1
|
Herrera GA, Teng J, Zeng C, Del Pozo-Yauner L, Liu B, Turbat-Herrera EA. Identifying gene expression and cellular pathways involved in glomerular AL-amyloidosis and correlation with experimental data: seeking novel therapeutic interventions. Ultrastruct Pathol 2025; 49:216-234. [PMID: 39985165 DOI: 10.1080/01913123.2025.2468708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
The prognosis of myeloma is based on controlling the plasma cell burden and thus, management of the production of monoclonal light chains has improved considerably, expanding survival and quality of life. However, the effects of the monoclonal light chains in the various organs result in alterations that may lead to renal failure. There is a crucial need to ameliorate or abolish renal damage. Organ-based therapies must be developed. Glomerulopathic light chains interact with mesangial cells using the SORL1 receptor and downstream effects lead to divergent mesangial alterations. While the multi-step process occurring when amyloidogenic light chains interact with mesangial cells has been elucidated in the laboratory, gene expression profiles and activated cellular pathways in human glomeruli have not been probed. Mesangial cells from five renal biopsies at different stages of glomerular amyloidosis were interrogated using spatial transcriptomics and compared with those from normal biopsy controls to identify cellular pathways and gene expression changes. The two most significant statistically overexpressed genes (FDR <0.05) when comparing control, early vs late cases were heat shock protein 90AB1 and HSPB1, known to be involved in protein misfolding and aggregation. The overexpressed genes exercise function and regulation over cellular pathways promoting apoptosis, vesicular transport, metalloproteinase activation, collagen degradation, gap junction degradation, GTPase cycle activation, and organelle biogenesis. This data confirmed the results previously reached in the research laboratory. Spatial transcriptomics demonstrated uniquely activated genes and cellular pathways in mesangial cells involved in the initiation and progression of glomerular amyloidosis, uncovering novel genes and new therapeutic targets.
Collapse
Affiliation(s)
| | - Jiamin Teng
- Department of Pathology, University of South Alabama, Mobile, AL, USA
| | - Chun Zeng
- Department of Pathology, University of South Alabama, Mobile, AL, USA
| | | | - Bing Liu
- Department of Pathology, University of South Alabama, Mobile, AL, USA
| | - Elba A Turbat-Herrera
- Department of Pathology, University of South Alabama, Mobile, AL, USA
- Mitchell Cancer Center Department of Pathology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
2
|
Peng HY, Chang CW, Wu PH, Li LJ, Lin YL, Hsiao M, Chang JY, Chang PMH, Lee HL, Chang WM. Oral Cancer-Derived miR-762 Suppresses T-Cell Infiltration and Activation by Horizontal Inhibition of CXCR3 Expression. Int J Mol Sci 2025; 26:1077. [PMID: 39940842 PMCID: PMC11817288 DOI: 10.3390/ijms26031077] [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: 11/11/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an immune-cold tumor characterized by an immunosuppressive microenvironment with low cytotoxic activity to eliminate tumor cells. Tumor escape is one of the initial steps in cancer development. Understanding the underlying mechanisms of cancer escape can help researchers develop new treatment strategies. In this study, we prove the oral oncogenic miR-762 can suppress T-cell recruitment and cytotoxic activation in the tumor microenvironment (TME) through horizontal transmission from OSCC cells to adaptive immune T cells. Public database analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine the prognosis and expression of miR-762 in OSCC. T-cell activation by flow cytometry, qRT-PCR, IL-12 secretion, and T-cell recruitment and cytotoxicity abilities were conducted in the miR-762 manipulation T-cell and OSCC-T-cell co-culture system. A luciferase reporter and CXCR3 protein expression were also carried out to validate the direct interaction between CXCR3 and microRNA (miR)-762. This horizontal transmission of miR-762 directly suppresses CXCR3 expression in T cells, inhibiting CXCR3-induced T-cell migration and downstream T-cell cytotoxic activity by disrupting AKT activation. Additionally, miR-762 transmission suppressed T-cell activation marker expression, T-cell proliferation, IL-12 secretion, and T-cell cytotoxicity. In conclusion, our findings reveal a novel miR-762/CXCR3 axis that regulates the immunosuppressive microenvironment in OSCC and may be a potential RNA-targeted therapeutic approach to restore the anti-tumor immune response in OSCC treatment.
Collapse
Affiliation(s)
- Hsuan-Yu Peng
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Chia-Wei Chang
- Division of Family Dentistry, Department of Dentistry, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Ping-Hsiu Wu
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei 110, Taiwan
| | - Li-Jie Li
- Ph.D. Program of School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Yu-Lung Lin
- The Ph.D. Program for Translational Medicine, College of Medical Sciences and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Jang-Yang Chang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Min Chang
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
3
|
Cini JK, Kenney RT, Dexter S, McAndrew SJ, Eraslan RN, Brody R, Rezac DJ, Boohaker R, Lapi SE, Mohan P. SON-1010: an albumin-binding IL-12 fusion protein that improves cytokine half-life, targets tumors, and enhances therapeutic efficacy. Front Immunol 2024; 15:1493257. [PMID: 39697343 PMCID: PMC11652653 DOI: 10.3389/fimmu.2024.1493257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Background Cytokines have been promising cancer immunotherapeutics for decades, yet only two are licensed to date. Interleukin-12 (IL-12) is a potent regulator of cell-mediated immunity that activates NK cells and interferon-γ (IFNγ) production. It plays a central role in multiple pathways that can enhance cancer cell death and modify the tumor microenvironment (TME). Attempts to dose rIL-12 were initially successful but IFNγ toxicity in Phase 2 complicated further development in the late 1990s. Since then, better dosing strategies have been developed, but none have achieved the level of cancer control seen in preclinical models. We set out to develop a novel strategy to deliver fully functional IL-12 and other biologics to the TME by binding albumin, taking advantage of its ability to be concentrated and retained in the tumor. Methods Single-chain variable fragments (scFv) were identified from a human phage display library that bound human, mouse, and cynomolgus macaque serum albumin, both at physiologic and acidic conditions. These were taken through a series of steps to identify strongly binding molecules that don't interfere with the normal physiology of albumin to bind FcRn, giving it prolonged half-life in serum, along with SPARC/GP60, which allows albumin to target the TME. A final molecule was chosen and a single mutation was made that minimizes the potential for immunogenicity. This fully human albumin-binding (FHAB®) domain was characterized and manufacturing processes were developed to bring the first drug candidate into the clinic. Results Once identified, the murine form of mIL12-FHAB was studied preclinically to understand its mechanism of action and biodistribution. It was found to be much more efficient at blocking tumor growth compared to murine IL-12, while stimulating significant IFNγ production with minimal toxicity. SON-1010, which uses the human IL-12 sequence, passed through all of the characterization and required toxicology and is currently being studied in the clinic. Conclusions We identified and developed a platform technology with prolonged half-life that can target IL-12 and other immune modulators to the TME. Safety and efficacy are being studied using SON-1010 as monotherapy and in combination with checkpoint blockade strategies.
Collapse
Affiliation(s)
- John K. Cini
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| | | | - Susan Dexter
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| | | | | | - Rich Brody
- InfinixBio, Inc., Athens, OH, United States
| | | | | | - Suzanne E. Lapi
- Radiology, Chemistry, and Biomedical Engineering, University of Alabama, Birmingham, AL, United States
| | - Pankaj Mohan
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| |
Collapse
|
4
|
Takács L, Abuaish S, Kaňková Š, Hill M, Ullmann J, Včelák J, Monk C. Maternal negative affect in pregnancy predicts cytokine levels which in turn predict birth outcomes - A prospective longitudinal study in a low-risk population. J Affect Disord 2024; 366:345-353. [PMID: 39191312 DOI: 10.1016/j.jad.2024.08.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 08/01/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Stress and negative mood in pregnancy have been linked to less favorable birth outcomes, but the mechanisms underlying this effect remain largely unknown. We examined associations between emotions in pregnancy, pro- and anti-inflammatory cytokines (IFN-γ, IL-6, IL-8, IL-10, IL-12, IL-17, MCP-1, MIP-1β, TNF-α) and birth outcomes (gestational age at birth and birth weight) in a low-risk sample. METHODS At each trimester of pregnancy, participants (N = 74) completed the Positive and Negative Affect Schedule, Perceived Stress Scale, Edinburgh Postnatal Depression Scale, and State-Trait Anxiety Inventory. They provided blood samples in the third trimester. Multivariate regression with a reduction of dimensionality (orthogonal projection to latent structures) was used to assess associations between maternal emotions, cytokine levels, and birth outcomes. RESULTS We found significant positive associations between negative mood (depressive symptoms in the second and third trimesters and negative affect in the third trimester) and anti-inflammatory cytokine IL-10 levels, and negative associations between maternal distress in the second and third trimesters and pro-/anti-inflammatory cytokine ratios (IFN-γ/IL-10, TNF-α/IL-10 and IL-6/IL-10). Higher levels of pro-inflammatory cytokines IFN-γ, IL-12, IL-17, and TNF-α were associated with younger gestational age at birth and lower birth weight. LIMITATIONS We did not control for relevant factors such as social support, health-related behaviors, or cortisol levels. CONCLUSIONS Negative mood in mid- and late pregnancy may shift cytokine balance toward the anti-inflammatory cytokine dominance. Our results provide further evidence for the negative association between pro-inflammatory cytokines in late pregnancy and gestational age at birth/birth weight, which we observed even in a low-risk population.
Collapse
Affiliation(s)
- Lea Takács
- Department of Psychology, Faculty of Arts, Charles University, Prague, Czech Republic.
| | - Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Šárka Kaňková
- Department of Philosophy and History of Science, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martin Hill
- Department of Steroid Hormones and Proteohormones, Institute of Endocrinology, Prague, Czech Republic
| | - Jana Ullmann
- Department of Philosophy and History of Science, Faculty of Science, Charles University, Prague, Czech Republic
| | - Josef Včelák
- Department of Molecular Endocrinology, Institute of Endocrinology, Prague, Czech Republic
| | - Catherine Monk
- Department of Obstetrics & Gynecology, and Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
5
|
Vandoren R, Boeren M, Schippers J, Bartholomeus E, Mullan K, Michels N, Aerts O, Leysen J, Bervoets A, Lambert J, Leuridan E, Wens J, Peeters K, Emonds MP, Jansens H, Casanova JL, Bastard P, Suls A, Van Tendeloo V, Ponsaerts P, Delputte P, Ogunjimi B, Laukens K, Meysman P. Unraveling the Immune Signature of Herpes Zoster: Insights Into the Pathophysiology and Human Leukocyte Antigen Risk Profile. J Infect Dis 2024; 230:706-715. [PMID: 38195164 PMCID: PMC11420803 DOI: 10.1093/infdis/jiad609] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
The varicella-zoster virus (VZV) infects >95% of the population. VZV reactivation causes herpes zoster (HZ), known as shingles, primarily affecting the elderly and individuals who are immunocompromised. However, HZ can occur in otherwise healthy individuals. We analyzed the immune signature and risk profile in patients with HZ using a genome-wide association study across different UK Biobank HZ cohorts. Additionally, we conducted one of the largest HZ human leukocyte antigen association studies to date, coupled with transcriptomic analysis of pathways underlying HZ susceptibility. Our findings highlight the significance of the major histocompatibility complex locus for HZ development, identifying 5 protective and 4 risk human leukocyte antigen alleles. This demonstrates that HZ susceptibility is largely governed by variations in the major histocompatibility complex. Furthermore, functional analyses revealed the upregulation of type I interferon and adaptive immune responses. These findings provide fresh molecular insights into the pathophysiology and activation of innate and adaptive immune responses triggered by symptomatic VZV reactivation.
