1
|
Zhang J, Cui J, Gao J, Zhang D, Lin D, Lin J. Polysaccharides of Plantago asiatica enhance antitumor activity via regulating macrophages to M1-like phenotype. Biomed Pharmacother 2023; 159:114246. [PMID: 36652734 DOI: 10.1016/j.biopha.2023.114246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Monocyte-derived macrophages can be polarized into antitumor M1 phenotype, which inhibited the growth of tumors, and immune-suppressive M2 phenotype, which promoted the development and metastasis of tumors. Plantain polysaccharide (PLP), extracted from the Plantago asiatica, has shown its various biological activities. However, the ability of PLP involved in immune regulation was still obscure. Accordingly, we aimed to investigate whether PLP could polarize macrophages and further inhibit 4T1 tumor cells in vivo and in vitro. In this research, in vitro results showed that PLP displayed the potential in polarizing RAW264.7 macrophages into M1 phenotype and indirect inhibiting migratory effect on 4T1 cells. Furthermore, the phagocytosis and the release of reactive oxygen species (ROS) of macrophages were enhanced. In vivo anti-tumor results demonstrated that PLP could effectively inhibit the growth of 4T1 breast tumors by promoting accumulation of macrophages and T cells in the spleen and lymph node. In conclusion, these findings indicated that PLP inhibited the proliferation and progression of breast tumors by accumulating CD4+, CD8+ T cells and M1-like macrophages in lymph node and spleen, and therefore provided an experimental basis for PLP as a potential antitumor adjunctive therapy in preclinical and clinical trials.
Collapse
Affiliation(s)
- Jiatong Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingwen Cui
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiafeng Gao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China; Center of Research and Innovation of Chinese Traditional Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
2
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
3
|
Zhang Z, Li Y, Shi J, Zhu L, Dai Y, Fu P, Liu S, Hong M, Zhang J, Wang J, Jiang C. Lymphocyte-Related Immunomodulatory Therapy with Siponimod (BAF-312) Improves Outcomes in Mice with Acute Intracerebral Hemorrhage. Aging Dis 2022; 14:966-991. [PMID: 37191423 DOI: 10.14336/ad.2022.1102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Modulators of the sphingosine-1-phosphate receptor (S1PR) have been proposed as a promising strategy for treating stroke. However, the detailed mechanisms and the potential translational value of S1PR modulators for intracerebral hemorrhage (ICH) therapy warrant exploration. Using collagenase VII-S-induced ICH in the left striatum of mice, we investigated the effects of siponimod on cellular and molecular immunoinflammatory responses in the hemorrhagic brain in the presence or absence of anti-CD3 monoclonal antibodies (Abs). We also assessed the severity of short- and long-term brain injury and evaluated the efficacy of siponimod in long-term neurologic function. Siponimod treatment significantly decreased brain lesion volume and brain water content on day 3 and the volume of the residual lesion and brain atrophy on day 28. It also inhibited neuronal degeneration on day 3 and improved long-term neurologic function. These protective effects may be associated with a reduction in the expression of lymphotactin (XCL1) and T-helper 1 (Th1)-type cytokines (interleukin 1β and interferon-γ). It may also be associated with inhibition of neutrophil and lymphocyte infiltration and alleviation of T lymphocyte activation in perihematomal tissues on day 3. However, siponimod did not affect the infiltration of natural killer cells (NK) or the activation of CD3-negative immunocytes in perihematomal tissues. Furthermore, it did not influence the activation or proliferation of microglia or astrocytes around the hematoma on day 3. Siponimod appears to have a profound impact on infiltration and activation of T lymphocytes after ICH. The effects of neutralized anti-CD3 Abs-induced T-lymphocyte tolerance on siponimod immunomodulation further confirmed that siponimod alleviated the cellular and molecular Th1 response in the hemorrhagic brain. This study provides preclinical evidence that encourages future investigation of immunomodulators, including siponimod, which target the lymphocyte-related immunoinflammatory reaction in ICH therapy.
