1
|
Liu Q, Deng G, Jiang X, Fu Y, Zhang J, Wu X, Li X, Ai J, Liu H, Tan G. Macrophage-mediated activation of the IL4I1/AhR axis is a key player in allergic rhinitis. Int Immunopharmacol 2025; 152:114439. [PMID: 40080924 DOI: 10.1016/j.intimp.2025.114439] [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: 11/07/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Epidemiological evidence suggests that environmental pollutants precipitate the occurrence of allergic rhinitis (AR). The aryl hydrocarbon receptor (AhR), a receptor or sensor for various contaminants, is closely related to immunomodulation and the polarization of M2 macrophages. However, the mechanisms involving AhR and M2 macrophages in AR remain unclear. METHODS Bioinformatics analysis of GEO datasets (GSE180697 and GSE180697) assessed AhR and IL4I1 expression levels, which were then verified in the nasal mucosa, monocytes and serum of patients with AR using western blotting, quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence, and enzyme-linked immunosorbent assay (ELISA). Primary human mononuclear cells were isolated from peripheral blood using a magnetic separation technique, and THP-1 cell lines with IL4I1 overexpression or downexpression were established through lentiviral constructs. M2 macrophages were induced with the cytokines CSF, IL4 and IL13 and then treated with the AhR agonist FICZ or inhibitor CH223191. The polarization of M2 macrophages was measured by flow cytometry and western blotting. Furthermore, primary nasal epithelial cells and macrophages were co-cultured to assess the epithelial-mesenchymal transition (EMT) in epithelial cells. The AR murine model was established using ovalbumin (OVA). Inflammation within the nasal mucosa and lung tissue was examined after CH223191 or IL4I1 treatment. RESULTS Nuclear translocation of AhR and upregulation of IL4I1 was observed in peripheral mononuclear cells and nasal mucosal tissue of patients with AR. Through the activation of AhR, IL4I1 promoted M2 macrophage polarization. Furthermore, modulation of the IL4I1/AhR axis regulated the migratory impact of OVA on T-M2 cells. The IL4I1/AhR axis was involved in the regulation of M2 macrophage-associated EMT and contributed to the expression of IL-33 and STAT6 phosphorylation in epithelial cells. In AR mice, increased AhR nuclear translocation and higher expression of IL4I1 and the M2 macrophage marker CD206 in the lungs was observed. The IL4I1/AhR axis exacerbated allergic symptoms in AR mice, fostering allergic inflammation within the nasal mucosa and lungs. CONCLUSIONS The IL4I1/AhR axis is activated within the mononuclear phagocyte system of patients with AR. This activation facilitates the polarization of mononuclear cells into M2 macrophages, which further aggravates EMT in epithelial cells and exacerbates inflammation in AR. This study may provide novel strategies for the precise treatment of AR.
