1
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
2
|
Kumar V, Kaushik NK, Tiwari SK, Singh D, Singh B. Green synthesis of iron nanoparticles: Sources and multifarious biotechnological applications. Int J Biol Macromol 2023:127017. [PMID: 37742902 DOI: 10.1016/j.ijbiomac.2023.127017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Green synthesis of iron nanoparticles is a highly fascinating research area and has gained importance due to reliable, sustainable and ecofriendly protocol for synthesizing nanoparticles, along with the easy availability of plant materials and their pharmacological significance. As an alternate to physical and chemical synthesis, the biological materials, like microorganisms and plants are considered to be less costly and environment-friendly. Iron nanoparticles with diverse morphology and size have been synthesized using biological extracts. Microbial (bacteria, fungi, algae etc.) and plant extracts have been employed in green synthesis of iron nanoparticles due to the presence of various metabolites and biomolecules. Physical and biochemical properties of biologically synthesized iron nanoparticles are superior to that are synthesized using physical and chemical agents. Iron nanoparticles have magnetic property with thermal and electrical conductivity. Iron nanoparticles below a certain size (generally 10-20 nm), can exhibit a unique form of magnetism called superparamagnetism. They are non-toxic and highly dispersible with targeted delivery, which are suitable for efficient drug delivery to the target. Green synthesized iron nanoparticles have been explored for multifarious biotechnological applications. These iron nanoparticles exhibited antimicrobial and anticancerous properties. Iron nanoparticles adversely affect the cell viability, division and metabolic activity. Iron nanoparticles have been used in the purification and immobilization of various enzymes/proteins. Iron nanoparticles have shown potential in bioremediation of various organic and inorganic pollutants. This review describes various biological sources used in the green synthesis of iron nanoparticles and their potential applications in biotechnology, diagnostics and mitigation of environmental pollutants.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Biotechnology, Central University of Haryana, Jant-Pali, Mahendergarh 123031, Haryana, India
| | - Naveen Kumar Kaushik
- Amity Institute of Virology and Immunology, Amity University, Sector-125, Noida 201313, U.P., India
| | - S K Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Davender Singh
- Department of Physics, RPS Degree College, Balana, Satnali Road, Mahendragarh 123029, Haryana, India
| | - Bijendar Singh
- Department of Biotechnology, Central University of Haryana, Jant-Pali, Mahendergarh 123031, Haryana, India; Laboratory of Bioprocess Technology, Department of Microbiology, Maharishi Dayanand University, Rohtak 124001, Haryana, India.
| |
Collapse
|
3
|
Matos AI, Peres C, Carreira B, Moura LIF, Acúrcio RC, Vogel T, Wegener E, Ribeiro F, Afonso MB, Santos FMF, Martínez‐Barriocanal Á, Arango D, Viana AS, Góis PMP, Silva LC, Rodrigues CMP, Graca L, Jordan R, Satchi‐Fainaro R, Florindo HF. Polyoxazoline-Based Nanovaccine Synergizes with Tumor-Associated Macrophage Targeting and Anti-PD-1 Immunotherapy against Solid Tumors. Adv Sci (Weinh) 2023; 10:e2300299. [PMID: 37434063 PMCID: PMC10477894 DOI: 10.1002/advs.202300299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Immune checkpoint blockade reaches remarkable clinical responses. However, even in the most favorable cases, half of these patients do not benefit from these therapies in the long term. It is hypothesized that the activation of host immunity by co-delivering peptide antigens, adjuvants, and regulators of the transforming growth factor (TGF)-β expression using a polyoxazoline (POx)-poly(lactic-co-glycolic) acid (PLGA) nanovaccine, while modulating the tumor-associated macrophages (TAM) function within the tumor microenvironment (TME) and blocking the anti-programmed cell death protein 1 (PD-1) can constitute an alternative approach for cancer immunotherapy. POx-Mannose (Man) nanovaccines generate antigen-specific T-cell responses that control tumor growth to a higher extent than poly(ethylene glycol) (PEG)-Man nanovaccines. This anti-tumor effect induced by the POx-Man nanovaccines is mediated by a CD8+ -T cell-dependent mechanism, in contrast to the PEG-Man nanovaccines. POx-Man nanovaccine combines with pexidartinib, a modulator of the TAM function, restricts the MC38 tumor growth, and synergizes with PD-1 blockade, controlling MC38 and CT26 tumor growth and survival. This data is further validated in the highly aggressive and poorly immunogenic B16F10 melanoma mouse model. Therefore, the synergistic anti-tumor effect induced by the combination of nanovaccines with the inhibition of both TAM- and PD-1-inducing immunosuppression, holds great potential for improving immunotherapy outcomes in solid cancer patients.