Collapse
Affiliation(s)
- Romi Vandoren
- Adrem Data Lab, Department of Computer Science, University of Antwerp
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Biomedical Informatics Research Network Antwerp
| | - Marlies Boeren
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Laboratory of Microbiology, Parasitology and Hygiene and Infla-Med Center of Excellence
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute
- Antwerp Center for Translational Immunology and Virology, Vaccine and Infectious Disease Institute
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute
| | - Jolien Schippers
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Antwerp Center for Translational Immunology and Virology, Vaccine and Infectious Disease Institute
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute
| | - Esther Bartholomeus
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Antwerp Center for Translational Immunology and Virology, Vaccine and Infectious Disease Institute
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute
| | - Kerry Mullan
- Adrem Data Lab, Department of Computer Science, University of Antwerp
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Biomedical Informatics Research Network Antwerp
| | - Nele Michels
- Department of Family Medicine and Population Health, Center for General Practice/Family Medicine, University of Antwerp
| | - Olivier Aerts
- Department of Dermatology, Antwerp University Hospital and University of Antwerp
| | - Julie Leysen
- Department of Dermatology, Antwerp University Hospital and University of Antwerp
| | - An Bervoets
- Department of Dermatology, Antwerp University Hospital and University of Antwerp
| | - Julien Lambert
- Department of Dermatology, Antwerp University Hospital and University of Antwerp
| | - Elke Leuridan
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp
| | - Johan Wens
- Department of Family Medicine and Population Health, Center for General Practice/Family Medicine, University of Antwerp
| | - Karin Peeters
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Antwerp Center for Translational Immunology and Virology, Vaccine and Infectious Disease Institute
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute
| | - Marie-Paule Emonds
- Histocompatibility and Immunogenetic Laboratory, Rode Kruis-Vlaanderen, Mechelen
| | - Hilde Jansens
- Department of Clinical Microbiology, Antwerp University Hospital, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris
- Imagine Institute, Paris Cité University, France
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University
- Howard Hughes Medical Institute, New York, New York
- Department of Pediatrics, Necker Hospital for Sick Children, Paris
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris
- Imagine Institute, Paris Cité University, France
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique–Hôpitaux de Paris, France
| | - Arvid Suls
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Medical Genetics, University of Antwerp and Antwerp University Hospital
| | - Viggo Van Tendeloo
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene and Infla-Med Center of Excellence
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Antwerp Center for Translational Immunology and Virology, Vaccine and Infectious Disease Institute
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute
- Department of Paediatrics, Antwerp University Hospital, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Biomedical Informatics Research Network Antwerp
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing, University of Antwerp
- Biomedical Informatics Research Network Antwerp
| |
Collapse
|
6
|
Toffoli EC, van Vliet AA, Forbes C, Arns AJ, Verheul HWM, Tuynman J, van der Vliet HJ, Spanholtz J, de Gruijl TD. Allogeneic NK cells induce the in vitro activation of monocyte-derived and conventional type-2 dendritic cells and trigger an inflammatory response under cancer-associated conditions. Clin Exp Immunol 2024; 216:159-171. [PMID: 38330230 PMCID: PMC11036108 DOI: 10.1093/cei/uxae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/27/2023] [Accepted: 02/06/2024] [Indexed: 02/10/2024] Open
Abstract
Natural killer (NK) cells are innate lymphocytes capable to recognize and kill virus-infected and cancer cells. In the past years, the use of allogeneic NK cells as anti-cancer therapy gained interest due to their ability to induce graft-versus-cancer responses without causing graft-versus-host disease and multiple protocols have been developed to produce high numbers of activated NK cells. While the ability of these cells to mediate tumor kill has been extensively studied, less is known about their capacity to influence the activity of other immune cells that may contribute to a concerted anti-tumor response in the tumor microenvironment (TME). In this study, we analyzed how an allogeneic off-the-shelf cord blood stem cell-derived NK-cell product influenced the activation of dendritic cells (DC). Crosstalk between NK cells and healthy donor monocyte-derived DC (MoDC) resulted in the release of IFNγ and TNF, MoDC activation, and the release of the T-cell-recruiting chemokines CXCL9 and CXCL10. Moreover, in the presence of prostaglandin-E2, NK cell/MoDC crosstalk antagonized the detrimental effect of IL-10 on MoDC maturation leading to higher expression of multiple (co-)stimulatory markers. The NK cells also induced activation of conventional DC2 (cDC2) and CD8+ T cells, and the release of TNF, GM-CSF, and CXCL9/10 in peripheral blood mononuclear cells of patients with metastatic colorectal cancer. The activated phenotype of MoDC/cDC2 and the increased release of pro-inflammatory cytokines and T-cell-recruiting chemokines resulting from NK cell/DC crosstalk should contribute to a more inflamed TME and may thus enhance the efficacy of T-cell-based therapies.
Collapse
Affiliation(s)
- E C Toffoli
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - A A van Vliet
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Glycostem Therapeutics, Oss, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - C Forbes
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - A J Arns
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - H W M Verheul
- Department of Medical Oncology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - J Tuynman
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - H J van der Vliet
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Lava Therapeutics, Utrecht, The Netherlands
| | - J Spanholtz
- Glycostem Therapeutics, Oss, The Netherlands
| | - T D de Gruijl
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Feng Q, Shen J, Zhang L, Wang Q, Luo S, Luo M, Chen W. Expression of IL-4 and IL-12 in the aqueous humor of patients with chronic primary angle-closure glaucoma. Front Med (Lausanne) 2024; 11:1323829. [PMID: 38651060 PMCID: PMC11033499 DOI: 10.3389/fmed.2024.1323829] [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: 10/18/2023] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Angle-closure glaucoma is a common type of glaucoma in Asian populations. However, the role of inflammatory cytokines in angle-closure glaucoma is yet to be elucidated. Therefore, this study aimed to examine the expression of interleukin-4 (IL-4) and IL-12 in the aqueous humor of patients with chronic primary angle-closure glaucoma (CPACG) and elucidate the correlations between IL-4 and IL-12 concentrations in the aqueous humor, the degree of visual field defects, and retinal nerve fiber layer (RNFL) thickness in patients with CPACG. Methods Aqueous humor samples were obtained from 31 patients diagnosed with CPACG at the Shaoxing People's Hospital between April 2022 and March 2023 and from 30 individuals with cataract (control). Based on the degree of the mean defect (MD), patients were divided into three groups: group A (MD ≤ -6dB, n= 10), group B (-6dB< MD< -12dB, n= 9), and group C (MD ≥ - 12dB, n= 12). RNFL thickness was measured using an optical coherence tomograph, and the concentrations of IL-4 and IL-12 in the aqueous humor were measured using Luminex technology. Results Aqueous humor concentration of IL-4 was significantly higher (p = 0.036) in the CPACG group than in the cataract group. However, there was no significant difference (p > 0.05) in IL- 12 concentration between the two groups. Additionally, there were no significant differences (p > 0.05) in IL-4 and IL-12 levels among patients with varying degrees of visual field defects (groups A, B, and C). Spearman's correlation analysis showed that IL-4 and IL-12 concentrations were not correlated (p > 0.05) with RNFL thickness around the optic disc and the degree of visual field defects. Discussion Conclusively, IL-4 may play an important role in the pathogenesis of CPACG. Given that IL-4 and IL-12 concentrations in the aqueous humor were not significantly correlated with RNFL thickness and the degree of visual field defects, the increase in IL-4 and IL-12 expression may not induce apoptosis and loss of retinal ganglion cells or affect RNFL thickness as well as the degree of visual field defects.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Chen
- Department of Ophthalmology, Shaoxing People's Hospital, Zhejiang University, Shaoxing, China
| |
Collapse
|
8
|
Zhang S, Liu G, Zhang Y, Wang C, Xu X, Zhao Y, Xiang Z, Wu W, Yang L, Chen J, Guo A, Chen Y. Investigation of the safety and protective efficacy of an attenuated and marker M. bovis-BoHV-1 combined vaccine in bovines. Front Immunol 2024; 15:1367253. [PMID: 38646533 PMCID: PMC11027501 DOI: 10.3389/fimmu.2024.1367253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/26/2024] [Indexed: 04/23/2024] Open
Abstract
Bovine respiratory disease (BRD) is one of the most common diseases in the cattle industry worldwide; it is caused by multiple bacterial or viral coinfections, of which Mycoplasma bovis (M. bovis) and bovine herpesvirus type 1 (BoHV-1) are the most notable pathogens. Although live vaccines have demonstrated better efficacy against BRD induced by both pathogens, there are no combined live and marker vaccines. Therefore, we developed an attenuated and marker M. bovis-BoHV-1 combined vaccine based on the M. bovis HB150 and BoHV-1 gG-/tk- strain previously constructed in our lab and evaluated in rabbits. This study aimed to further evaluate its safety and protective efficacy in cattle using different antigen ratios. After immunization, all vaccinated cattle had a normal rectal temperature and mental status without respiratory symptoms. CD4+, CD8+, and CD19+ cells significantly increased in immunized cattle and induced higher humoral and cellular immune responses, and the expression of key cytokines such as IL-4, IL-12, TNF-α, and IFN-γ can be promoted after vaccination. The 1.0 × 108 CFU of M. bovis HB150 and 1.0 × 106 TCID50 BoHV-1 gG-/tk- combined strain elicited the most antibodies while significantly increasing IgG and cellular immunity after challenge. In conclusion, the M. bovis HB150 and BoHV-1 gG-/tk- combined strain was clinically safe and protective in calves; the mix of 1.0 × 108 CFU of M. bovis HB150 and 1.0 × 106 TCID50 BoHV-1 gG-/tk- strain was most promising due to its low amount of shedding and highest humoral and cellular immune responses compared with others. This study introduces an M. bovis-BoHV-1 combined vaccine for application in the cattle industry.