Collapse
Affiliation(s)
- Qian Liu
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China; Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Guohao Deng
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xian Jiang
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yanpeng Fu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Afliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China
| | - Jian Zhang
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xue Wu
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xinlong Li
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Jingang Ai
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Honghui Liu
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| | - Guolin Tan
- Department of Otolaryngology - Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
2
|
Wechsler R, Bendele A, Pinkus R, Sarfati G, Rotem R, Kasem H, Turjeman K, Barenholz Y. Empty large liposomes reduce cartilage degeneration in osteoarthritic rats by forming a lubricative coating. Osteoarthritis Cartilage 2025:S1063-4584(25)00815-5. [PMID: 39978572 DOI: 10.1016/j.joca.2025.02.774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE To explore the mechanism of action and structural effects of MM-II, a dispersion of "empty" multilamellar large liposomes composed of dimyristoyl phosphatidylcholine (DMPC) and dipalmitoyl phosphatidylcholine (DPPC), which durably reduced pain in a phase 2b study in knee osteoarthritis (OA) patients. METHOD MM-II liposomes were manufactured using a defined ratio of DMPC and DPPC, resulting in a lipid phase transition temperature range overlapping with the temperature of human OA knees. MM-II cartilage coating in the presence and absence of compression load was assessed using labeled MM-II. Lubrication of damaged cartilage by MM-II was tested in cartilage-on-glass friction tests and pin-on-disc wear tests. Knee distribution of intra-articularly injected MM-II was assessed in healthy and OA rabbit knees. Structural effects were assessed using a rat OA model comparing to DPPC liposomes, DMPC liposomes, a mixture of DPPC and DMPC liposomes, and vehicle. RESULTS In a pin-on-disc model, MM-II reduced cartilage wear by up to 44% compared to control. MM-II liposomes bound to cartilage discs preferentially under compression load. Coating of cartilage and menisci with MM-II was also observed in both healthy and OA rabbit knees. Cartilage-bound MM-II efficiently lubricated damaged cartilage discs. In a rat OA model, MM-II demonstrated 53% reduction in tibial cartilage degeneration and the least associated mononuclear cell recruitment compared to other groups. CONCLUSIONS Reduction in cartilage degeneration by MM-II in OA rats is likely mediated through formation of a lubricative layer at the cartilage surface, though additional mechanisms could be mediating pain reduction demonstrated in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Haytam Kasem
- Azrieli College of Engineering, Jerusalem, Israel.
| | - Keren Turjeman
- The Laboratory of Membrane and Liposome Research IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| | - Yechezkel Barenholz
- The Laboratory of Membrane and Liposome Research IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
3
|
Shi L, Gu H. Cell membrane-camouflaged liposomes and neopeptide-loaded liposomes with TLR agonist R848 provides a prime and boost strategy for efficient personalized cancer vaccine therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102648. [PMID: 36584738 DOI: 10.1016/j.nano.2022.102648] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/01/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Recent advances in bioinformatics and nanotechnology offer great opportunities for personalized cancer vaccine development. However, the timely identification of neoantigens and unsatisfactory efficacy of therapeutic cancer vaccines remain two obstacles for clinical transformation. We propose a "prime and boost" strategy to facilitate neoantigen-based immunotherapy. To prime the immune system, we first constructed personalized liposomes with cancer cell membranes and adjuvant R848 to provide immunostimulatory efficacy and time for identifying tumor antigens. Liposomes loaded with personalized neopeptides and adjuvants were used to boost the immune response. In vitro experiments verified potent immune responses, including macrophage polarization, dendritic cell maturation, and T lymphocyte activation. In vivo B16F10 and TC-1 cancer model were used to investigate efficient tumor growth suppression. Liposomal vaccines with neopeptides could stimulate human dendritic cells and T lymphocytes in vitro. These results demonstrate that the "prime and boost" strategy provides simple, quick, and efficient personalized vaccines for cancer therapy.