Collapse
Affiliation(s)
- Ana I. Matos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Carina Peres
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Barbara Carreira
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liane I. F. Moura
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Rita C. Acúrcio
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Theresa Vogel
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Erik Wegener
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Marta B. Afonso
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Fábio M. F. Santos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Águeda Martínez‐Barriocanal
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Ana S. Viana
- Centro de Química EstruturalDepartamento de Química e BioquímicaInstitute of Molecular SciencesFaculty of SciencesUniversidade de LisboaLisbon1749‐016Portugal
| | - Pedro M. P. Góis
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liana C. Silva
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Cecília M. P. Rodrigues
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Luis Graca
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Rainer Jordan
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineSagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Helena F. Florindo
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| |
Collapse
|
4
|
Zou Y, Ghaderpour A, Munkhbileg B, Seo SU, Seong SY. Taurodeoxycholate ameliorates DSS-induced colitis in mice. Int Immunopharmacol 2023; 122:110628. [PMID: 37454634 DOI: 10.1016/j.intimp.2023.110628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is typically managed using medications such as 5-aminosalicylic acid (5-ASA), glucocorticoids, anti-TNFα Ab, or anti-IL-12/23 Ab. However, some patients do not respond well to these treatments or frequently experience relapses. Therefore, alternative therapeutic options are needed. Since the activation of the inflammasome is crucial to the pathogenesis of IBD, inhibiting the inflammasome may be beneficial for patients. MATERIALS AND METHODS We tested the efficacy of taurodeoxycholate (TDCA), which is a known G-protein coupled receptor 19 (GPCR19) agonist, in a mouse colitis model induced by dextran sodium sulfate (DSS). RESULTS In the mouse colitis model, TDCA prevented loss of body weight, shortening of the colon, production of pro-inflammatory cytokines, infiltration of pro-inflammatory cells, and mucosal ulceration in the colon. In vitro, TDCA inhibited the activation of NF-κB in bone marrow-derived macrophages (BMDMs) by activating the cAMP-PKA axis. TDCA downregulated the expression of purinergic receptor P2X7 (P2X7R) and enhanced the colocalization of P2X7R with GPCR19, and inhibited the Ca2+ mobilization of BMDMs when stimulated with ATP or BzATP, which plays a pivotal role in activating the NLRP3 inflammasome (N3I) via P2X7R. TDCA inhibited the oligomerization of NLRP3-ASC and downregulated the expression of NLRP3 and ASC, as well as suppressed the maturation of pro-caspase-1 and pro-IL-1β. TDCA also increased the percentage of M2 macrophages while decreasing the number of M1 macrophages, Th1, Th2, and Th17 cells in the colon. CONCLUSION TDCA ameliorated DSS-induced colitis in mice, possibly by inhibiting both the priming phase (via the GPCR19-cAMP-PKA-NF-κB axis) and the activation phase (via the GPCR19-P2X7R-NLRP3-Caspase 1-IL-1β axis) of N3I signaling.
