP-/- mice. These data support that VIP plays an important role in maintaining microbiota balance, biodiversity, and GIT function, and its genetic removal results in significant gut microbiota restructuring and weight loss.
Collapse
Affiliation(s)
- Manpreet Bains
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, ND, United States
| | - Caleb Laney
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, ND, United States
| | - Annie E. Wolfe
- Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Megan Orr
- Department of Statistics, College of Science and Math, North Dakota State University, Fargo, ND, United States
| | - James A. Waschek
- Intellectual and Developmental Disabilities Research Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aaron C. Ericsson
- Metagenomics Center, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Glenn P. Dorsam
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
6
|
Atanasova KR, Reznikov LR. Neuropeptides in asthma, chronic obstructive pulmonary disease and cystic fibrosis. Respir Res 2018; 19:149. [PMID: 30081920 PMCID: PMC6090699 DOI: 10.1186/s12931-018-0846-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
The nervous system mediates key airway protective behaviors, including cough, mucus secretion, and airway smooth muscle contraction. Thus, its involvement and potential involvement in several airway diseases has become increasingly recognized. In the current review, we focus on the contribution of select neuropeptides in three distinct airway diseases: asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis. We present data on some well-studied neuropeptides, as well as call attention to a few that have not received much consideration. Because mucus hypersecretion and mucus obstruction are common features of many airway diseases, we place special emphasis on the contribution of neuropeptides to mucus secretion. Finally, we highlight evidence implicating involvement of neuropeptides in mucus phenotypes in asthma, COPD and cystic fibrosis, as well as bring to light knowledge that is still lacking in the field.
Collapse
Affiliation(s)
- Kalina R Atanasova
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA
| | - Leah R Reznikov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA.
| |
Collapse
|
7
|
Petersen CT, Li JM, Waller EK. Administration of a vasoactive intestinal peptide antagonist enhances the autologous anti-leukemia T cell response in murine models of acute leukemia. Oncoimmunology 2017. [PMID: 28638725 DOI: 10.1080/2162402x.2017.1304336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) is a neuroendocrine peptide hormone that has potent anti-inflammatory activities. VIP signaling through its receptor VPAC1 on T cells leads to reduced proliferation and a reduction in pro-inflammatory cytokine secretion. We report here that inhibition of the VIP pathway with a peptide antagonist significantly enhances a T-cell-dependent, autologous anti-leukemia response in murine models of acute myeloid leukemia and T lymphoblastic leukemia. Subcutaneous administration of the VIP antagonist, VIPhyb, resulted in reduced tumor burden and significantly enhanced survival (30-50% survival) over vehicle-treated controls (0-20% survival). The T cells in mice treated with VIPhyb expressed lower levels of the co-inhibitory PD-1 and secreted higher levels of IFNγ. Furthermore, T cells from VIPhyb-treated survivors were protective against C1498 following adoptive transfer. These data highlight the potential for the VIP pathway as a novel target for immunomodulation in settings of hematological malignancies.
Collapse
Affiliation(s)
- Christopher T Petersen
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jian-Ming Li
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Edmund K Waller
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Galoian K, Patel P. Epigenetic control of cancer by neuropeptides. Biomed Rep 2016; 6:3-7. [PMID: 28123699 DOI: 10.3892/br.2016.804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/26/2016] [Indexed: 12/26/2022] Open
Abstract
Neuropeptides act as neurohormones, neurotransmitters and/or neuromodulators. Neuropeptides maintain physiological homeostasis and are paramount in molecular mechanisms of disease progression and regulation, including in cancer. Neuropeptides, by their definition, originate and are secreted from the neuronal cells, they are able to signal to neighboring cells or are released into the blood flow, if they act as neurohormones. The majority of neuropeptides exert their functions through G protein-coupled receptors, with certain exceptions. Although previous studies indicate that neuropeptides function in supporting proliferation of malignant cells in many types of solid tumor, the antitumorigenic action of the neuropeptides and their receptors, for example, in gastric cancers and chondrosarcoma, were also reported. It is known that epigenetically modified chromatin regulates molecular mechanisms involved in gene expression and malignant progression. The epigenetic modifications are genetically heritable, although they do not cause changes in DNA sequence. DNA methylation, histone modifications and miRNA expression are subject to those modifications. While there is substantial data on epigenetic regulation of neuropeptides, the epigenetic control of cancer by neuropeptides is considered to be uncharted territory. The aim of the current review is to describe the involvement of neuropeptides in the epigenetic machinery of cancer based on data obtained from our laboratory and from other authors.