Collapse
Affiliation(s)
- Lu Shi
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Hongchen Gu
- Nano Biomedical Research Center, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| |
Collapse
|
4
|
Giordano G, Teresa Bochicchio M, Niro G, Lucchesi A, Napolitano M. Genetic regulation of iron homeostasis in sideropenic patients with mild COVID-19 disease under a new oral iron formulation: Lessons from a different perspective. Immunobiology 2022; 227:152297. [PMID: 36327544 PMCID: PMC9597571 DOI: 10.1016/j.imbio.2022.152297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022]
Abstract
Background Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) needs iron to replicate itself. Coronaviruses are able to upregulate Chop/Gadd153 and Arg1 genes, consequently leading to CD8 lymphocytes decrease, degradation of asparagine and decreased nitric oxide (NO), thus impairing immune response and antithrombotic functions. Little is known about regulation of genes involved in iron metabolism in paucisymptomatic patients with COVID-19 disease or in patients with iron deficiency treated with sucrosomial iron. Methods Whole blood was taken from the COVID-19 patients and from patients with sideropenic anemia, treated or not (control group) with iron supplementations. Enrolled patients were: affected by COVID19 under sucrosomal iron support (group A), affected by COVID-19 not under oral iron support (group B), iron deficiency not under treatment, not affected by COVID19 (control group). After RNA extraction and complementary DNA (cDNA) synthesis of Arg1, Hepcidin and Chop/Gadd153, gene expression from the 3 groups was measured by qRT-PCR. M2 macrophages were detected by cytofluorimetry using CD163 and CD14 markers. Results Forty patients with COVID-19 (group A), 20 patients with iron deficiency treated with sucrosomial iron (group B) and 20 patients with iron deficiency not under treatment (control group) were enrolled. In all the patients supported with oral sucrosomial iron, the gene expression of Chop, Arg1 and Hepcidin genes was lower than in sideropenic patients not supported with iron, M1 macrophages polarization and functional iron deficiency was also lower in group A and B, than observed in the control group. Conclusions New oral iron formulations, as sucrosomial iron, are able to influence the expression of genes like Chop and Arg1 and to influence M2 macrophage polarization mainly in the early phase of COVID-19 disease.
Collapse
Affiliation(s)
- Giulio Giordano
- Division of Internal Medicine, Hematology Service, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Giovanna Niro
- Division of Laboratory Medicine, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, Meldola (FC), Italy,Corresponding author
| | - Mariasanta Napolitano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), Haematology Unit, University Hospital “P. Giaccone”, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
5
|
Hallan SS, Sguizzato M, Esposito E, Cortesi R. Challenges in the Physical Characterization of Lipid Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13040549. [PMID: 33919859 PMCID: PMC8070758 DOI: 10.3390/pharmaceutics13040549] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Nano-sized drug transporters have become an efficient approach with considerable commercial values. Nanomedicine is not only limited to drug delivery by means of different administration routes, such as intravenous, oral, transdermal, nasal, pulmonary, and more, but also has applications in a multitude of areas, such as a vaccine, antibacterial, diagnostics and imaging, and gene delivery. This review will focus on lipid nanosystems with a wide range of applications, taking into consideration their composition, properties, and physical parameters. However, designing suitable protocol for the physical evaluation of nanoparticles is still conflicting. The main obstacle is concerning the sensitivity, reproducibility, and reliability of the adopted methodology. Some important techniques are compared and discussed in this report. Particularly, a comparison between different techniques involved in (a) the morphologic characterization, such as Cryo-TEM, SEM, and X-ray; (b) the size measurement, such as dynamic light scattering, sedimentation field flow fractionation, and optical microscopy; and (c) surface properties, namely zeta potential measurement, is described. In addition, an amperometric tool in order to investigate antioxidant activity and the response of nanomaterials towards the skin membrane has been presented.
Collapse
Affiliation(s)
- Supandeep Singh Hallan
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy; (S.S.H.); (M.S.); (E.E.)
| | - Maddalena Sguizzato
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy; (S.S.H.); (M.S.); (E.E.)
| | - Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy; (S.S.H.); (M.S.); (E.E.)
| | - Rita Cortesi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, I-44121 Ferrara, Italy; (S.S.H.); (M.S.); (E.E.)