Collapse
MESH Headings
- Animals
- Cattle
- Herpesvirus 1, Bovine/immunology
- Vaccines, Combined/immunology
- Vaccines, Combined/administration & dosage
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Mycoplasma bovis/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/adverse effects
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/adverse effects
- Cytokines/metabolism
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Mycoplasma Infections/prevention & control
- Mycoplasma Infections/veterinary
- Mycoplasma Infections/immunology
- Vaccines, Marker/immunology
- Vaccines, Marker/administration & dosage
- Vaccination/veterinary
- Vaccine Efficacy
- Immunity, Humoral
- Bovine Respiratory Disease Complex/prevention & control
- Bovine Respiratory Disease Complex/immunology
- Bovine Respiratory Disease Complex/virology
Collapse
Affiliation(s)
- Sen Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Guoxing Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Yisheng Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Chen Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Xiaowen Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Yuhao Zhao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Zhijie Xiang
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Wenying Wu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Li Yang
- Wuhan Keqian Biology Co., Ltd, Research and Development Department, Wuhan, China
| | - Jianguo Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Aizhen Guo
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| | - Yingyu Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan, China
| |
Collapse
|
9
|
Kenney RT, Cini JK, Dexter S, DaFonseca M, Bingham J, Kuan I, Chawla SP, Polasek TM, Lickliter J, Ryan PJ. A phase I trial of SON-1010, a tumor-targeted, interleukin-12-linked, albumin-binding cytokine, shows favorable pharmacokinetics, pharmacodynamics, and safety in healthy volunteers. Front Immunol 2024; 15:1362775. [PMID: 38487528 PMCID: PMC10937388 DOI: 10.3389/fimmu.2024.1362775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
Background The benefits of recombinant interleukin-12 (rIL-12) as a multifunctional cytokine and potential immunotherapy for cancer have been sought for decades based on its efficacy in multiple mouse models. Unexpected toxicity in the first phase 2 study required careful attention to revised dosing strategies. Despite some signs of efficacy since then, most rIL-12 clinical trials have encountered hurdles such as short terminal elimination half-life (T½), limited tumor microenvironment targeting, and substantial systemic toxicity. We developed a strategy to extend the rIL-12 T½ that depends on binding albumin in vivo to target tumor tissue, using single-chain rIL-12 linked to a fully human albumin binding (FHAB) domain (SON-1010). After initiating a dose-escalation trial in patients with cancer (SB101), a randomized, double-blind, placebo-controlled, single-ascending dose (SAD) phase 1 trial in healthy volunteers (SB102) was conducted. Methods SB102 (NCT05408572) focused on safety, tolerability, pharmacokinetic (PK), and pharmacodynamic (PD) endpoints. SON-1010 at 50-300 ng/kg or placebo administered subcutaneously on day 1 was studied at a ratio of 6:2, starting with two sentinels; participants were followed through day 29. Safety was reviewed after day 22, before enrolling the next cohort. A non-compartmental analysis of PK was performed and correlations with the PD results were explored, along with a comparison of the SON-1010 PK profile in SB101. Results Participants receiving SON-1010 at 100 ng/kg or higher tolerated the injection but generally experienced more treatment-emergent adverse effects (TEAEs) than those receiving the lowest dose. All TEAEs were transient and no other dose relationship was noted. As expected with rIL-12, initial decreases in neutrophils and lymphocytes returned to baseline by days 9-11. PK analysis showed two-compartment elimination in SB102 with mean T½ of 104 h, compared with one-compartment elimination in SB101, which correlated with prolonged but controlled and dose-related increases in interferon-gamma (IFNγ). There was no evidence of cytokine release syndrome based on minimal participant symptoms and responses observed with other cytokines. Conclusion SON-1010, a novel presentation for rIL-12, was safe and well-tolerated in healthy volunteers up to 300 ng/kg. Its extended half-life leads to a prolonged but controlled IFNγ response, which may be important for tumor control in patients. Clinical trial registration https://clinicaltrials.gov/study/NCT05408572, identifier NCT05408572.
Collapse
Affiliation(s)
| | - John K. Cini
- Sonnet BioTherapeutics, Inc, Princeton, NJ, United States
| | - Susan Dexter
- Sonnet BioTherapeutics, Inc, Princeton, NJ, United States
| | | | | | | | - Sant P. Chawla
- Sarcoma Oncology Center, Santa Monica, CA, United States
| | - Thomas M. Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, VIC, Australia
- InClin, Inc, San Mateo, CA, United States
| | | | | |
Collapse
|
10
|
Zhang S, Liu G, Wu W, Yang L, Shirani I, Guo A, Chen Y. Investigation of the Optimal Immunization Dose and Protective Efficacy of an Attenuated and Marker M. bovis-Bovine Herpesvirus Type 1 Combined Vaccine in Rabbits. Animals (Basel) 2024; 14:748. [PMID: 38473133 DOI: 10.3390/ani14050748] [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: 01/30/2024] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Bovine respiratory disease (BRD) is one of the most common diseases in the cattle industry; it is a globally prevalent multifactorial infection primarily caused by viral and bacterial coinfections. In China, Mycoplasma bovis (M. bovis) and bovine herpesvirus type 1 (BoHV-1) are the most notable pathogens associated with BRD. Our previous study attempted to combine the two vaccines and conducted a preliminary investigation of their optimal antigenic ratios. Based on this premise, the research extended its investigation by administering varying vaccine doses in a rabbit model to identify the most effective immunization dosage. After immunization, all rabbits in other immunization dose groups had a normal rectal temperature without obvious clinical symptoms. Furthermore, assays performed on the samples collected from immunized rabbits indicated that there were increased humoral and cellular immunological reactions. Moreover, the histological analysis of the lungs showed that immunized rabbits had more intact lung tissue than their unimmunized counterparts after the challenge. Additionally, there appears to be a positive correlation between the protective efficacy and the immunization dose. In conclusion, the different immunization doses of the attenuated and marker M. bovis HB150 and BoHV-1 gG-/tk- combined vaccine were clinically safe in rabbits; the mix of 2.0 × 108 CFU of M. bovis HB150 and 2.0 × 106 TCID50 BoHV-1 gG-/tk- strain was most promising due to its highest humoral and cellular immune responses and a more complete morphology of the lung tissue compared with others. These findings determined the optimal immunization dose of the attenuated and marker M. bovis HB150 and BoHV-1 gG-/tk- combined vaccine, laying a foundation for its clinical application.
Collapse
Affiliation(s)
- Sen Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| | - Guoxing Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| | - Wenying Wu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| | - Li Yang
- Wuhan Keqian Biology Co., Ltd., Wuhan 430200, China
| | - Ihsanullah Shirani
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| | - Aizhen Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| | - Yingyu Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China
| |
Collapse
|
11
|
Lee YJ, Pan Y, Lim D, Park SH, Sin SI, Kwack K, Park KY. Broccoli Cultivated with Deep Sea Water Mineral Fertilizer Enhances Anti-Cancer and Anti-Inflammatory Effects of AOM/DSS-Induced Colorectal Cancer in C57BL/6N Mice. Int J Mol Sci 2024; 25:1650. [PMID: 38338927 PMCID: PMC10855752 DOI: 10.3390/ijms25031650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
This study aimed to determine the alleviating effect of broccoli grown with deep sea water mineral (DSWM) fertilizer extracted from deep sea water on the development of colorectal cancer in C57BL/6N mice treated with AOM/DSS. Naturaldream Fertilizer Broccoli (NFB) cultured with deep sea water minerals (DSWM) showed a higher antioxidant effect and mineral content. In addition, orally administered NFB, showed a level of recovery in the colon and spleen tissues of mice compared with those in normal mice through hematoxylin and eosin (H&E) staining. Orally administered NFB showed the inhibition of the expression of inflammatory cytokine factors IL-1β, IL-6, TNF, IFN-γ, and IL-12 while increasing the expression of IL-10. Furthermore, the expression of inflammatory cytokines and NF-κB in the liver tissue was inhibited, and that of inflammatory enzymes, such as COX-2 and iNOS, was reduced. In the colon tissue, the expression of p53 and p21 associated with cell cycle arrest increased, and that of Bcl-2 associated with apoptosis decreased. Additionally, the expression of Bax, Bad, Bim, Bak, caspase 9, and caspase 3 increased, indicating enhanced activation of apoptosis-related factors. These results demonstrate that oral administration of broccoli cultivated using DSWM significantly restores spleen and colon tissues and simultaneously inhibits the NF-κB pathway while significantly decreasing cytokine expression. Moreover, by inducing cell cycle arrest and activating cell apoptosis, they also suggest alleviating AOM/DSS-induced colon cancer symptoms in C57BL/6N mice.
Collapse
Affiliation(s)
- Yeon-Jun Lee
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea; (Y.-J.L.); (Y.P.); (D.L.)
| | - Yanni Pan
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea; (Y.-J.L.); (Y.P.); (D.L.)
| | - Daewoo Lim
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea; (Y.-J.L.); (Y.P.); (D.L.)
| | - Seung-Hwan Park
- Agriculture Research Center for Carbon Neutral and Healing, Gurye-gun 57607, Republic of Korea
| | - Sin-Il Sin
- Agriculture Research Center for Carbon Neutral and Healing, Gurye-gun 57607, Republic of Korea
| | - KyuBum Kwack
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea; (Y.-J.L.); (Y.P.); (D.L.)
| | - Kun-Young Park
- Graduate School of Integrative Medicine, CHA University, Seongnam 13488, Republic of Korea
| |
Collapse
|
12
|
Oguma Y, Alessio N, Aprile D, Dezawa M, Peluso G, Di Bernardo G, Galderisi U. Meta-analysis of senescent cell secretomes to identify common and specific features of the different senescent phenotypes: a tool for developing new senotherapeutics. Cell Commun Signal 2023; 21:262. [PMID: 37770897 PMCID: PMC10537976 DOI: 10.1186/s12964-023-01280-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
DNA damage resulting from genotoxic injury can initiate cellular senescence, a state characterized by alterations in cellular metabolism, lysosomal activity, and the secretion of factors collectively known as the senescence-associated secretory phenotype (SASP). Senescence can have beneficial effects on our bodies, such as anti-cancer properties, wound healing, and tissue development, which are attributed to the SASP produced by senescent cells in their intermediate stages. However, senescence can also promote cancer and aging, primarily due to the pro-inflammatory activity of SASP.Studying senescence is complex due to various factors involved. Genotoxic stimuli cause random damage to cellular macromolecules, leading to variations in the senescent phenotype from cell to cell, despite a shared program. Furthermore, senescence is a dynamic process that cannot be analyzed as a static endpoint, adding further complexity.Investigating SASP is particularly intriguing as it reveals how a senescence process triggered in a few cells can spread to many others, resulting in either positive or negative consequences for health. In our study, we conducted a meta-analysis of the protein content of SASP obtained from different research groups, including our own. We categorized the collected omic data based on: i) cell type, ii) harmful agent, and iii) senescence stage (early and late senescence).By employing Gene Ontology and Network analysis on the omic data, we identified common and specific features of different senescent phenotypes. This research has the potential to pave the way for the development of new senotherapeutic drugs aimed at combating the negative consequences associated with the senescence process. Video Abstract.