Collapse
|
4
|
Chaudhry SR, Kinfe TM, Lamprecht A, Niemelä M, Dobreva G, Hänggi D, Muhammad S. Elevated level of cerebrospinal fluid and systemic chemokine CCL5 is a predictive biomarker of clinical outcome after aneurysmal subarachnoid hemorrhage (aSAH). Cytokine 2020; 133:155142. [PMID: 32485621 DOI: 10.1016/j.cyto.2020.155142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pathophysiology of aneurysmal subarachnoid hemorrhage (aSAH) is highly complex. Bleeding from ruptured aneurysm causes increase in intracranial pressure that disrupts blood-brain barrier leading to infiltration of peripheral immune cells. Interactions between the infiltrated leukocytes and the resident brain cells in the injured tissue mainly determine the delayed tissue damage. Recruitment of leukocytes in the injured brain is mainly mediated by the chemokines. Chemokine C-C motif ligand 5 (CCL5) is a potent pro-inflammatory chemokine shown to be upregulated in preclinical SAH studies. However, detailed clinical investigations exploring the association of cerebrospinal fluid (CSF) and systemic CCL5 and post-aSAH complications and clinical outcome are still lacking. This study investigated CSF and systemic CCL5 after aSAH and its association with clinical outcome and post-aSAH complications. METHODS CSF and serum from control and aSAH patients were obtained after centrifugation of the CSF and peripheral blood, and were preserved at -80 °C until quantification by an enzyme-linked immunoassay. Patient pertinent data, post-aSAH complications and clinical outcome (modified Rankin scale [mRS] and Glasgow outcome scale [GOS]) were retrieved from patient records. RESULTS A significant increase in CSF and serum CCL5 levels was observed on post-aSAH day 1 and day 7 compared to control patients. Dichotomization of patients to poor (mRS 3-6 or GOS 1-3) and good (mRS 0-2 or GOS 4-5) clinical outcomes showed significantly higher serum CCL5 levels in patients with good clinical outcome at discharge, but lower CSF CCL5 levels. Interestingly, significantly lower serum CCL5 levels were observed on post-aSAH day 7 in patients who have additional intracerebral bleeding or the patients who developed chronic hydrocephalus or pneumonia. Whereas, CSF CCL5 levels significantly increased on post-aSAH day 1 in patients developing chronic hydrocephalus, delayed ischemic neurological deficits and intraventricular hemorrhage. CSF CCL5 levels on post-aSAH day 1 were correlated with poor clinical outcome, however, serum CCL5 levels on post-aSAH day 7 were correlated with good clinical outcome. CONCLUSION Systemic and CSF CCL5 levels were elevated after aSAH and levels of serum CCL5 on day 7 were associated independently with clinical outcome (GOS and mRS) at discharge. Therapeutic approaches targeting CCL5 might be beneficial in aSAH.
Collapse
Affiliation(s)
- Shafqat Rasul Chaudhry
- Department of Neurosurgery, University Hospital Bonn, University of Bonn, D-53105 Bonn, Germany; Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, D-53121 Bonn, Germany; Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, 44000 Islamabad, Pakistan
| | - Thomas Mehari Kinfe
- Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, D-53121 Bonn, Germany
| | - Mika Niemelä
- Department of Neurosurgery, University Hospital Helsinki, University of Helsinki, FI-00029 Helsinki, Finland
| | - Gergana Dobreva
- Department of Anatomy and Developmental Biology, Center for Biomedicine and Medical Technology Mannheim (CBTM) and European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Daniel Hänggi
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University of Düsseldorf, D-40225 Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University of Düsseldorf, D-40225 Düsseldorf, Germany; Department of Neurosurgery, University Hospital Bonn, University of Bonn, D-53105 Bonn, Germany; Department of Neurosurgery, University Hospital Helsinki, University of Helsinki, FI-00029 Helsinki, Finland; Department of Anatomy and Developmental Biology, Center for Biomedicine and Medical Technology Mannheim (CBTM) and European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany.