- Biotechnology Interuniversity Consortium (C.I.B.), Ferrara Section, University of Ferrara, I-44121 Ferrara, Italy
- Correspondence:
| |
Collapse
|
6
|
Immunological and Toxicological Considerations for the Design of Liposomes. NANOMATERIALS 2020; 10:nano10020190. [PMID: 31978968 PMCID: PMC7074910 DOI: 10.3390/nano10020190] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/25/2022]
Abstract
Liposomes hold great potential as gene and drug delivery vehicles due to their biocompatibility and modular properties, coupled with the major advantage of attenuating the risk of systemic toxicity from the encapsulated therapeutic agent. Decades of research have been dedicated to studying and optimizing liposomal formulations for a variety of medical applications, ranging from cancer therapeutics to analgesics. Some effort has also been made to elucidate the toxicities and immune responses that these drug formulations may elicit. Notably, intravenously injected liposomes can interact with plasma proteins, leading to opsonization, thereby altering the healthy cells they come into contact with during circulation and removal. Additionally, due to the pharmacokinetics of liposomes in circulation, drugs can end up sequestered in organs of the mononuclear phagocyte system, affecting liver and spleen function. Importantly, liposomal agents can also stimulate or suppress the immune system depending on their physiochemical properties, such as size, lipid composition, pegylation, and surface charge. Despite the surge in the clinical use of liposomal agents since 1995, there are still several drawbacks that limit their range of applications. This review presents a focused analysis of these limitations, with an emphasis on toxicity to healthy tissues and unfavorable immune responses, to shed light on key considerations that should be factored into the design and clinical use of liposomal formulations.
Collapse
|
7
|
Nanomaterials for direct and indirect immunomodulation: A review of applications. Eur J Pharm Sci 2020; 142:105139. [DOI: 10.1016/j.ejps.2019.105139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 01/03/2023]
|
8
|
Reichel D, Tripathi M, Perez JM. Biological Effects of Nanoparticles on Macrophage Polarization in the Tumor Microenvironment. Nanotheranostics 2019; 3:66-88. [PMID: 30662824 PMCID: PMC6328304 DOI: 10.7150/ntno.30052] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/02/2018] [Indexed: 12/11/2022] Open
Abstract
Biological interactions between tumor-associated macrophages (TAMs), cancer cells and other cells within the tumor microenvironment contribute to tumorigenesis, tumor growth, metastasis and therapeutic resistance. TAMs can remodel the tumor microenvironment to reduce growth barriers such as the dense extracellular matrix and shift tumors towards an immunosuppressive microenvironment that protects cancer cells from targeted immune responses. Nanoparticles can interrupt these biological interactions within tumors by altering TAM phenotypes through a process called polarization. Macrophage polarization within tumors can shift TAMs from a growth-promoting phenotype towards a cancer cell-killing phenotype that predicts treatment efficacy. Because many types of nanoparticles have been shown to preferentially accumulate within macrophages following systemic administration, there is considerable interest in identifying nanoparticle effects on TAM polarization, evaluating nanoparticle-induced TAM polarization effects on cancer treatment using drug-loaded nanoparticles and identifying beneficial types of nanoparticles for effective cancer treatment. In this review, the macrophage polarization effects of nanoparticles will be described based on their primary chemical composition. Because of their strong macrophage-polarizing and antitumor effects compared to other types of nanoparticles, the effects of iron oxide nanoparticles on macrophages will be discussed in detail. By comparing the macrophage polarization effects of various nanoparticle treatments reported in the literature, this review aims to both elucidate nanoparticle material effects on macrophage polarization and to provide insight into engineering nanoparticles with more beneficial immunological responses for cancer treatment.