Collapse
Affiliation(s)
- Yunyun Zou
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Aziz Ghaderpour
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bolormaa Munkhbileg
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Yong Seong
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea; Shaperon Inc., Seoul, Republic of Korea.
| |
Collapse
|
5
|
He JJ, Li QQ, Zhao C, Zhou J, Wu J, Zhang HB, Zhao YQ, Zhang HH, Lei TY, Zhao XY, You Z, Song QB, Xu B. Advancement and Applications of Nanotherapy for Cancer Immune Microenvironment. Curr Med Sci 2023; 43:631-646. [PMID: 37558863 DOI: 10.1007/s11596-023-2763-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/27/2023] [Indexed: 08/11/2023]
Abstract
Cancer treatment has evolved rapidly due to major advances in tumor immunity research. However, due to the complexity, heterogeneity, and immunosuppressive microenvironment of tumors, the overall efficacy of immunotherapy is only 20%. In recent years, nanoparticles have attracted more attention in the field of cancer immunotherapy because of their remarkable advantages in biocompatibility, precise targeting, and controlled drug delivery. However, the clinical application of nanomedicine also faces many problems concerning biological safety, and the synergistic mechanism of nano-drugs with immunity remains to be elucidated. Our study summarizes the functional characteristics and regulatory mechanisms of nanoparticles in the cancer immune microenvironment and how nanoparticles activate and long-term stimulate innate immunity and adaptive immunity. Finally, the current problems and future development trends regarding the application of nanoparticles are fully discussed and prospected to promote the transformation and application of nanomedicine used in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ju He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qing-Qing Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Zhou
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hui-Bo Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ya-Qi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hao-Han Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tian-Yu Lei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin-Yi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zuo You
- Department of Traditional Chinese Medicine, Xianfeng County People's Hospital, Enshi, 445000, China
| | - Qi-Bin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
6
|
Pusta A, Tertis M, Crăciunescu I, Turcu R, Mirel S, Cristea C. Recent Advances in the Development of Drug Delivery Applications of Magnetic Nanomaterials. Pharmaceutics 2023; 15:1872. [PMID: 37514058 PMCID: PMC10383769 DOI: 10.3390/pharmaceutics15071872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
With the predicted rise in the incidence of cancer, there is an ever-growing need for new cancer treatment strategies. Recently, magnetic nanoparticles have stood out as promising nanostructures for imaging and drug delivery systems as they possess unique properties. Moreover, magnetic nanomaterials functionalized with other compounds can lead to multicomponent nanoparticles with innovative structures and synergetic performance. The incorporation of chemotherapeutic drugs or RNA in magnetic drug delivery systems represents a promising alternative that can increase efficiency and reduce the side effects of anticancer therapy. This review presents a critical overview of the recent literature concerning the advancements in the field of magnetic nanoparticles used in drug delivery, with a focus on their classification, characteristics, synthesis and functionalization methods, limitations, and examples of magnetic drug delivery systems incorporating chemotherapeutics or RNA.