| |
Collapse
|
5
|
|
6
|
Mormile R. Hepatitis B vaccine non response: A predictor of latent autoimmunity? Med Hypotheses 2017; 104:45-47. [PMID: 28673589 DOI: 10.1016/j.mehy.2017.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
Unresponsiveness to Hepatitis B virus (HBV) vaccine has been associated with interleukins involved with Th1 functioning including Interleukin-8 (IL-18) and Interferon-γ (IFN-γ). IL-18 and IFN-γ have also been implicated in the onset of different types of immune-mediate inflammatory conditions such as Type 1 Diabetes (T1D), Celiac disease (CD), rheumatoid arthritis (RA), obesity and systemic lupus erythematosus (SLE). Taking into account that HBV vaccination is provided in the 1st year of life worldwide, I propose that all babies should be tested for anti-HBs response after completion of the vaccine series. And I suggest that children with undetectable anti-HBs titers after recommended immunization schedule as well as the additional booster doses should be followed up over time because they may be at risk of developing a number of autoimmune disorders. In this light, the non-responsiveness to HBV vaccine might be a predictor of latent autoimmunity. For that reason, research studies are needed in order to verify the existence of potential IL-18 and IFN-γ gene polymorphisms to utilize as biomarkers of latent autoimmunity. As a final point, administration of neutralizing antibodies against IFN-γ and/or IL-18 might represent a future target for immune-modulatory therapeutic approach to halt or even reverse autoimmune phenomena.
Collapse
Affiliation(s)
- Raffaella Mormile
- Division of Pediatrics and Neonatology, Moscati Hospital, Aversa, Italy.
| |
Collapse
|
7
|
Gordy JT, Luo K, Zhang H, Biragyn A, Markham RB. Fusion of the dendritic cell-targeting chemokine MIP3α to melanoma antigen Gp100 in a therapeutic DNA vaccine significantly enhances immunogenicity and survival in a mouse melanoma model. J Immunother Cancer 2016; 4:96. [PMID: 28018602 PMCID: PMC5168589 DOI: 10.1186/s40425-016-0189-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/07/2016] [Indexed: 01/17/2023] Open
Abstract
Background Although therapeutic cancer vaccines have been mostly disappointing in the clinic, the advent of novel immunotherapies and the future promise of neoantigen-based therapies have created the need for new vaccine modalities that can easily adapt to current and future developments in cancer immunotherapy. One such novel platform is a DNA vaccine fusing the chemokine Macrophage Inflammatory Protein-3α (MIP-3α) to an antigen, here melanoma antigen gp100. Previous published work has indicated that MIP-3α targets nascent peptides to immature dendritic cells, leading to processing by class I and II MHC pathways. This platform has shown enhanced efficacy in prophylactic melanoma and therapeutic lymphoma model systems. Methods The B16F10 melanoma syngeneic mouse model system was utilized, with a standard therapeutic protocol: challenge with lethal dose of B16F10 cells (5 × 104) on day 0 and then vaccinate by intramuscular electroporation with 50 μg plasmid on days three, 10, and 17. Efficacy was assessed by analysis of tumor burden, tumor growth, and mouse survival, using the statistical tests ANOVA, mixed effects regression, and log-rank, respectively. Immunogenicity was assessed by ELISA and flow cytometric methods, including intracellular cytokine staining to assess vaccine-specific T-cell responses, all tested by ANOVA. Results We demonstrate that the addition of MIP3α to gp100 significantly enhances systemic anti-gp100 immunological parameters. Further, chemokine-fusion vaccine therapy significantly reduces tumor burden, slows tumor growth, and enhances mouse overall survival compared to antigen-only, irrelevant-antigen, and mock vaccines, with efficacy mediated by both CD4+ and CD8+ effector T cells. Antigen-only, irrelevant-antigen, and chemokine-fusion vaccines elicit significantly higher and similar CD4+ and CD8+ tumor-infiltrating lymphocyte (TIL) levels compared to mock vaccine. However, vaccine-specific CD8+ TILs are significantly higher in the chemokine-fusion vaccine group, indicating that the critical step induced by the fusion vaccine construct is the enhancement of vaccine-specific T-cell effectors. Conclusions The current study shows that fusion of MIP3α to melanoma antigen gp100 enhances the immunogenicity and efficacy of a DNA vaccine in a therapeutic B16F10 mouse melanoma model. This study analyzes an adaptable and easily produced MIP3α-antigen modular vaccine platform that could lend itself to a variety of functionalities, including combination treatments and neoantigen vaccination in the pursuit of personalized cancer therapy. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0189-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- James T Gordy
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Kun Luo
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Hong Zhang
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, 251 Bayview, Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Richard B Markham
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| |
Collapse
|
8
|