Collapse
Affiliation(s)
- Karina Galoian
- Department of Orthopedics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Parthik Patel
- Department of Orthopedics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
9
|
Seoane IV, Ortiz AM, Piris L, Lamana A, Juarranz Y, García-Vicuña R, González-Álvaro I, Gomariz RP, Martínez C. Clinical Relevance of VPAC1 Receptor Expression in Early Arthritis: Association with IL-6 and Disease Activity. PLoS One 2016; 11:e0149141. [PMID: 26881970 PMCID: PMC4755558 DOI: 10.1371/journal.pone.0149141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/27/2016] [Indexed: 11/19/2022] Open
Abstract
Background The vasoactive intestinal peptide (VIP) receptors VPAC1 and VPAC2 mediate anti-inflammatory and immunoregulatory responses in rheumatoid arthritis (RA). Data on the expression of these receptors could complement clinical assessment in the management of RA. Our goal was to investigate the correlation between expression of both receptors and the 28-Joint Disease Activity Score (DAS28) in peripheral blood mononuclear cells (PBMCs) from patients with early arthritis (EA). We also measured expression of IL-6 to evaluate the association between VIP receptors and systemic inflammation. Methods We analyzed 250 blood samples collected at any of the 5 scheduled follow-up visits from 125 patients enrolled in the Princesa Early Arthritis Register Longitudinal study. Samples from 22 healthy donors were also analyzed. Sociodemographic, clinical, and therapeutic data were systematically recorded. mRNA expression levels were determined using real-time PCR. Then, longitudinal multivariate analyses were performed. Results PBMCs from EA patients showed significantly higher expression of VPAC2 receptors at baseline compared to healthy donors (p<0.001). With time, however, VPAC2 expression tended to be significantly lower while VPAC1 receptor expression increased in correlation with a reduction in DAS28 index. Our results reveal that more severe inflammation, based on high levels of IL-6, is associated with lower expression of VPAC1 (p<0.001) and conversely with increased expression of VPAC2 (p<0.001). A major finding of this study is that expression of VPAC1 is lower in patients with increased disease activity (p = 0.001), thus making it possible to differentiate between patients with various degrees of clinical disease activity. Conclusion Patients with more severe inflammation and higher disease activity show lower levels of VPAC1 expression, which is associated with patient-reported impairment. Therefore, VPAC1 is a biological marker in EA.
Collapse
MESH Headings
- Adult
- Aged
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/physiopathology
- Case-Control Studies
- Female
- Gene Expression Regulation
- Humans
- Interleukin-6/blood
- Interleukin-6/metabolism
- Leukocytes, Mononuclear/metabolism
- Longitudinal Studies
- Male
- Middle Aged
- Receptors, Vasoactive Intestinal Peptide, Type II/blood
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I/blood
- Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
- Severity of Illness Index
- Signal Transduction
- Social Class
- Time Factors
- Vasoactive Intestinal Peptide/metabolism
Collapse
Affiliation(s)
- Iria V. Seoane
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana M. Ortiz
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa, Madrid, Spain
| | - Lorena Piris
- Unidad de Apoyo Metodológico, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa, Madrid, Spain
| | - Amalia Lamana
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa, Madrid, Spain
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosario García-Vicuña
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa, Madrid, Spain
| | - Isidoro González-Álvaro
- Servicio de Reumatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria la Princesa, Madrid, Spain
| | - Rosa P. Gomariz
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Carmen Martínez
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
10
|
Jimeno R, Leceta J, Garín M, Ortiz AM, Mellado M, Rodríguez-Frade JM, Martínez C, Pérez-García S, Gomariz RP, Juarranz Y. Th17 polarization of memory Th cells in early arthritis: the vasoactive intestinal peptide effect. J Leukoc Biol 2015; 98:257-69. [PMID: 25957307 DOI: 10.1189/jlb.3a0714-327r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 04/12/2015] [Indexed: 01/15/2023] Open
Abstract
Several studies in humans indicate the implication of Th17 cells in RA. Therapies targeting their pathogenicity, as well as their plasticity to the Th17/1 phenotype, could ameliorate the progression of the pathology. The neuroendocrine environment has a major impact on the differentiation of lymphoid cells. VIP is present in the microenvironment of the joint, and its known therapeutic effects are supported by several studies on RA. We examine the ability of VIP to modulate the differentiation of Th17 cells. Peripheral blood CD4(+)CD45RO(+) T cells from HD and eRA patients were expanded under Th17-polarizing conditions in the presence of TGF-β. After 7 days, the higher IL-17A, IL-21, and IL-9 levels and lower IL-22 levels indicate the nonpathogenic profile for Th17 cells in HD. In contrast, Th17 cells from eRA patients produced significantly more IL-22 and IFN-γ, and these cells show a more Th17/1 profile, indicating a pathogenic phenotype. Interestingly, when VIP was present in the Th17 conditioned medium, increased levels of IL-10 and IL-9 were detected in HD and eRA patients. VIP also reduced the levels of IL-22 in eRA patients. These data suggest that VIP reduces the pathogenic profile of the Th17-polarized cells. This effect was accompanied by an increased in the Treg/Th17 profile, as shown by the increase levels of Foxp3. In conclusion, this report addresses a novel and interesting question on the effect of VIP on human Th17 cells and adds clinical relevance by analyzing, in parallel, HD and eRA patients.