Collapse
Affiliation(s)
- Alexandra Pusta
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
- Department of Medical Devices, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Izabell Crăciunescu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Rodica Turcu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Simona Mirel
- Department of Medical Devices, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry and Instrumental Analysis, Iuliu Hațieganu University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| |
Collapse
|
7
|
Koncz G, Jenei V, Tóth M, Váradi E, Kardos B, Bácsi A, Mázló A. Damage-mediated macrophage polarization in sterile inflammation. Front Immunol 2023; 14:1169560. [PMID: 37465676 PMCID: PMC10351389 DOI: 10.3389/fimmu.2023.1169560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/07/2023] [Indexed: 07/20/2023] Open
Abstract
Most of the leading causes of death, such as cardiovascular diseases, cancer, dementia, neurodegenerative diseases, and many more, are associated with sterile inflammation, either as a cause or a consequence of these conditions. The ability to control the progression of inflammation toward tissue resolution before it becomes chronic holds significant clinical potential. During sterile inflammation, the initiation of inflammation occurs through damage-associated molecular patterns (DAMPs) in the absence of pathogen-associated molecules. Macrophages, which are primarily localized in the tissue, play a pivotal role in sensing DAMPs. Furthermore, macrophages can also detect and respond to resolution-associated molecular patterns (RAMPs) and specific pro-resolving mediators (SPMs) during sterile inflammation. Macrophages, being highly adaptable cells, are particularly influenced by changes in the microenvironment. In response to the tissue environment, monocytes, pro-inflammatory macrophages, and pro-resolution macrophages can modulate their differentiation state. Ultimately, DAMP and RAMP-primed macrophages, depending on the predominant subpopulation, regulate the balance between inflammatory and resolving processes. While sterile injury and pathogen-induced reactions may have distinct effects on macrophages, most studies have focused on macrophage responses induced by pathogens. In this review, which emphasizes available human data, we illustrate how macrophages sense these mediators by examining the expression of receptors for DAMPs, RAMPs, and SPMs. We also delve into the signaling pathways induced by DAMPs, RAMPs, and SPMs, which primarily contribute to the regulation of macrophage differentiation from a pro-inflammatory to a pro-resolution phenotype. Understanding the regulatory mechanisms behind the transition between macrophage subtypes can offer insights into manipulating the transition from inflammation to resolution in sterile inflammatory diseases.
Collapse
Affiliation(s)
- Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Márta Tóth
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eszter Váradi
- Institute of Genetics, Biological Research Centre, Eotvos Lorand Research Network, Szeged, Hungary
- Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Balázs Kardos
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Allergology Research Group, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
8
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. Adv Sci (Weinh) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
9
|
Su X, Gao Y, Yang R. Gut microbiota derived bile acid metabolites maintain the homeostasis of gut and systemic immunity. Front Immunol 2023; 14:1127743. [PMID: 37256134 PMCID: PMC10225537 DOI: 10.3389/fimmu.2023.1127743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 06/01/2023] Open
Abstract
Bile acids (BAs) as cholesterol-derived molecules play an essential role in some physiological processes such as nutrient absorption, glucose homeostasis and regulation of energy expenditure. They are synthesized in the liver as primary BAs such as cholic acid (CA), chenodeoxycholic acid (CDCA) and conjugated forms. A variety of secondary BAs such as deoxycholic acid (DCA) and lithocholic acid (LCA) and their derivatives is synthesized in the intestine through the involvement of various microorganisms. In addition to essential physiological functions, BAs and their metabolites are also involved in the differentiation and functions of innate and adaptive immune cells such as macrophages (Macs), dendritic cells (DCs), myeloid derived suppressive cells (MDSCs), regulatory T cells (Treg), Breg cells, T helper (Th)17 cells, CD4 Th1 and Th2 cells, CD8 cells, B cells and NKT cells. Dysregulation of the BAs and their metabolites also affects development of some diseases such as inflammatory bowel diseases. We here summarize recent advances in how BAs and their metabolites maintain gut and systemic homeostasis, including the metabolism of the BAs and their derivatives, the role of BAs and their metabolites in the differentiation and function of immune cells, and the effects of BAs and their metabolites on immune-associated disorders.