Mirzaee V, Shahriari J, Hajghani M. CCR5 on the NK Cells and its Ligand (RANTES) Expressions are Disrupted in South-Eastern Iranian Patients With Chronic Hepatitis B Infection. IRANIAN RED CRESCENT MEDICAL JOURNAL 2014; 16:e12458. [PMID: 24910790 PMCID: PMC4028763 DOI: 10.5812/ircmj.12458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/12/2013] [Accepted: 06/15/2013] [Indexed: 11/30/2022]
Abstract
Background: CCR5 is a receptor for CCL3 (MIP-1 α), CCL4 (MIP-1 α) and CCL5 (regulated on activation normal T cell expressed and secreted (RANTES)) and play important roles in recruitment of NK cells to the HBV infected liver. Objectives: The main purpose of this study was to investigate the expression levels of CCR5 on the NK cells and also serum levels of RANTES in chronic HBV infected (CHI) patients. Materials and Methods: In this descriptive study 63 CHI patients and 96 healthy controls were evaluated regarding CCR5 expression on the NK cells and serum levels of RANTES using flow cytometry and ELISA techniques, respectively. Real-Time PCR technique also was used for HBV-DNA quantification. Results: The results revealed that CCR5 expressing NK cells and serum levels of RANTES were decreased significantly in the CHI patients in compare to healthy control. Conclusions: Based on the results it can be concluded that NK cells of Iranian CHI patients are unable to express adequate levels of CCR5 and expression levels of RANTES by immune cells also are defected in CHI patients, hence, the migration of NK cells to the infected hepatocytes and HBV eradication from the cells is interrupted.
Collapse
Affiliation(s)
- Vahid Mirzaee
- Department of Internal Medicine, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, IR Iran
- Corresponding Author: Vahid Mirzaee, Department of Internal Medicine, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, IR Iran, Tel: +98-39182200019, Fax: +98-03918220022, E-mail:
| | - Jahanbano Shahriari
- Hematology Laboratory, Bahonar Hospital, Kerman University of Medical Sciences, Kerman, IR Iran
| | - Masomeh Hajghani
- Hematology Laboratory, Bahonar Hospital, Kerman University of Medical Sciences, Kerman, IR Iran
| |
Collapse
|
9
|
Flingai S, Czerwonko M, Goodman J, Kudchodkar SB, Muthumani K, Weiner DB. Synthetic DNA vaccines: improved vaccine potency by electroporation and co-delivered genetic adjuvants. Front Immunol 2013; 4:354. [PMID: 24204366 PMCID: PMC3816528 DOI: 10.3389/fimmu.2013.00354] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/20/2013] [Indexed: 01/07/2023] Open
Abstract
In recent years, DNA vaccines have undergone a number of technological advancements that have incited renewed interest and heightened promise in the field. Two such improvements are the use of genetically engineered cytokine adjuvants and plasmid delivery via in vivo electroporation (EP), the latter of which has been shown to increase antigen delivery by nearly 1000-fold compared to naked DNA plasmid delivery alone. Both strategies, either separately or in combination, have been shown to augment cellular and humoral immune responses in not only mice, but also in large animal models. These promising results, coupled with recent clinical trials that have shown enhanced immune responses in humans, highlight the bright prospects for DNA vaccines to address many human diseases.
Collapse
Affiliation(s)
- Seleeke Flingai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| | | | | | | | | | | |
Collapse
|
10
|
Role of regulated upon activation normal T-cell expressed and secreted in a model of retransplantation acute rejection mediated by alloreactive memory CD4+ T cells. Transplant Proc 2013; 45:546-51. [PMID: 23498790 DOI: 10.1016/j.transproceed.2012.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/13/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND It is unknown what role Regulated Upon Activation Normal T-Cell Exposed and Secreted may play in retransplantation or T-cell memory-transfer models. The experiment observed the influence of the chemokine RANTES in a mouse model of acute cardiac allograft rejection induced by adoptive transfer of alloreactive CD4(+) memory T (Tm) cells. METHODS Alloreactive CD4(+) Tm cells from spleens of skin-grafted C57BL/6 were adoptively transferred to naïve C57BL/6 recipients prior to heterotopic heart transplantation. We measured the median survival time of cardiac grafts and performed some tests. RESULTS Spleens from skin-grafted C57BL/6 contained 26.83% CD4(+) Tm cells. The median graft survival time of heterotopic heart transplantations (n = 6) was 5.17 ± 0.17 days for hosts receiving CD4(+) Tm cells compared with 7.76 ± 0.21 days among controls (n = 6; P < .001). The mean rejection activity in histological sections of cardiac allografts at day 5 postgrafting was 3.92 ± 0.08 in the CD4(+) Tm cell recipient group (n = 6) compared with 2.67 ± 0.14 in the controls (n = 6; P < .001). Gene expression of Ccl5, interferon (IFN)-γ and interleukin2 was significantly higher among CD4(+) Tm recipients compared with controls. Serum concentrations of RANTES and IFN-γ were higher in the heterotopic heart transplantation group receiving CD4(+) Tm compared with controls. CONCLUSIONS Alloreactive CD4(+) Tm cells contribute to increased expression and secretion of RANTES, and to the Tm and other inflammatory cells migration into the graft.