Collapse
Affiliation(s)
- Yo Oguma
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA.
- Dip. Medicina Sperimentale, Via Luigi De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
13
|
Mandt T, Bangar A, Sauceda C, Das M, Moderbacher C, Ghani M, Webster N, Newton I. Stimulating Antitumoral Immunity by Percutaneous Cryoablation and Combination Immunoadjuvant Therapy in a Murine Model of Hepatocellular Carcinoma. J Vasc Interv Radiol 2023; 34:1516-1527.e6. [PMID: 37178816 PMCID: PMC10852103 DOI: 10.1016/j.jvir.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/18/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
PURPOSE To test the hypothesis that antitumoral immunity can be induced after cryoablation (cryo) of hepatocellular carcinoma (HCC) through coadministration of the immunostimulant CpG and an immune checkpoint (programmed cell death 1 [PD-1]) inhibitor. MATERIALS AND METHODS Sixty-three immunocompetent C57BL/6J mice were generated with 2 orthotopic HCC tumor foci: 1 for treatment and 1 to observe for antitumoral immunity. Tumors were treated with incomplete cryo alone or intratumoral CpG and/or a PD-1 inhibitor. The primary endpoint was death or when the following criteria for sacrifice were met: tumor > 1 cm (determined using ultrasound) or moribund state. Antitumoral immunity was assessed using flow cytometry and histology (tumor and liver) as well as enzyme-linked immunosorbent assay (serum). Analysis of variance was used for statistical comparisons. RESULTS At 1 week, the nonablated satellite tumor growth was reduced by 1.9-fold (P = .047) in the cryo + CpG group and by 2.8-fold (P = .007) in the cryo + CpG + PD-1 group compared with that in the cryo group. Compared with cryo alone, the time to tumor progression to endpoints was also prolonged for cryo + CpG + PD-1 and cryo + CpG mice, with log-rank hazard ratios of 0.42 (P = .031) and 0.27 (P < .001), respectively. Flow cytometry and histology showed increased cytotoxic T-cell infiltration (P = .002) and serum levels of the proinflammatory cytokine interferon-γ (P = .015) in tumors and serum of cryo + CpG mice compared with those in tumors and serum of mice treated with cryo alone. High serum levels of the anti-inflammatory cytokine tumor growth factor-β and the proangiogenesis chemokine C-X-C motif chemokine ligand 1 were correlated with a shorter time to endpoints and faster tumor growth. CONCLUSIONS Cryo combined with the immunostimulant CpG promoted cytotoxic T-cell infiltration into tumors, slowed tumor growth, and prolonged the time to progression to endpoints in an aggressive murine HCC model.
Collapse
Affiliation(s)
- Tyler Mandt
- Health Department of Radiology, University of California San Diego, San Diego
| | - Amandip Bangar
- Health Department of Radiology, University of California San Diego, San Diego
| | - Consuelo Sauceda
- Health Department of Radiology, University of California San Diego, San Diego
| | - Manasi Das
- Health Department of Radiology, University of California San Diego, San Diego
| | | | - Mansur Ghani
- Health Department of Radiology, University of California San Diego, San Diego
| | - Nicholas Webster
- San Diego Veteran's Affairs, University of California San Diego, San Diego
| | - Isabel Newton
- San Diego Veteran's Affairs, University of California San Diego, San Diego.
| |
Collapse
|
14
|
Jou E. Type 1 and type 2 cytokine-mediated immune orchestration in the tumour microenvironment and their therapeutic potential. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:474-497. [PMID: 37455828 PMCID: PMC10345208 DOI: 10.37349/etat.2023.00146] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/25/2023] [Indexed: 07/18/2023] Open
Abstract
Cancer remains the second leading cause of death worldwide despite modern breakthroughs in medicine, and novel treatments are urgently needed. The revolutionary success of immune checkpoint inhibitors in the past decade serves as proof of concept that the immune system can be effectively harnessed to treat cancer. Cytokines are small signalling proteins with critical roles in orchestrating the immune response and have become an attractive target for immunotherapy. Type 1 immune cytokines, including interferon γ (IFNγ), interleukin-12 (IL-12), and tumour necrosis factor α (TNFα), have been shown to have largely tumour suppressive roles in part through orchestrating anti-tumour immune responses mediated by natural killer (NK) cells, CD8+ T cells and T helper 1 (Th1) cells. Conversely, type 2 immunity involving group 2 innate lymphoid cells (ILC2s) and Th2 cells are involved in tissue regeneration and wound repair and are traditionally thought to have pro-tumoural effects. However, it is found that the classical type 2 immune cytokines IL-4, IL-5, IL-9, and IL-13 may have conflicting roles in cancer. Similarly, type 2 immunity-related cytokines IL-25 and IL-33 with recently characterised roles in cancer may either promote or suppress tumorigenesis in a context-dependent manner. Furthermore, type 1 cytokines IFNγ and TNFα have also been found to have pro-tumoural effects under certain circumstances, further complicating the overall picture. Therefore, the dichotomy of type 1 and type 2 cytokines inhibiting and promoting tumours respectively is not concrete, and attempts of utilising these for cancer immunotherapy must take into account all available evidence. This review provides an overview summarising the current understanding of type 1 and type 2 cytokines in tumour immunity and discusses the prospects of harnessing these for immunotherapy in light of previous and ongoing clinical trials.
Collapse
Affiliation(s)
- Eric Jou
- Queens’ College, University of Cambridge, CB3 9ET Cambridge, UK
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| |
Collapse
|
15
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
16
|
Multifunctional Modified Tumor Cell Membranes-Coated Adjuvant PTX against Melanoma. Biomolecules 2023; 13:biom13010179. [PMID: 36671563 PMCID: PMC9855842 DOI: 10.3390/biom13010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/01/2023] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Melanoma is the deadliest type of skin cancer. Anti-tumor immunotherapy has made great progress in increasing the overall survival of patients. However, many physiological barriers cause low bioavailability of drugs. Cell membranes are becoming increasingly prevalent for assisting drug delivery because of the significant benefits of avoiding host cell barriers. Herein, B16F10 cell membranes (BFMs) were prepared in this study. BFMs could not only act as antigens but also serve as vesicles for vaccines. To trigger potent immunity, BFMs must be taken up by dendritic cells (DCs) and combined with adjuvants to make BFMs overcome the immune tolerance. To avoid circulating BFMs into tumors and quickly internalized by DCs after subcutaneously injection, the antigen-cell penetrating fusion peptide WT(YGRKKRRQRSRRYVDFFVWL) was used to modify BFMs. Additionally, a low dosage of paclitaxel (PTX) can activate DCs via toll-like receptor-4 (TLR-4). Therefore, we developed PTX-loaded micelles using Pluronic® F127. Then, WT-modified BFMs (WT-BFMs) were coated F127-PTX to yield WT-BFMs/ F127-PTX. Optimized WT-BFMs/F127-PTX promoted the cellular uptake and showed remarkable efficacy in eliciting robust antigen-specific cellular and humoral immune responses.
Collapse
|
17
|
Synthetic selenium nanoparticles as co-adjuvant improved immune responses against methicillin-resistant Staphylococcus aureus. World J Microbiol Biotechnol 2023; 39:16. [PMID: 36401129 PMCID: PMC9676803 DOI: 10.1007/s11274-022-03455-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the leading causes of hospital-acquired infections worldwide, which is resistant to many antibiotics, resulting in significant mortality in societies. Vaccination is a well-known approach to preventing disease. Autolysin, a surface-associated protein in S. aureus with multiple functions, is a suitable candidate for vaccine development. As a co-adjuvant, selenium nanoparticles (SeNPs) can increase the immune system, presumably resulting in increased vaccine efficacy. The present study evaluated the immunogenicity and defense of recombinant autolysin formulated in SeNPs and Alum adjuvants against MRSA. r-Autolysin was expressed and purified by the Ni-NTA affinity chromatography. SeNPs were synthetically obtained from sodium dioxide, followed by an assessment of shape and size using SEM and DLS. Balb/c mice were injected subcutaneously with 20 mg of r-autolysin formulated in Alum and SeNps adjuvants three times with the proper control group in 2 weeks intervals. Cytokine profile and isotyping ELISA were conducted to determine the type of induced immunity. Opsonophagocytosis tests assessed the functional activity of the vaccine, and the bacterial burden from the infected tissues was determined. Results showed that mice receiving SeNps and r-Autolysin had higher levels of total IgG and isotypes (IgG1 and IgG2a) and increased cytokine levels (IFN-γ, TNF-α, IL-12, and IL-4) as compared with those only receiving autolysin and PBS as a control. More importantly, mice immunized with SeNps and r-Autolysin exhibited a decrease in mortality and bacterial burden compared to the control group. We concluded that SeNps could stimulate immune responses and can be used as an adjuvant element in vaccine formulation.
Collapse
|
18
|
Zhang Z, Liu L, Ma C, Chen W. A Computational Model of Cytokine Release Syndrome during CAR T-cell Therapy. ADVANCED THERAPEUTICS 2022; 5:2200130. [PMID: 36590643 PMCID: PMC9797206 DOI: 10.1002/adtp.202200130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Indexed: 01/05/2023]
Abstract
Cytokine release syndrome (CRS) is a lethal adverse event in chimeric antigen receptor (CAR) T-cell therapy, hindering this promising therapy for cancers, such as B-cell acute lymphoblastic leukemia (B-ALL). Clinical management of CRS requires a better understanding of its underlying mechanisms. In this study, a computational model of CRS during CAR T-cell therapy is built to depict how the cellular interactions among CAR T-cells, B-ALL cells, and bystander monocytes, as well as the accompanying molecular interactions among various inflammatory cytokines, influence the severity of CRS. The model successfully defines the factors related to severe CRS and studied the effects of immunomodulatory therapy on CRS. The use of the model is also demonstrated as a precision medicine tool to optimize the treatment scheme, including personalized choice of CAR T-cell products and control of switchable CAR T-cell activity, for a more efficient and safer immunotherapy. This new computational oncology model can serve as a precision medicine tool to guide the clinical management of CRS during CAR T cell therapy.