Collapse
Affiliation(s)
- Rebeca Jimeno
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Javier Leceta
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marina Garín
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Ana M Ortiz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mario Mellado
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jose Miguel Rodríguez-Frade
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carmen Martínez
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Rosa P Gomariz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de *Biología y Medicina, Universidad Complutense de Madrid, Madrid, Spain; División de Terapias Innovadoras en el S. Hematopoyético, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad Mixta de Terapias Avanzadas, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/IIS Fundación Jiménez Díaz, Madrid, Spain; Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa, Madrid, Spain; and Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
11
|
Vasoactive intestinal peptide maintains the nonpathogenic profile of human th17-polarized cells. J Mol Neurosci 2014; 54:512-25. [PMID: 24805298 DOI: 10.1007/s12031-014-0318-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/28/2014] [Indexed: 01/01/2023]
Abstract
The cytokine microenvironment modulates CD4 T cell differentiation causing the shift of naïve CD4 T cells into different cell subsets. This process is also regulated by modulators such as vasoactive intestinal peptide (VIP), a neuropeptide with known immunomodulatory properties on CD4 T cells that exert this action through specific receptors, vasoactive intestinal peptide receptor (VPAC)1 and VPAC2. Our results show that the pattern of VIP receptors expression ratio is modified during Th17 differentiation. In this report, we evaluate the capacity of VIP to modulate naïve human cells into Th17 cells in vitro by analyzing their functional phenotype. The presence of VIP maintains the nonpathogenic profile of Th17-polarized cells, increases the proliferation rate, and decreases their Th1 potential. VIP induces the upregulation of the STAT3 gene interaction with the VPAC1 receptor during the onset of Th17 differentiation. Moreover, RAR-related orphan receptor C (RORC), RAR-related orphan receptor A (RORA), and interleukin (IL)-17A genes are upregulated in the presence of VIP through interaction with VPAC1 and VPAC2 receptors. Interestingly, VIP induces the expression of the IL-23R gene through interaction with the VPAC2 receptor during the expansion phase. This is the first report that describes the differentiation of naïve human T cells to Th17-polarized cells in the presence of VIP and demonstrates how this differentiation regulates the expression of the VIP receptors.
Collapse
|
12
|
Dovat S. Ikaros in hematopoiesis and leukemia. World J Biol Chem 2011; 2:105-7. [PMID: 21765974 PMCID: PMC3135855 DOI: 10.4331/wjbc.v2.i6.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 06/10/2011] [Accepted: 06/17/2011] [Indexed: 02/05/2023] Open
Abstract
Ikaros is a gene whose activity is essential for normal hematopoiesis. Ikaros acts as a master regulator of lymphoid and myeloid development as well as a tumor suppressor. In cells, Ikaros regulates gene expression via chromatin remodeling. During the past 15 years tremendous advances have been made in understanding the role of Ikaros in hematopoiesis and leukemogenesis. In this Topic Highlights series of reviews, several groups of international experts in this field summarize the experimental data that is shaping the emerging picture of Ikaros function at the biochemical and cellular levels. The articles provide detailed analyses of recent scientific advancements and present models that will serve as a basis for future studies aimed at developing a better understanding of normal hematopoiesis and hematological malignancies and at accelerating the application of this knowledge in clinical practice.
Collapse
Affiliation(s)
- Sinisa Dovat
- Sinisa Dovat, Department of Pediatrics, Pennsylvania State University, College of Medicine, Hershey, PA 17033-0850, United States
| |
Collapse
|