Collapse
|
11
|
Mohit E, Rafati S. Chemokine-based immunotherapy: delivery systems and combination therapies. Immunotherapy 2013; 4:807-40. [PMID: 22947009 DOI: 10.2217/imt.12.72] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A major role of chemokines is to mediate leukocyte migration through interaction with G-protein-coupled receptors. Various delivery systems have been developed to utilize the chemokine properties for combating disease. Viral and mutant viral vectors expressing chemokines, genetically modified dendritic cells with chemokine or chemokine receptors, engineered chemokine-expressing tumor cells and pDNA encoding chemokines are among these methods. Another approach for inducing a targeted immune response is fusion of a targeting antibody or antibody fragment to a chemokine. In addition, chemokines induce more effective antitumor immunity when used as adjuvants. In this regard, chemokines are codelivered along with antigens or fused as a targeting unit with antigenic moieties. In this review, several chemokines with their role in inducing immune response against different diseases are discussed, with a major emphasis on cancer.
Collapse
Affiliation(s)
- Elham Mohit
- Molecular Immunology & Vaccine Research Lab, Pasteur Institute of Iran, Tehran 13164, Iran
| | | |
Collapse
|
12
|
Immunomodulatory effects of IP-10 chemokine along with PEI600-Tat delivery system in DNA vaccination against HPV infections. Mol Immunol 2013; 53:149-60. [DOI: 10.1016/j.molimm.2012.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 07/18/2012] [Accepted: 07/26/2012] [Indexed: 12/23/2022]
|
13
|
Al-Qahtani A, Alarifi S, Al-Okail M, Hussain Z, Abdo A, Sanai F, Al-Anazi M, Khalaf N, Al-Humaidan H, Al-Ahdal M, Almajhdi FN. RANTES gene polymorphisms (-403G>A and -28C>G) associated with hepatitis B virus infection in a Saudi population. GENETICS AND MOLECULAR RESEARCH 2012; 11:855-62. [PMID: 22576913 DOI: 10.4238/2012.april.10.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Besides the host immune response, genetic and environmental factors play crucial roles in the manifestation of hepatitis B virus (HBV) infection. "Regulated on activation normal T-cell expressed and secreted" factor (RANTES) plays a vital role in CD4(+), CD8(+) T-lymphocyte and dendritic cell activation and proliferation in inflammation. Single nucleotide polymorphisms (SNPs) in the RANTES gene are associated with several viral and non-viral diseases. Association studies have invariably indicated a lack of association between RANTES gene SNPs and HBV infection in ethnic populations, even though RANTES gene SNPs exhibit distinct ethnic distributions. Despite the high prevalence of HBV infections in Saudi Arabia, no studies have been made concerning a possible relationship between RANTES gene polymorphisms and susceptibility to and progression of HBV infection. We examined -403G>A and -28C>G RANTES gene variants in 473 healthy controls and 484 HBV patients in ethnic Saudi populations. Significant differences were found in the genotype and allele distributions of the SNPs between the controls and the HBV patients. Both SNPs were significantly linked to viral clearance in these subjects. Our data demonstrate for the first time in a Saudi population, a relationship between the RANTES gene polymorphisms and the clinical course of HBV infection and underscore the importance of evaluating the genetic background of the affected individual to determine how it may affect disease progression.