Collapse
Affiliation(s)
- Xiaomin Su
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
10
|
Luo Y, Vivaldi Marrero E, Choudhary V, Bollag WB. Phosphatidylglycerol to Treat Chronic Skin Wounds in Diabetes. Pharmaceutics 2023; 15:pharmaceutics15051497. [PMID: 37242739 DOI: 10.3390/pharmaceutics15051497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
This review proposes the use of dioleoylphosphatidylglycerol (DOPG) to enhance diabetic wound healing. Initially, the characteristics of diabetic wounds are examined, focusing on the epidermis. Hyperglycemia accompanying diabetes results in enhanced inflammation and oxidative stress in part through the generation of advanced glycation end-products (AGEs), in which glucose is conjugated to macromolecules. These AGEs activate inflammatory pathways; oxidative stress results from increased reactive oxygen species generation by mitochondria rendered dysfunctional by hyperglycemia. These factors work together to reduce the ability of keratinocytes to restore epidermal integrity, contributing to chronic diabetic wounds. DOPG has a pro-proliferative action on keratinocytes (through an unclear mechanism) and exerts an anti-inflammatory effect on keratinocytes and the innate immune system by inhibiting the activation of Toll-like receptors. DOPG has also been found to enhance macrophage mitochondrial function. Since these DOPG effects would be expected to counteract the increased oxidative stress (attributable in part to mitochondrial dysfunction), decreased keratinocyte proliferation, and enhanced inflammation that characterize chronic diabetic wounds, DOPG may be useful in stimulating wound healing. To date, efficacious therapies to promote the healing of chronic diabetic wounds are largely lacking; thus, DOPG may be added to the armamentarium of drugs to enhance diabetic wound healing.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Edymarie Vivaldi Marrero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Vivek Choudhary
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA
| | - Wendy B Bollag
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA
- Department of Dermatology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
11
|
Zhen W, Weichselbaum RR, Lin W. Nanoparticle-Mediated Radiotherapy Remodels the Tumor Microenvironment to Enhance Antitumor Efficacy. Adv Mater 2023; 35:e2206370. [PMID: 36524978 PMCID: PMC10213153 DOI: 10.1002/adma.202206370] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/12/2022] [Indexed: 05/26/2023]
Abstract
Radiotherapy (RT) uses ionizing radiation to eradicate localized tumors and, in rare cases, control tumors outside of the irradiated fields via stimulating an antitumor immune response (abscopal effect). However, the therapeutic effect of RT is often limited by inherent physiological barriers of the tumor microenvironment (TME), such as hypoxia, abnormal vasculature, dense extracellular matrix (ECM), and an immunosuppressive TME. Thus, it is critical to develop new RT strategies that can remodel the TME to overcome radio-resistance and immune suppression. In the past decade, high-Z-element nanoparticles have been developed to increase radiotherapeutic indices of localized tumors by reducing X-ray doses and side effects to normal tissues and enhance abscopal effects by activating the TME to elicit systemic antitumor immunity. In this review, the principles of RT and radiosensitization, the mechanisms of radio-resistance and immune suppression, and the use of various nanoparticles to sensitize RT and remodel TMEs for enhanced antitumor efficacy are discussed. The challenges in clinical translation of multifunctional TME-remodeling nanoradiosensitizers are also highlighted.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
12
|
Lv Y, Pu R, Tao Y, Yang X, Mu H, Wang H, Sun W. Applications and Future Prospects of Micro/Nanorobots Utilizing Diverse Biological Carriers. Micromachines (Basel) 2023; 14:mi14050983. [PMID: 37241607 DOI: 10.3390/mi14050983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023]
Abstract
Targeted drug delivery using micro-nano robots (MNRs) is a rapidly advancing and promising field in biomedical research. MNRs enable precise delivery of drugs, addressing a wide range of healthcare needs. However, the application of MNRs in vivo is limited by power issues and specificity in different scenarios. Additionally, the controllability and biological safety of MNRs must be considered. To overcome these challenges, researchers have developed bio-hybrid micro-nano motors that offer improved accuracy, effectiveness, and safety for targeted therapies. These bio-hybrid micro-nano motors/robots (BMNRs) use a variety of biological carriers, blending the benefits of artificial materials with the unique features of different biological carriers to create tailored functions for specific needs. This review aims to give an overview of the current progress and application of MNRs with various biocarriers, while exploring the characteristics, advantages, and potential hurdles for future development of these bio-carrier MNRs.