Collapse
Affiliation(s)
- Zhuoyu Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
19
|
Saber S, Alomar SY, Yahya G. Blocking prostanoid receptors switches on multiple immune responses and cascades of inflammatory signaling against larval stages in snail fever. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43546-43555. [PMID: 35396684 PMCID: PMC9200668 DOI: 10.1007/s11356-022-20108-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 04/01/2022] [Indexed: 05/27/2023]
Abstract
Schistosomiasis, also known as snail fever or bilharziasis, is a worm infection caused by trematode called schistosomes that affects humans and animals worldwide. Schistosomiasis endemically exists in developing countries. Inflammatory responses elicited in the early phase of infection represent the rate limiting step for parasite migration and pathogenesis and could be a valuable target for therapeutic interventions. Prostaglandin E2 (PGE2) and interleukin (IL)-10 were found to be differentially affected in case of immune-modulation studies and cytokine analysis of hosts infected with either normal or radiation-attenuated parasite (RA) which switches off the development of an effective immune response against the migrating parasite in the early phase of schistosomiasis. Normal parasites induce predominantly a T helper 2 (Th2)-type cytokine response (IL-4 and IL-5) which is essential for parasite survival; here, we discuss in detail the downstream effects and cascades of inflammatory signaling of PGE2 and IL10 induced by normal parasites and the effect of blocking PGE2 receptors. We suggest that by selectively constraining the production of PGE2 during vaccination or therapy of susceptible persons or infected patients of schistosomiasis, this would boost IL-12 and reduce IL-10 production leading to a polarization toward the anti-worm Thl cytokine synthesis (IL-2 and Interferon (IFN)-γ).
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Suliman Y. Alomar
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharkia, 44519 Egypt
| |
Collapse
|
20
|
Dendritic Cells and Their Immunotherapeutic Potential for Treating Type 1 Diabetes. Int J Mol Sci 2022; 23:ijms23094885. [PMID: 35563276 PMCID: PMC9099521 DOI: 10.3390/ijms23094885] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic beta cells through a process that is primarily mediated by T cells. Emerging evidence suggests that dendritic cells (DCs) play a crucial role in initiating and developing this debilitating disease. DCs are professional antigen-presenting cells with the ability to integrate signals arising from tissue infection or injury that present processed antigens from these sites to naïve T cells in secondary lymphoid organs, thereby triggering naïve T cells to differentiate and modulate adaptive immune responses. Recent advancements in our knowledge of the various subsets of DCs and their cellular structures and methods of orchestration over time have resulted in a better understanding of how the T cell response is shaped. DCs employ various arsenal to maintain their tolerance, including the induction of effector T cell deletion or unresponsiveness and the generation and expansion of regulatory T cell populations. Therapies that suppress the immunogenic effects of dendritic cells by blocking T cell costimulatory pathways and proinflammatory cytokine production are currently being sought. Moreover, new strategies are being developed that can regulate DC differentiation and development and harness the tolerogenic capacity of these cells. Here, in this report, we focus on recent advances in the field of DC immunology and evaluate the prospects of DC-based therapeutic strategies to treat T1D.
Collapse
|
21
|
Chawda C, McMorrow R, Gaspar N, Zambito G, Mezzanotte L. Monitoring Immune Cell Function Through Optical Imaging: a Review Highlighting Transgenic Mouse Models. Mol Imaging Biol 2022; 24:250-263. [PMID: 34735680 PMCID: PMC8983637 DOI: 10.1007/s11307-021-01662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022]
Abstract
Transgenic mouse models have facilitated research of human diseases and validation of therapeutic approaches. Inclusion of optical reporter genes (fluorescent or bioluminescent genes) in the targeting vectors used to develop such models makes in vivo imaging of cellular and molecular events possible, from the microscale to the macroscale. In particular, transgenic mouse models expressing optical reporter genes allowed accurately distinguishing immune cell types from trafficking in vivo using intravital microscopy or whole-body optical imaging. Besides lineage tracing and trafficking of different subsets of immune cells, the ability to monitor the function of immune cells is of pivotal importance for investigating the effects of immunotherapies against cancer. Here, we introduce the reader to state-of-the-art approaches to develop transgenics, optical imaging techniques, and several notable examples of transgenic mouse models developed for immunology research by critically highlighting the models that allow the following of immune cell function.
Collapse
Affiliation(s)
- Chintan Chawda
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Natasa Gaspar
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Leleux JA, Albershardt TC, Reeves R, James R, Krull J, Parsons AJ, ter Meulen J, Berglund P. Intratumoral expression of IL-12 from lentiviral or RNA vectors acts synergistically with TLR4 agonist (GLA) to generate anti-tumor immunological memory. PLoS One 2021; 16:e0259301. [PMID: 34855754 PMCID: PMC8638928 DOI: 10.1371/journal.pone.0259301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/15/2021] [Indexed: 11/20/2022] Open
Abstract
Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Expression Regulation
- Genetic Vectors/administration & dosage
- Genetic Vectors/pharmacology
- Glucosides/pharmacology
- Immunity, Innate/drug effects
- Immunity, Innate/genetics
- Immunologic Memory/drug effects
- Immunologic Memory/genetics
- Immunotherapy/methods
- Interferon-gamma/blood
- Interleukin-12/blood
- Interleukin-12/genetics
- Interleukin-12/immunology
- Lentivirus/genetics
- Lipid A/pharmacology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Toll-Like Receptor 4/agonists
- Mice
Collapse
Affiliation(s)
- Jardin A. Leleux
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Tina C. Albershardt
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Rebecca Reeves
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Reice James
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Jordan Krull
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Andrea J. Parsons
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Jan ter Meulen
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Peter Berglund
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| |
Collapse
|
23
|
Gomez-Lopez N, Garcia-Flores V, Chin PY, Groome HM, Bijland MT, Diener KR, Romero R, Robertson SA. Macrophages exert homeostatic actions in pregnancy to protect against preterm birth and fetal inflammatory injury. JCI Insight 2021; 6:146089. [PMID: 34622802 PMCID: PMC8525593 DOI: 10.1172/jci.insight.146089] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
Macrophages are commonly thought to contribute to the pathophysiology of preterm labor by amplifying inflammation — but a protective role has not previously been considered to our knowledge. We hypothesized that given their antiinflammatory capability in early pregnancy, macrophages exert essential roles in maintenance of late gestation and that insufficient macrophages may predispose individuals to spontaneous preterm labor and adverse neonatal outcomes. Here, we showed that women with spontaneous preterm birth had reduced CD209+CD206+ expression in alternatively activated CD45+CD14+ICAM3– macrophages and increased TNF expression in proinflammatory CD45+CD14+CD80+HLA-DR+ macrophages in the uterine decidua at the materno-fetal interface. In Cd11bDTR/DTR mice, depletion of maternal CD11b+ myeloid cells caused preterm birth, neonatal death, and postnatal growth impairment, accompanied by uterine cytokine and leukocyte changes indicative of a proinflammatory response, while adoptive transfer of WT macrophages prevented preterm birth and partially rescued neonatal loss. In a model of intra-amniotic inflammation–induced preterm birth, macrophages polarized in vitro to an M2 phenotype showed superior capacity over nonpolarized macrophages to reduce uterine and fetal inflammation, prevent preterm birth, and improve neonatal survival. We conclude that macrophages exert a critical homeostatic regulatory role in late gestation and are implicated as a determinant of susceptibility to spontaneous preterm birth and fetal inflammatory injury.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Holly M Groome
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Melanie T Bijland
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kerrilyn R Diener
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, US Department of Health and Human Services; Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Krieger SS, Zwart SR, Mehta S, Wu H, Simpson RJ, Smith SM, Crucian B. Alterations in Saliva and Plasma Cytokine Concentrations During Long-Duration Spaceflight. Front Immunol 2021; 12:725748. [PMID: 34504500 PMCID: PMC8422944 DOI: 10.3389/fimmu.2021.725748] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/05/2021] [Indexed: 01/01/2023] Open
Abstract
Long-duration spaceflight is known to cause immune dysregulation in astronauts. Biomarkers of immune system function are needed to determine both the need for and effectiveness of potential immune countermeasures for astronauts. Whereas plasma cytokine concentrations are a well-established biomarker of immune status, salivary cytokine concentrations are emerging as a sensitive indicator of stress and inflammation. For this study, to aid in characterizing immune dysregulation during spaceflight, plasma and saliva cytokines were monitored in astronauts before, during and after long-duration spaceflight onboard the International Space Station. Blood was collected from 13 astronauts at 3 timepoints before, 5 timepoints during and 3 timepoints after spaceflight. Saliva was collected from 6 astronauts at 2 timepoints before spaceflight, 2 timepoints during and 3 timepoints following spaceflight. Samples were analyzed using multiplex array technology. Significant increases in the plasma concentration of IL-3, IL-15, IL-12p40, IFN-α2, and IL-7 were observed during spaceflight compared to before flight baseline. Significant decreases in saliva GM-CSF, IL-12p70, IL-10 and IL-13 were also observed during spaceflight as compared to compared to before flight baseline concentrations. Additionally, plasma TGFβ1 and TGFβ2 concentrations tended to be consistently higher during spaceflight, although these did not reach statistical significance. Overall, the findings confirm an in-vivo hormonal dysregulation of immunity, appearing pro-inflammatory and Th1 in nature, persists during long-duration orbital spaceflight. These biomarkers may therefore have utility for monitoring the effectiveness of biomedical countermeasures for astronauts, with potential application in terrestrial research and medicine.