Collapse
Affiliation(s)
- A Al-Qahtani
- Department of Biological and Medical Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Despite many years of research, human DNA vaccines have yet to fulfill their early promise. Over the past 15 years, multiple generations of DNA vaccines have been developed and tested in preclinical models for prophylactic and therapeutic applications in the areas of infectious disease and cancer, but have failed in the clinic. Thus, while DNA vaccines have achieved successful licensure for veterinary applications, their poor immunogenicity in humans when compared with traditional protein-based vaccines has hindered their progress. Many strategies have been attempted to improve DNA vaccine potency including use of more efficient promoters and codon optimization, addition of traditional or genetic adjuvants, electroporation, intradermal delivery and various prime-boost strategies. This review summarizes these advances in DNA vaccine technologies and attempts to answer the question of when DNA vaccines might eventually be licensed for human use.
Collapse
Affiliation(s)
- Fadi Saade
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia
- Department of Diabetes and Endocrinology, Flinders Medical Centre/Flinders University, Adelaide 5042, Australia
| |
Collapse
|
15
|
Arathy DS, Nair S, Soman SS, Issac A, Sreekumar E. Functional characterization of the CC chemokine RANTES from Pekin duck (Anas platyrhynchos). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:142-150. [PMID: 20850473 DOI: 10.1016/j.dci.2010.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 09/08/2010] [Accepted: 09/08/2010] [Indexed: 05/29/2023]
Abstract
RANTES (Regulated upon Activation, Normal T-cell Expressed and Secreted) is a key pro-inflammatory cytokine that belongs to the CC-group of chemokines. The present study was carried out to functionally characterize the previously identified RANTES homologue in domestic duck (GenBank Accession No. AY641435). Recombinant duck RANTES was expressed in Escherichia coli-based and HEK293T cell-based systems. A tRNA supplementation strategy was required to express the protein in E. coli due to the presence of rare codons. In biological assays using HEK293T cell-expressed protein, RANTES was found to mediate chemotaxis of DT-40 chicken B cells and primary duck splenocytes at a concentration of 0.505μg/ml (0.6μM). Immunostaining of the migrated splenocytes using anti-duck CD4 and CD8 monoclonal antibodies and subsequent flow cytometric analysis showed enhanced chemotaxis of CD8+ cells. The recombinant RANTES exhibited in vitro antiviral activity by inhibiting infection of chicken embryo fibroblast cells with duck enteritis virus (DEV) at the same concentration. The effect could be neutralized by rabbit anti-duck RANTES polyclonal serum. The mechanism seems to be direct on viral particles as evidenced by the need for co-incubation of RANTES with DEV prior to the infection for antiviral activity, and also by the enhanced binding of DEV to E. coli expressed purified RANTES on ELISA-based assays. Our results show that the duck RANTES has overlapping biological properties with its mammalian orthologue, and also has possible functional cross-reactivity with chicken immune cells indicated by the chemotaxis of DT-40 cells.
Collapse
Affiliation(s)
- D S Arathy
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram 695014, Kerala, India
| | | | | | | | | |
Collapse
|
16
|
Li J, Diao H, Zhao D, Zhang J. Reduced tumourigenicity of EG7 after RANTES gene transfer and the underlying mechanism. Arch Med Sci 2010; 6:829-36. [PMID: 22427753 PMCID: PMC3302691 DOI: 10.5114/aoms.2010.19287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/28/2010] [Accepted: 08/25/2010] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Chemokine ligand 5, also known as CCL5 or regulated on activation normal T-cell expressed and secreted (RANTES), is a chemokine expressed in inflamed tissue and capable of inducing migration of immature dendritic cells (DCs) or Langerhans cells. In this study, we explored the effect of RANTES on EG7 cells. MATERIAL AND METHODS In vivo, RANTES gene transfer reduced the tumourigenic capacity of EG7 and prolonged the survival of tumour-bearing mice. To reveal the underlying mechanism, we performed the following experiments and provided evidence to support our hypothesis of RANTES gene therapy for EG7. Higher natural killer (NK) cell and cytotoxic T lymphocyte (CTL) activity was induced after RANTES gene transfer, accompanied by higher levels of Th1 type cytokines (IL-2 and IFN-γ). RESULTS Tumour necrosis was also markedly observed in the tumour tissues after RANTES gene transfer, which was attributed to reduced expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP-2). CONCLUSIONS We draw the conclusion that reduced tumourigenicity of EG7 after RANTES gene transfer can be attributed to higher NK cell and CTL activity, anti-angiogenesis and higher levels of Th1 type cytokines induced by RANTES. These results support the notion that higher chemokine expression in tumour tissue elicits potent anti-tumour immunity.