Collapse
Affiliation(s)
- Yu Lv
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ruochen Pu
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yining Tao
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiyu Yang
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hongsheng Wang
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wei Sun
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
13
|
Perez-Potti A, Rodríguez-Pérez M, Polo E, Pelaz B, Del Pino P. Nanoparticle-based immunotherapeutics: from the properties of nanocores to the differential effects of administration routes. Adv Drug Deliv Rev 2023; 197:114829. [PMID: 37121275 DOI: 10.1016/j.addr.2023.114829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
The engagement with the immune system is one of the main cornerstones in the development of nanotechnologies for therapy and diagnostics. Recent advances have made possible the tuning of features like size, shape and biomolecular modifications that influence such interactions, however, the capabilities for immune modulation of nanoparticles are still not well defined and exploited. This review focuses on recent advances made in preclinical research for the application of nanoparticles to modulate immune responses, and the main features making them relevant for such applications. We review and discuss newest evidence in the field, which include in vivo experiments with an extensive physicochemical characterization as well as detailed study of the induced immune response. We emphasize the need of incorporating knowledge about immune response development and regulation in the design and application of nanoparticles, including the effect by parameters such as the administration route and the differential interactions with immune subsets.
Collapse
Affiliation(s)
- André Perez-Potti
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Pérez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
14
|
Dietz-Fricke C, Tacke F, Zöllner C, Demir M, Schmidt HH, Schramm C, Willuweit K, Lange CM, Weber S, Denk G, Berg CP, Grottenthaler JM, Merle U, Olkus A, Zeuzem S, Sprinzl K, Berg T, van Bömmel F, Wiegand J, Herta T, Seufferlein T, Zizer E, Dikopoulos N, Thimme R, Neumann-Haefelin C, Galle PR, Sprinzl M, Lohse AW, Schulze zur Wiesch J, Kempski J, Geier A, Reiter FP, Schlevogt B, Gödiker J, Hofmann WP, Buggisch P, Kahlhöfer J, Port K, Maasoumy B, Cornberg M, Wedemeyer H, Deterding K. Treating hepatitis D with bulevirtide - Real-world experience from 114 patients. JHEP Rep 2023; 5:100686. [PMID: 37025462 PMCID: PMC10071092 DOI: 10.1016/j.jhepr.2023.100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 04/08/2023] Open
Abstract
Background & Aims Bulevirtide is a first-in-class entry inhibitor of hepatitis B surface antigen. In July 2020, bulevirtide was conditionally approved for the treatment of hepatitis D, the most severe form of viral hepatitis, which frequently causes end-stage liver disease and hepatocellular carcinoma. Herein, we report the first data from a large multicenter real-world cohort of patients with hepatitis D treated with bulevirtide at a daily dose of 2 mg without additional interferon. Methods In a joint effort with 16 hepatological centers, we collected anonymized retrospective data from patients treated with bulevirtide for chronic hepatitis D. Results Our analysis is based on data from 114 patients, including 59 (52%) with cirrhosis, receiving a total of 4,289 weeks of bulevirtide treatment. A virologic response defined as an HDV RNA decline of at least 2 log or undetectable HDV RNA was observed in 87/114 (76%) cases with a mean time to virologic response of 23 weeks. In 11 cases, a virologic breakthrough (>1 log-increase in HDV RNA after virologic response) was observed. After 24 weeks of treatment, 19/33 patients (58%) had a virologic response, while three patients (9%) did not achieve a 1 log HDV RNA decline. No patient lost hepatitis B surface antigen. Alanine aminotransferase levels improved even in patients not achieving a virologic response, including five patients who had decompensated cirrhosis at the start of treatment. Treatment was well tolerated and there were no reports of drug-related serious adverse events. Conclusions In conclusion, we confirm the safety and efficacy of bulevirtide monotherapy in a large real-world cohort of patients with hepatitis D treated in Germany. Future studies need to explore the long-term benefits and optimal duration of bulevirtide treatment. Impact and implications Clinical trials proved the efficacy of bulevirtide for chronic hepatitis D and led to conditional approval by the European Medical Agency. Now it is of great interest to investigate the effects of bulevirtide treatment in a real-world setting. In this work, we included data from 114 patients with chronic hepatitis D who were treated with bulevirtide at 16 German centers. A virologic response was seen in 87/114 cases. After 24 weeks of treatment, only a small proportion of patients did not respond to treatment. At the same time, signs of liver inflammation improved. This observation was independent from changes in hepatitis D viral load. The treatment was generally well tolerated. In the future, it will be of interest to investigate the long-term effects of this new treatment.