Collapse
Affiliation(s)
| | - Sara R Zwart
- University of Texas Medical Branch (UTMB), Preventive Medicine and Population Health, Galveston, TX, United States
| | - Satish Mehta
- JES Tech, Human Health and Performance Directorate, Houston, TX, United States
| | - Honglu Wu
- National Aeronautics and Space Administration (NASA) Johnson Space Center, Human Health and Performance Directorate, Houston, TX, United States
| | - Richard J Simpson
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,The University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States
| | - Scott M Smith
- National Aeronautics and Space Administration (NASA) Johnson Space Center, Human Health and Performance Directorate, Houston, TX, United States
| | - Brian Crucian
- National Aeronautics and Space Administration (NASA) Johnson Space Center, Human Health and Performance Directorate, Houston, TX, United States
| |
Collapse
|
25
|
Han SH, Ahn Y, Lee HJ, Suh HJ, Jo K. Antioxidant and Immunostimulatory Activities of a Submerged Culture of Cordyceps sinensis Using Spent Coffee. Foods 2021; 10:foods10081697. [PMID: 34441476 PMCID: PMC8394004 DOI: 10.3390/foods10081697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Spent coffee grounds (SCG) are inexpensive materials that have been used as a source of antioxidants and polysaccharides with immunostimulatory activity. In this study, we performed a microbial fermentation of SCG using Cordyceps sinensis and investigated the radical scavenging and immunostimulatory activity of fermented SCG. SCG fermentation using C. sinensis was performed at 25 °C for 8 d. The polyphenol content of the fermented SCG increased from 1022.4 to 1562.0 μg/mL. The glucosamine content of the mycelia also continuously increased during fermentation. The main polyphenol compounds of fermented SCG were chlorogenic acid and p-coumaric acid, which were increased by fermentation. Fermented SCG also showed significantly higher content of chlorogenic acid isomers than unfermented SCG. The fermented SCG exhibited significantly higher 2,2-diphenyl-2-picrylhydrazyl hydrate (half maximal inhibitory concentration: IC50, 0.37 mg/mL) and 2,2-azinobis (3-ethylbenzothiazoline-6-sulfonic acid) (IC50, 0.93 mg/mL) radical scavenging activities than those of the control (0.54 mg/mL and 1.20 mg/mL, respectively; p < 0.05). The fermented SCG stimulated macrophages and promoted the production of various immunostimulatory cytokines (IL-12, IL-6, and TNF-α) compared to control; therefore, microbial fermentation of SCG using C. sinensis is an effective means of generating antioxidant and immunostimulatory materials.
Collapse
Affiliation(s)
- Sung Hee Han
- Institute of Human Behavior & Genetic, College of Medicine, Korea University, Seoul 02841, Korea;
| | - Yejin Ahn
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Korea; (Y.A.); (H.J.S.)
| | - Hyun Jung Lee
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID 83844, USA;
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Korea; (Y.A.); (H.J.S.)
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Korea
| | - Kyungae Jo
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Korea; (Y.A.); (H.J.S.)
- Correspondence: ; Tel.: +82-2-940-2764; Fax: +82-2-921-7207
| |
Collapse
|
26
|
Qaseem MF, Shaheen H, Wu AM. Cell wall hemicellulose for sustainable industrial utilization. RENEWABLE AND SUSTAINABLE ENERGY REVIEWS 2021; 144:110996. [DOI: 10.1016/j.rser.2021.110996] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
27
|
Tsai CK, Cheng HH, Hsu TY, Wang JY, Hung CH, Tsai CC, Lai YJ, Lin YJ, Huang HC, Chan JYH, Tain YL, Chen CC, Tsai TA, Yu HR. Prenatal Exposure to Di-Ethyl Phthalate (DEP) Is Related to Increasing Neonatal IgE Levels and the Altering of the Immune Polarization of Helper-T Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18126364. [PMID: 34208324 PMCID: PMC8296186 DOI: 10.3390/ijerph18126364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022]
Abstract
Introduction: Phthalates are substances that are added to plastic products to increase their plasticity. These substances are released easily into the environment and can act as endocrine disruptors. Epidemiological studies in children have showed inconsistent findings regarding the relationship between prenatal or postnatal exposure to phthalates and the risk of allergic disease. Our hypothesis is that prenatal exposure to phthalates may contribute to the development of allergies in children. Material and methods: The objective of this study was to determine the associations between urinary phthalate metabolite concentrations in pregnant women, maternal atopic diathesis, maternal lifestyle, and cord blood IgE. Pregnant mothers and paired newborns (n = 101) were enrolled from an antenatal clinic. The epidemiologic data and the clinical information were collected using standard questionnaires and medical records. The maternal blood and urine samples were collected at 24–28 weeks gestation, and cord blood IgE, IL-12p70, IL-4, and IL-10 levels were determined from the newborns at birth. The link between phthalates and maternal IgE was also assessed. To investigate the effects of phthalates on neonatal immunity, cord blood mononuclear cells (MNCs) were used for cytokine induction in another in vitro experiment. Results: We found that maternal urine monoethyl phthalate (MEP) (a metabolite of di-ethyl phthalate (DEP)) concentrations are positively correlated with the cord blood IgE of the corresponding newborns. The cord blood IL-12p70 levels of mothers with higher maternal urine MEP groups (high DEP exposure) were lower than mothers with low DEP exposure. In vitro experiments demonstrated that DEP could enhance IL-4 production of cord blood MNCs rather than adult MNCs. Conclusion: Prenatal DEP exposure is related to neonatal IgE level and alternation of cytokines relevant to Th1/Th2 polarization. This suggests the existence of a link between prenatal exposure to specific plasticizers and the future development of allergies.
Collapse
Affiliation(s)
- Chang-Ku Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Hsin-Hsin Cheng
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Kaohsiung 83301, Taiwan; (H.-H.C.); (T.-Y.H.); (C.-C.T.); (Y.-J.L.); (Y.-J.L.)
| | - Te-Yao Hsu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Kaohsiung 83301, Taiwan; (H.-H.C.); (T.-Y.H.); (C.-C.T.); (Y.-J.L.); (Y.-J.L.)
| | - Jiu-Yao Wang
- Department of Pediatrics, College of Medicine, National Cheng Kung University, Tainan City 704302, Taiwan;
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Ching-Chang Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Kaohsiung 83301, Taiwan; (H.-H.C.); (T.-Y.H.); (C.-C.T.); (Y.-J.L.); (Y.-J.L.)
| | - Yun-Ju Lai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Kaohsiung 83301, Taiwan; (H.-H.C.); (T.-Y.H.); (C.-C.T.); (Y.-J.L.); (Y.-J.L.)
| | - Yu-Ju Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Centre, Kaohsiung 83301, Taiwan; (H.-H.C.); (T.-Y.H.); (C.-C.T.); (Y.-J.L.); (Y.-J.L.)
| | - Hsin-Chun Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Julie Y. H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Chih-Cheng Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Ti-An Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (C.-K.T.); (H.-C.H.); (Y.-L.T.); (C.-C.C.); (T.-A.T.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8713); Fax: +886-7-733-8009
| |
Collapse
|
28
|
Lin M, Huang W, Kabbani N, Theiss MM, Hamilton JF, Ecklund JM, Conley YP, Vodovotz Y, Brienza D, Wagner AK, Robbins E, Sowa GA, Lipsky RH. Effect of CHRFAM7A Δ2bp gene variant on secondary inflammation after spinal cord injury. PLoS One 2021; 16:e0251110. [PMID: 33956875 PMCID: PMC8101719 DOI: 10.1371/journal.pone.0251110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/20/2021] [Indexed: 11/18/2022] Open
Abstract
The α7 neuronal nicotinic acetylcholine receptors (α7nAChRs) are essential for anti-inflammatory responses. The human-specific CHRFAM7A gene and its 2bp deletion polymorphism (Δ2bp variant) encodes a structurally-deficient α7nAChRs that may impact the anti-inflammatory function. We studied 45 spinal cord injury (SCI) patients for up to six weeks post SCI to investigate the role of the Δ2bp variant on multiple circulating inflammatory mediators and two outcome measures (neuropathic pain and risk of pressure ulcers). The patient's SCI were classified as either severe or mild. Missing values were imputed. Overall genetic effect was conducted with independent sample t-test and corrected with false discovery rate (FDR). Univariate analysis and regression analysis were applied to evaluate the Δ2bp effects on temporal variation of inflammatory mediators post SCI and their interaction with outcome measures. In severe SCI, the Δ2bp carriers showed higher levels of circulating inflammatory mediators than the Δ2bp non-carriers in TNF-α (FDR = 9.6x10-4), IFN-γ (FDR = 1.3x10-3), IL-13 (FDR = 1.6x10-3), CCL11 (FDR = 2.1x10-3), IL-12p70 (FDR = 2.2x10-3), IL-8 (FDR = 2.2x10-3), CXCL10 (FDR = 3.1x10-3), CCL4 (FDR = 5.7x10-3), IL-12p40 (FDR = 7.1x10-3), IL-1b (FDR = 0.014), IL-15 (FDR = 0.024), and IL-2 (FDR = 0.037). IL-8 and CCL2 were negatively associated with days post injury (DPI) for the Δ2bp carriers (P = 2x10-7 and P = 2x10-8, respectively) and IL-5 was positively associated with DPI for the Δ2bp non-carriers (P = 0.015). Neuropathic pain was marginally positively associated with IL-13 for the Δ2bp carriers (P = 0.056). In mild SCI, the Δ2bp carriers had lower circulating levels of IL-15 (FDR = 0.04) than the Δ2bp non-carriers. Temporal variation of inflammatory mediators post SCI was not associated with the Δ2bp variant. For the mild SCI Δ2bp carriers, risk of pressure ulcers was positively associated with circulating levels of IFN-γ, CXCL10, and CCL4 and negatively associated with circulating levels of IL-12p70. These findings support an important role for the human-specific CHRFAM7A Δ2bp gene variant in modifying anti-inflammatory function of α7nAChRs following SCI.
Collapse
Affiliation(s)
- Mingkuan Lin
- School of Systems Biology, George Mason University, Fairfax, Virginia, United States of America
- Inova Neuroscience and Spine Institute, Inova Health System, Falls Church, Virginia, United States of America
- * E-mail:
| | - Wan Huang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Nadine Kabbani
- School of Systems Biology, George Mason University, Fairfax, Virginia, United States of America
| | - Mark M. Theiss
- Department of Orthopedic Services, Inova Health System, Falls Church, Virginia, United States of America
| | - John F. Hamilton
- Inova Neuroscience and Spine Institute, Inova Health System, Falls Church, Virginia, United States of America
| | - James M. Ecklund
- Inova Neuroscience and Spine Institute, Inova Health System, Falls Church, Virginia, United States of America
| | - Yvette P. Conley
- School of Nursing and Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yoram Vodovotz
- Department of Surgery, Center for Inflammation & Regenerative Modeling in McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - David Brienza
- Rehabilitation Science &Technology, Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amy K. Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Emily Robbins
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Gwendolyn A. Sowa
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robert H. Lipsky
- School of Systems Biology, George Mason University, Fairfax, Virginia, United States of America
- Inova Neuroscience and Spine Institute, Inova Health System, Falls Church, Virginia, United States of America
| |
Collapse
|
29
|
Molagoda IMN, Jayasingha JACC, Choi YH, Jayasooriya RGPT, Kang CH, Kim GY. Fisetin inhibits lipopolysaccharide-induced inflammatory response by activating β-catenin, leading to a decrease in endotoxic shock. Sci Rep 2021; 11:8377. [PMID: 33863923 PMCID: PMC8052411 DOI: 10.1038/s41598-021-87257-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Fisetin is a naturally occurring flavonoid that possesses several pharmacological benefits including anti-inflammatory activity. However, its precise anti-inflammatory mechanism is not clear. In the present study, we found that fisetin significantly inhibited the expression of proinflammatory mediators, such as nitric oxide (NO) and prostaglandin E2 (PGE2), and cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Additionally, fisetin attenuated LPS-induced mortality and abnormalities in zebrafish larvae and normalized the heart rate. Fisetin decreased the recruitment of macrophages and neutrophils to the LPS-microinjected inflammatory site in zebrafish larvae, concomitant with a significant downregulation of proinflammatory genes, such as inducible NO synthase (iNOS), cyclooxygenase-2a (COX-2a), IL-6, and TNF-α. Fisetin inhibited the nuclear localization of nuclear factor-kappa B (NF-κB), which reduced the expression of pro-inflammatory genes. Further, fisetin inactivated glycogen synthase kinase 3β (GSK-3β) via phosphorylation at Ser9, and inhibited the degradation of β-catenin, which consequently promoted the localization of β-catenin into the nucleus. The pharmacological inhibition of β-catenin with FH535 reversed the fisetin-induced anti-inflammatory activity and restored NF-κB activity, which indicated that fisetin-mediated activation of β-catenin results in the inhibition of LPS-induced NF-κB activity. In LPS-microinjected zebrafish larvae, FH535 promoted the migration of macrophages to the yolk sac and decreased resident neutrophil counts in the posterior blood island and induced high expression of iNOS and COX-2a, which was accompanied by the inhibition of fisetin-induced anti-inflammatory activity. Altogether, the current study confirmed that the dietary flavonoid, fisetin, inhibited LPS-induced inflammation and endotoxic shock through crosstalk between GSK-3β/β-catenin and the NF-κB signaling pathways.