Collapse
Affiliation(s)
- Jiuzhou Li
- People’s Hospital of Binzhou City, Binzhou, China
| | - Huiling Diao
- Department of Physiology, Binzhou Medical College, Binzhou, China
| | - Dongmei Zhao
- Department of Anatomy, Binzhou Medical College, Binzhou, China
| | - Jianbin Zhang
- The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
17
|
Kallal LE, Schaller MA, Lindell DM, Lira SA, Lukacs NW. CCL20/CCR6 blockade enhances immunity to RSV by impairing recruitment of DC. Eur J Immunol 2010; 40:1042-52. [PMID: 20101616 DOI: 10.1002/eji.200939778] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chemokines are important mediators of the immune response to pathogens, but can also promote chronic inflammatory states. Chemokine receptor 6 (CCR6) is found on immature DC and effector/memory T cells, and binds a single ligand, CCL20, with high affinity. Here, we investigated the role of CCL20 and CCR6 in a pulmonary viral infection caused by RSV, a ubiquitous virus that can cause severe pulmonary complications. Neutralization of CCL20 during RSV infection significantly reduced lung pathology and favored a Th1 effector response. CCR6-deficient animals recapitulated this phenotype, and additionally showed enhanced viral clearance when compared with WT mice. No differences were observed in migration of T cells to the lungs of CCR6(-/-) animals; however, a significant reduction was observed in numbers of conventional DC (cDC), but not plasmacytoid DC, in CCR6(-/-) mice. A pathogenic phenotype could be reconstituted in CCR6(-/-) mice by supplying cDC into the airway, indicating that mere number of cDC dictates the adverse response. Our data suggest that blockade of the CCL20/CCR6 pathway provides an environment whereby the attenuated recruitment of cDC alters the balance of innate immune cells and mediates the efficient antiviral response to RSV.
Collapse
Affiliation(s)
- Lara E Kallal
- Department of Molecular & Cellular Pathology, The University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
18
|
Abstract
The interaction between immune responses and hepatitis B virus (HBV) is coordinated between innate and adaptive immunity. Anti-HBs antibodies protect the host by blocking the binding ability of HBV. Anti-HBc antibodies are detected with persistent HBV infection. The presence of anti-HBe antibodies is often associated with recovery from active diseases and is clinically used as a benchmark to assess response to treatment. Our studies have revealed that the anti-HBV immunoglobulins secreted are different in subclass patterns in different HBV infection status populations. These revelations may help to understand HBV escape and persistent infection and to develop strategies for prevention and therapeutic management of HBV infection.
Collapse
Affiliation(s)
- Hsiu-Ting Tsai
- School of Nursing, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | | | | | | |
Collapse
|
19
|
Song S, Liu C, Wang J, Zhang Y, You H, Wang Y, Liu F, Sun S. Vaccination with combination of Fit3L and RANTES in a DNA prime-protein boost regimen elicits strong cell-mediated immunity and antitumor effect. Vaccine 2008; 27:1111-8. [PMID: 19100302 DOI: 10.1016/j.vaccine.2008.11.095] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 11/22/2008] [Indexed: 12/21/2022]
Abstract
With accumulating evidence indicating the importance of cytotoxic T lymphocytes (CTLs) in the antitumor response, strategies are being pursued to elicit augmented CD8(+) T-cell responses against tumors with tumor vaccines. Here, we report the protective efficacy of vaccine-elicited antitumor immune responses with an aggressive HBc-expressing B16-HBc melanoma, which expressed HBc as a self and model antigen, tumor model. We demonstrated that the significantly better memory responses or marked inhibition on tumor growth could be achieved after coadministration of cytokine adjuvants RANTES and Flt3L in a DNA prime-protein boost regimen. Furthermore, the augmentation of DNA prime-protein boost regimens by cytokines gene was due to the improvement the immunopotency of DNA vaccine and subsequently the augmented Ag-specific and IFN-gamma mediating CD8(+) T-cell responses after protein boosting. Hence, this study demonstrates for the first time that combinatorial use of chemotactic and potent DC-specific growth factor molecules provides a useful strategy for enhancing antitumor responses.
Collapse
Affiliation(s)
- Shuxia Song
- Department of Molecular Biology and Key Lab of Laboratory Animal, Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | | | | | |
Collapse
|