Collapse
Affiliation(s)
- Derek Reichel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Manisha Tripathi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Current Address: Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - J. Manuel Perez
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
9
|
Ordoñez C, Savage HP, Tarajia M, Rivera R, Weeks‐Galindo C, Sambrano D, Riley L, Fernandez PL, Baumgarth N, Goodridge A. Both B-1a and B-1b cells exposed to Mycobacterium tuberculosis lipids differentiate into IgM antibody-secreting cells. Immunology 2018; 154:613-623. [PMID: 29455451 PMCID: PMC6050208 DOI: 10.1111/imm.12909] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/29/2018] [Accepted: 02/08/2018] [Indexed: 01/22/2023] Open
Abstract
Tuberculosis is an infectious disease caused by Mycobacterium tuberculosis. The cellular immune response to mycobacteria has been characterized extensively, but the antibody response remains underexplored. The present study aimed to examine whether host or bacterial phospholipids induce secretion of IgM, and specifically anti-phospholipid IgM, antibodies by B cells and to identify the responsible B-cell subset. Here we show that peritoneal B cells responded to lipid antigens by secreting IgM antibodies. Specifically, stimulation with M. tuberculosis H37Rv total lipids resulted in significant induction of total and anti-phosphatidylcholine IgM. Similarly, IgM antibody production increased significantly with stimulation by whole Mycobacterium bovis bacillus Calmette-Guérin. The B-1 subset was the dominant source of IgM antibodies after exposure to cardiolipin. Both CD5+ B-1a and CD5- B-1b cell subsets secreted total IgM antibodies after exposure to M. tuberculosis H37Rv total lipids in vitro. Overall, our results suggest that the poly-reactive B-1 cell repertoire contributes to non-specific anti-phospholipid IgM antibody secretion in response to M. tuberculosis lipids.
Collapse
Affiliation(s)
- Ciara Ordoñez
- Tuberculosis Biomarker Research UnitCentro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
- Department of BiotechnologyAcharya Nargajuna UniversityGunturIndia
| | - Hannah P. Savage
- Graduate Group in ImmunologyUniversity of CaliforniaDavisCAUSA
- Center for Comparative MedicineUniversity of CaliforniaDavisCAUSA
| | - Musharaf Tarajia
- Tuberculosis Biomarker Research UnitCentro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
- Department of BiotechnologyAcharya Nargajuna UniversityGunturIndia
| | - René Rivera
- Centro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
| | - Cheyenne Weeks‐Galindo
- Tuberculosis Biomarker Research UnitCentro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
- Sentara RMH Medical CenterHarrisonburgVAUSA
| | - Dilcia Sambrano
- Tuberculosis Biomarker Research UnitCentro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
| | - Lee Riley
- Division of Infectious Diseases and VaccinologySchool of Public HealthUniversity of CaliforniaBerkeleyCAUSA
| | - Patricia L. Fernandez
- Centro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
| | - Nicole Baumgarth
- Graduate Group in ImmunologyUniversity of CaliforniaDavisCAUSA
- Center for Comparative MedicineUniversity of CaliforniaDavisCAUSA
- Department of Pathology, Microbiology and ImmunologyUniversity of CaliforniaDavisCAUSA
| | - Amador Goodridge
- Tuberculosis Biomarker Research UnitCentro de Biología Molecular y Celular de Enfermedades‐Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT‐AIP)CIUDAD DEL SABERClaytonPanama
| |
Collapse
|
10
|
Liposome-induced immunosuppression and tumor growth is mediated by macrophages and mitigated by liposome-encapsulated alendronate. J Control Release 2017; 271:139-148. [PMID: 29277680 DOI: 10.1016/j.jconrel.2017.12.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022]
Abstract
Liposomal nanoparticles are the most commonly used drug nano-delivery platforms. However, recent reports show that certain pegylated liposomal nanoparticles (PLNs) and polymeric nanoparticles have the potential to enhance tumor growth and inhibit antitumor immunity in murine cancer models. We sought herein to identify the mechanisms and determine whether PLN-associated immunosuppression and tumor growth can be reversed using alendronate, an immune modulatory drug. By conducting in vivo and ex vivo experiments with the immunocompetent TC-1 murine tumor model, we found that macrophages were the primary cells that internalized PLN in the tumor microenvironment and that PLN-induced tumor growth was dependent on macrophages. Treatment with PLN increased immunosuppression as evidenced by increased expression of arginase-1 in CD11b+Gr1+ cells, diminished M1 functionality in macrophages, and globally suppressed T-cell cytokine production. Encapsulating alendronate in PLN reversed these effects on myeloid cells and shifted the profile of multi-cytokine producing T-cells towards an IFNγ+ perforin+ response, suggesting increased cytotoxic functionality. Importantly, we also found that PLN-encapsulated alendronate (PLN-alen), but not free alendronate, abrogated PLN-induced tumor growth and increased progression-free survival. In summary, we have identified a novel mechanism of PLN-induced tumor growth through macrophage polarization and immunosuppression that can be targeted and inactivated to improve the anticancer efficacy of PLN-delivered drugs. Importantly, we also determined that PLN-alen not only reversed protumoral effects of the PLN carrier, but also had moderate antitumor activity. Our findings strongly support the inclusion of immune-responsive tumor models and in-depth immune functional studies in the preclinical drug development paradigm for cancer nanomedicines, and the further development of chemo-immunotherapy strategies to co-deliver alendronate and chemotherapy for the treatment of cancer.