Collapse
Affiliation(s)
| | | | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan, 47227, Republic of Korea
| | | | - Chang-Hee Kang
- Bioresources Industrialization Support Department, Nakdonggang National Institute of Biological Resources, Sangju, 37242, Republic of Korea.
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
30
|
Chin WY, He CY, Chow TW, Yu QY, Lai LC, Miaw SC. Adenylate Kinase 4 Promotes Inflammatory Gene Expression via Hif1α and AMPK in Macrophages. Front Immunol 2021; 12:630318. [PMID: 33790902 PMCID: PMC8005550 DOI: 10.3389/fimmu.2021.630318] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages comprise the front line of defense against various pathogens. Classically activated macrophages (M1), induced by IFN-γ and LPS, highly express inflammatory cytokines and contribute to inflammatory processes. By contrast, alternatively activated macrophages (M2) are induced by IL-4 and IL-13, produce IL-10, and display anti-inflammatory activity. Adenylate kinase 4 (Ak4), an enzyme that transfers phosphate group among ATP/GTP, AMP, and ADP, is a key modulator of ATP and maintains the homeostasis of cellular nucleotides which is essential for cell functions. However, its role in regulating the function of macrophages is not fully understood. Here we report that Ak4 expression is induced in M1 but not M2 macrophages. Suppressing the expression of Ak4 in M1 macrophages with shRNA or siRNA enhances ATP production and decreases ROS production, bactericidal ability and glycolysis in M1 cells. Moreover, Ak4 regulates the expression of inflammation genes, including Il1b, Il6, Tnfa, Nos2, Nox2, and Hif1a, in M1 macrophages. We further demonstrate that Ak4 inhibits the activation of AMPK and forms a positive feedback loop with Hif1α to promote the expression of inflammation-related genes in M1 cells. Furthermore, RNA-seq analysis demonstrates that Ak4 also regulates other biological processes in addition to the expression of inflammation-related genes in M1 cells. Interestingly, Ak4 does not regulate M1/M2 polarization. Taken together, our study uncovers a potential mechanism linking energy consumption and inflammation in macrophages.
Collapse
Affiliation(s)
- Wei-Yao Chin
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Ying He
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tsun Wai Chow
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Qi-You Yu
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Liang-Chuan Lai
- Bioinformatics and Biostatistics Core, Center of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.,College of Medicine, Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Shi-Chuen Miaw
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
31
|
Respiratory and Systemic Toxicity of Inhaled Artificial Asian Sand Dust in Pigs. Life (Basel) 2021; 11:life11010025. [PMID: 33406620 PMCID: PMC7823853 DOI: 10.3390/life11010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 11/26/2022] Open
Abstract
Air pollution, particularly caused by Asian sand dust (ASD) and particulate matter (PM), has become one of the leading threats to public health. However, the majority of studies have primarily focused on epidemiological assessment, and in vivo toxicities of certain air pollutants have been poorly elucidated in medium/large-size laboratory animals. To investigate the impact of ASD in domestic animals, 16 Landrace pigs were exposed to an artificial ASD sandstorm for 6 h. All animals were divided in four cages, and a commercial yellow soil was used for generating artificial mineralogical particles. Blood samples were collected, and necropsies were performed before exposure and 6, 12, 24, and 72 h after exposure. Complete blood cell count and the levels of serum biochemical enzymes, blood gas, electrolytes, and a variety of inflammatory cytokines were evaluated. In addition, histopathological examination was conducted. Various test results proved acute lower airway disorders with systemic inflammation in pigs. To our knowledge, this study is the first to describe experimental research in domestic animals concerning the damage caused by artificial ASD exposure. The results of this study suggest that ASD has importance in terms of not only public health but also of ultimate economic losses in the pork industry.
Collapse
|
32
|
Dong Z, Liu Z, Dai H, Liu W, Feng Z, Zhao Q, Gao Y, Liu F, Zhang N, Dong X, Zhou X, Du J, Huang G, Tian X, Liu B. The Potential Role of Regulatory B Cells in Idiopathic Membranous Nephropathy. J Immunol Res 2020; 2020:7638365. [PMID: 33426094 PMCID: PMC7772048 DOI: 10.1155/2020/7638365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/22/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Regulatory B cells (Breg) are widely regarded as immunomodulatory cells which play an immunosuppressive role. Breg inhibits pathological autoimmune response by secreting interleukin-10 (IL-10), transforming growth factor-β (TGF-β), and adenosine and through other ways to prevent T cells and other immune cells from expanding. Recent studies have shown that different inflammatory environments induce different types of Breg cells, and these different Breg cells have different functions. For example, Br1 cells can secrete IgG4 to block autoantigens. Idiopathic membranous nephropathy (IMN) is an autoimmune disease in which the humoral immune response is dominant and the cellular immune response is impaired. However, only a handful of studies have been done on the role of Bregs in this regard. In this review, we provide a brief overview of the types and functions of Breg found in human body, as well as the abnormal pathological and immunological phenomena in IMN, and propose the hypothesis that Breg is activated in IMN patients and the proportion of Br1 can be increased. Our review aims at highlighting the correlation between Breg and IMN and proposes potential mechanisms, which can provide a new direction for the discovery of the pathogenesis of IMN, thus providing a new strategy for the prevention and early treatment of IMN.
Collapse
Affiliation(s)
- Zhaocheng Dong
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Zhiyuan Liu
- Shandong First Medical University, No. 619 Changcheng Road, Tai'an City, Shandong 271016, China
| | - Haoran Dai
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Station East 5, Shunyi District, Beijing 101300, China
| | - Wenbin Liu
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
| | - Zhendong Feng
- Beijing Chinese Medicine Hospital Pinggu Hospital, No. 6, Pingxiang Road, Pinggu District, Beijing 101200, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Yu Gao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Fei Liu
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Na Zhang
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Xuan Dong
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Xiaoshan Zhou
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Jieli Du
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Guangrui Huang
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
| | - Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| |
Collapse
|
33
|
IL-12 and IL-23-Close Relatives with Structural Homologies but Distinct Immunological Functions. Cells 2020; 9:cells9102184. [PMID: 32998371 PMCID: PMC7600943 DOI: 10.3390/cells9102184] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022] Open
Abstract
Cytokines of the IL-12 family show structural similarities but have distinct functions in the immune system. Prominent members of this cytokine family are the pro-inflammatory cytokines IL-12 and IL-23. These two cytokines share cytokine subunits and receptor chains but have different functions in autoimmune diseases, cancer and infections. Accordingly, structural knowledge about receptor complex formation is essential for the development of new therapeutic strategies preventing and/or inhibiting cytokine:receptor interaction. In addition, intracellular signaling cascades can be targeted to inhibit cytokine-mediated effects. Single nucleotide polymorphisms can lead to alteration in the amino acid sequence and thereby influencing protein functions or protein–protein interactions. To understand the biology of IL-12 and IL-23 and to establish efficient targeting strategies structural knowledge about cytokines and respective receptors is crucial. A highly efficient therapy might be a combination of different drugs targeting extracellular cytokine:receptor assembly and intracellular signaling pathways.
Collapse
|
34
|
Schulz-Kuhnt A, Wirtz S, Neurath MF, Atreya I. Regulation of Human Innate Lymphoid Cells in the Context of Mucosal Inflammation. Front Immunol 2020; 11:1062. [PMID: 32655549 PMCID: PMC7324478 DOI: 10.3389/fimmu.2020.01062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since their identification as a unique cell population, innate lymphoid cells (ILCs) have revolutionized our understanding of immune responses, leaving their impact on multiple inflammatory and fibrotic pathologies without doubt. Thus, a tightly controlled regulation of local ILC numbers and their activity is of crucial importance. Even though this has been extensively studied in murine ILCs in the last few years, our knowledge of human ILCs is still lagging behind. Our review article will therefore summarize recent insights into the function of human ILCs and will particularly focus on their regulation under inflammatory conditions. The quality and intensity of ILC involvement into local immune responses at mucosal sites of the human body can potentially be modulated via three different axes: (1) activation of tissue-resident mature ILCs, (2) plasticity and local transdifferentiation of specific ILC subsets, and (3) tissue migration and accumulation of peripheral ILCs. Despite a still ongoing scientific effort in this field, already existing data on the fate of human ILCs under different pathologic conditions clearly indicate that all three of these mechanisms are of relevance for the clinical course of chronic inflammatory and autoimmune diseases and might likewise provide new target structures for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
35
|
Nistor GI, Dillman RO. Cytokine network analysis of immune responses before and after autologous dendritic cell and tumor cell vaccine immunotherapies in a randomized trial. J Transl Med 2020; 18:176. [PMID: 32316978 PMCID: PMC7171762 DOI: 10.1186/s12967-020-02328-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background In a randomized phase II trial conducted in patients with metastatic melanoma, patient-specific autologous dendritic cell vaccines (DCV) were associated with longer survival than autologous tumor cell vaccines (TCV). Both vaccines presented antigens from cell-renewing autologous tumor cells. The current analysis was performed to better understand the immune responses induced by these vaccines, and their association with survival. Methods 110 proteomic markers were measured at a week-0 baseline, 1 week before the first of 3 weekly vaccine injections, and at week-4, 1 week after the third injection. Data was presented as a deviation from normal controls. A two-component principal component (PC) statistical analysis and discriminant analysis were performed on this data set for all patients and for each treatment cohort. Results At baseline PC-1 contained 64.4% of the variance and included the majority of cytokines associated with Th1 and Th2 responses, which positively correlated with beta-2-microglobulin (B2M), programmed death protein-1 (PD-1) and transforming growth factor beta (TGFβ1). Results were similar at baseline for both treatment cohorts. After three injections, DCV-treated patients showed correlative grouping among Th1/Th17 cytokines on PC-1, with an inverse correlation with B2M, FAS, and IL-18, and correlations among immunoglobulins in PC-2. TCV-treated patients showed a positive correlation on PC-1 among most of the cytokines and tumor markers B2M and FAS receptor. There were also correlative changes of IL12p40 with both Th1 and Th2 cytokines and TGFβ1. Discriminant analysis provided additional evidence that DCV was associated with innate, Th1/Th17, and Th2 responses while TCV was only associated with innate and Th2 responses. Conclusions These analyses confirm that DCV induced a different immune response than that induced by TCV, and these immune responses were associated with improved survival. Trial registration Clinical trials.gov NCT004936930 retrospectively registered 28 July 2009
Collapse
Affiliation(s)
- Gabriel I Nistor
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA
| | - Robert O Dillman
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA.
| |
Collapse
|
36
|
Azeem W, Bakke RM, Appel S, Øyan AM, Kalland KH. Dual Pro- and Anti-Inflammatory Features of Monocyte-Derived Dendritic Cells. Front Immunol 2020; 11:438. [PMID: 32292402 PMCID: PMC7120039 DOI: 10.3389/fimmu.2020.00438] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/25/2020] [Indexed: 01/01/2023] Open
Abstract
The transcription factor β-catenin is able to induce tolerogenic/anti-inflammatory features in different types of dendritic cells (DCs). Monocyte-derived dendritic cells (moDCs) have been widely used in dendritic cell-based cancer therapy, but so far with limited clinical efficacy. We wanted to investigate the hypothesis that aberrant differentiation or induction of dual pro- and anti-inflammatory features may be β-catenin dependent in moDCs. β-catenin was detectable in both immature and lipopolysaccharide (LPS)-stimulated DCs. The β-catenin inhibitor ICG-001 dose-dependently increased the pro-inflammatory signature cytokine IL-12p70 and decreased the anti-inflammatory signature molecule IL-10. The β-catenin activator 6-bromoindirubin-3′-oxime (6-BIO) dose-dependently increased total and nuclear β-catenin, and this was associated with decreased IL-12p70, increased IL-10, and reduced surface expression of activation markers, such as CD80 and CD86, and increased expression of inhibitory markers, such as PD-L1. 6-BIO and ICG-001 competed dose-dependently regarding these features. Genome-wide mRNA expression analyses further underscored the dual development of pro- and anti-inflammatory features of LPS-matured moDCs and suggest a role for β-catenin inhibition in production of more potent therapeutic moDCs.
Collapse
Affiliation(s)
- Waqas Azeem
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ragnhild Maukon Bakke
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| | - Silke Appel
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
| | - Anne Margrete Øyan
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| | - Karl-Henning Kalland
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Norway Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| |
Collapse
|
37
|
Li Y, Du Y, Zhang A, Jiang R, Nie X, Xiong X. Role of CCR7 on dendritic cell‑mediated immune tolerance in the airways of allergy‑induced asthmatic rats. Mol Med Rep 2019; 20:4425-4432. [PMID: 31545493 PMCID: PMC6797982 DOI: 10.3892/mmr.2019.10694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/26/2019] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) have an important role in initiating and maintaining the immune inflammatory response in allergic asthma, and CC chemokine receptor 7 (CCR7) is directly involved in the pathogenesis of DC- and T cell-mediated allergic asthma. The present study aimed to investigate the effects of CCR7 on DC-mediated immune tolerance in allergic asthma. In the present study, bone marrow-derived DCs were transfected with an adenovirus encoding the rat CCR7 gene or a short hairpin RNA targeting CCR7 (sh-CCR7). Rats injected with DCs overexpressing CCR7 or presenting CCR7 knockdown were examined. After the rats were injected with DCs via the tail vein, bronchoalveolar lavage fluid was collected to assess its cellular composition. The protein expression levels of CCR7 in DCs were determined using immunohistochemistry and western blot analysis. The protein expression levels of interferon-γ (IFN-γ), interleukin-4 (IL-4), IL-10, IL-12, transforming growth factor-β (TGF-β) and immunoglobulin E (IgE) were determined by ELISA. Compared with the control group, the protein expression level of CCR7 was significantly higher in the CCR7 overexpression group and significantly lower in sh-CCR7 group. Similarly, the number of DCs was higher in the CCR7 overexpression group and lower in the sh-CCR7 group. The protein expression levels of IL-10 and TGF-β were significantly lower in the CCR7 overexpression group and higher in the sh-CCR7 group. In addition, the expression levels of IL-4, IL-12, IFN-γ and IgE were higher in the CCR7 overexpression group and lower in the sh-CCR7 group. The present results suggested that the role of cytokines and IgE in immune inflammation and immune tolerance in allergic asthma may be associated with the expression level of CCR7 in DCs, suggesting that CCR7 may serve a role in DC-mediated immune tolerance in allergic asthma.
Collapse
Affiliation(s)
- Yi Li
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| | - Yongcheng Du
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| | - Aizhen Zhang
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| | - Rui Jiang
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| | - Xin Nie
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| | - Xue Xiong
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
38
|
Nahas MR, Rosenblatt J, Lazarus HM, Avigan D. Anti-cancer vaccine therapy for hematologic malignancies: An evolving era. Blood Rev 2018; 32:312-325. [PMID: 29475779 DOI: 10.1016/j.blre.2018.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/19/2022]
Abstract
The potential promise of therapeutic vaccination as effective therapy for hematologic malignancies is supported by the observation that allogeneic hematopoietic cell transplantation is curative for a subset of patients due to the graft-versus-tumor effect mediated by alloreactive lymphocytes. Tumor vaccines are being explored as a therapeutic strategy to re-educate host immunity to recognize and target malignant cells through the activation and expansion of effector cell populations. Via several mechanisms, tumor cells induce T cell dysfunction and senescence, amplifying and maintaining tumor cell immunosuppressive effects, resulting in failure of clinical trials of tumor vaccines and adoptive T cell therapies. The fundamental premise of successful vaccine design involves the introduction of tumor-associated antigens in the context of effective antigen presentation so that tolerance can be reversed and a productive response can be generated. With the increasing understanding of the role of both the tumor and tumor microenvironment in fostering immune tolerance, vaccine therapy is being explored in the context of immunomodulatory therapies. The most effective strategy may be to use combination therapies such as anti-cancer vaccines with checkpoint blockade to target critical aspects of this environment in an effort to prevent the re-establishment of tumor tolerance while limiting toxicity associated with autoimmunity.
Collapse
Affiliation(s)
- Myrna R Nahas
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jacalyn Rosenblatt
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David Avigan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
39
|
Magalhães TNC, Weiler M, Teixeira CVL, Hayata T, Moraes AS, Boldrini VO, dos Santos LM, de Campos BM, de Rezende TJR, Joaquim HPG, Talib LL, Forlenza OV, Cendes F, Balthazar MLF. Systemic Inflammation and Multimodal Biomarkers in Amnestic Mild Cognitive Impairment and Alzheimer’s Disease. Mol Neurobiol 2017; 55:5689-5697. [DOI: 10.1007/s12035-017-0795-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/26/2017] [Indexed: 12/01/2022]
|
40
|
Urzua U, Chacon C, Lizama L, Sarmiento S, Villalobos P, Kroxato B, Marcelain K, Gonzalez MJ. Parity History Determines a Systemic Inflammatory Response to Spread of Ovarian Cancer in Naturally Aged Mice. Aging Dis 2017; 8:546-557. [PMID: 28966800 PMCID: PMC5614320 DOI: 10.14336/ad.2017.0110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/10/2017] [Indexed: 01/09/2023] Open
Abstract
Aging intersects with reproductive senescence in women by promoting a systemic low-grade chronic inflammation that predisposes women to several diseases including ovarian cancer (OC). OC risk at menopause is significantly modified by parity records during prior fertile life. To date, the combined effects of age and parity on the systemic inflammation markers that are particularly relevant to OC initiation and progression at menopause remain largely unknown. Herein, we profiled a panel of circulating cytokines in multiparous versus virgin C57BL/6 female mice at peri-estropausal age and investigated how cytokine levels were modulated by intraperitoneal tumor induction in a syngeneic immunocompetent OC mouse model. Serum FSH, LH and TSH levels increased with age in both groups while prolactin (PRL) was lower in multiparous respect to virgin mice, a finding previously observed in parous women. Serum CCL2, IL-10, IL-5, IL-4, TNF-α, IL1-β and IL-12p70 levels increased with age irrespective of parity status, but were specifically reduced following OC tumor induction only in multiparous mice. Animals developed hemorrhagic ascites and tumor implants in the omental fat band and other intraperitoneal organs by 12 weeks after induction, with multiparous mice showing a significantly extended survival. We conclude that previous parity history counteracts aging-associated systemic inflammation possibly by reducing the immunosuppression that typically allows tumor spread. Results suggest a partial impairment of the M2 shift in tumor-associated macrophages as well as decreased stimulation of regulatory B-cells in aged mice. This long term, tumor-concurrent effect of parity on inflammation markers at menopause would be a contributing factor leading to decreased OC risk.
Collapse
Affiliation(s)
- Ulises Urzua
- 1Laboratorio de Genómica Aplicada, Facultad de Medicina, Universidad de Chile.,4Programa de Biología Celular y Molecular, ICBM.,5Departamento de Oncología Básica y Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carlos Chacon
- 1Laboratorio de Genómica Aplicada, Facultad de Medicina, Universidad de Chile
| | | | - Sebastián Sarmiento
- 1Laboratorio de Genómica Aplicada, Facultad de Medicina, Universidad de Chile
| | - Pía Villalobos
- 1Laboratorio de Genómica Aplicada, Facultad de Medicina, Universidad de Chile
| | - Belén Kroxato
- 1Laboratorio de Genómica Aplicada, Facultad de Medicina, Universidad de Chile
| | - Katherine Marcelain
- 3Programa de Genética Humana, ICBM.,5Departamento de Oncología Básica y Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | |
Collapse
|