Collapse
|
11
|
Laborde RJ, Sanchez-Ferras O, Luzardo MC, Cruz-Leal Y, Fernández A, Mesa C, Oliver L, Canet L, Abreu-Butin L, Nogueira CV, Tejuca M, Pazos F, Álvarez C, Alonso ME, Longo-Maugéri IM, Starnbach MN, Higgins DE, Fernández LE, Lanio ME. Novel Adjuvant Based on the Pore-Forming Protein Sticholysin II Encapsulated into Liposomes Effectively Enhances the Antigen-Specific CTL-Mediated Immune Response. THE JOURNAL OF IMMUNOLOGY 2017; 198:2772-2784. [PMID: 28258198 DOI: 10.4049/jimmunol.1600310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022]
Abstract
Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.
Collapse
Affiliation(s)
- Rady J Laborde
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Oraly Sanchez-Ferras
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María C Luzardo
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Yoelys Cruz-Leal
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Audry Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Circe Mesa
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liliana Oliver
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liem Canet
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Liane Abreu-Butin
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Catarina V Nogueira
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Mayra Tejuca
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Fabiola Pazos
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Carlos Álvarez
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María E Alonso
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Ieda M Longo-Maugéri
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Darren E Higgins
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Luis E Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba;
| | - María E Lanio
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba;
| |
Collapse
|
12
|
Ghosh S, Mukherjee S, Choudhury S, Gupta P, Adhikary A, Baral R, Chattopadhyay S. Reactive oxygen species in the tumor niche triggers altered activation of macrophages and immunosuppression: Role of fluoxetine. Cell Signal 2015; 27:1398-412. [DOI: 10.1016/j.cellsig.2015.03.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/05/2015] [Indexed: 02/07/2023]
|
13
|
Yan B, Xie S, Liu Z, Ran J, Li Y, Wang J, Yang Y, Zhou J, Li D, Liu M. HDAC6 deacetylase activity is critical for lipopolysaccharide-induced activation of macrophages. PLoS One 2014; 9:e110718. [PMID: 25330030 PMCID: PMC4199742 DOI: 10.1371/journal.pone.0110718] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/15/2014] [Indexed: 01/01/2023] Open
Abstract
Activated macrophages play an important role in both innate and adaptive immune responses, and aberrant activation of macrophages often leads to inflammatory and immune disorders. However, the molecular mechanisms of how macrophages are activated are not fully understood. In this study, we identify a novel role for histone deacetylse 6 (HDAC6) in lipopolysaccharide (LPS)-induced macrophage activation. Our data show that suppression of HDAC6 activity significantly restrains LPS-induced activation of macrophages and production of pro-inflammatory cytokines. Further study reveals that the regulation of macrophage activation by HDAC6 is independent of F-actin polymerization and filopodium formation; instead, it is mediated by the effects of HDAC6 on cell adhesion and microtubule acetylation. These data thus suggest that HDAC6 is an important regulator of LPS-induced macrophage activation and might be a potential target for the management of inflammatory disorders.
Collapse
Affiliation(s)
- Bing Yan
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Songbo Xie
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhu Liu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Ran
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuanyuan Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jian Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yang Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Min Liu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|