Collapse
Affiliation(s)
- Christopher Dietz-Fricke
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Caroline Zöllner
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Hartmut H. Schmidt
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Christoph Schramm
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Katharina Willuweit
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Christian M. Lange
- Department of Medicine II, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Sabine Weber
- Department of Medicine II, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Gerald Denk
- Department of Medicine II, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Christoph P. Berg
- Department of Gastroenterology, Gastrointestinal Oncology, Hepatology, Infectiology, and Geriatrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia M. Grottenthaler
- Department of Gastroenterology, Gastrointestinal Oncology, Hepatology, Infectiology, and Geriatrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University of Heidelberg, Heidelberg, Germany
| | - Alexander Olkus
- Department of Internal Medicine IV, University of Heidelberg, Heidelberg, Germany
| | - Stefan Zeuzem
- Internal Medicine Department, Goethe University Hospital, Frankfurt, Germany
| | - Kathrin Sprinzl
- Internal Medicine Department, Goethe University Hospital, Frankfurt, Germany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Florian van Bömmel
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Johannes Wiegand
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Toni Herta
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | | | - Eugen Zizer
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | | | - Robert Thimme
- Department of Medicine II, University Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Neumann-Haefelin
- Department of Medicine II, University Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter R. Galle
- Department of Medicine I, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Martin Sprinzl
- Department of Medicine I, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Ansgar W. Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze zur Wiesch
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg - Lübeck - Borstel - Riems, Hamburg, Germany
| | - Jan Kempski
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Geier
- University Hospital Würzburg, Division of Hepatology, Dept. of Medicine II, Würzburg, Germany
| | - Florian P. Reiter
- University Hospital Würzburg, Division of Hepatology, Dept. of Medicine II, Würzburg, Germany
| | | | - Juliana Gödiker
- Department of Medicine B, University Hospital Münster, Münster, Germany
| | | | - Peter Buggisch
- Ifi-Institute for Interdisciplinary Medicine, Hamburg, Germany
| | - Julia Kahlhöfer
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
| | - Kerstin Port
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
| | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
- D-SOLVE consortium, a EU Horizon Europe funded project (No 101057917)
- Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
- Excellence Cluster Resist, Hannover Medical School, Germany
- German Centre for Infection Research (DZIF), Hannover-Braunschweig, Germany
- D-SOLVE consortium, a EU Horizon Europe funded project (No 101057917)
| | - Katja Deterding
- Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany
- Corresponding author. Address: Department of Gastroenterology, Hepatology and Endocrinology at Hannover Medical School, Hannover, Germany.
| |
Collapse
|
15
|
Manjili MH. The adaptation model of immunity: A new insight into aetiology and treatment of multiple sclerosis. Scand J Immunol 2023; 97:e13255. [PMID: 36680379 DOI: 10.1111/sji.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/04/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Current research and drug development for multiple sclerosis (MS) is fully influenced by the self-nonself (SNS) model of immunity, suggesting that breakage of immunological tolerance towards self-antigens expressed in the central nervous system (CNS) is responsible for pathogenesis of MS; thus, immune suppressive drugs are recommended for the management of the disease. However, this model provides incomplete understanding of the causes and pathways involved in the onset and progression of MS and limits our ability to effectively treat this neurological disease. Some pre-clinical and clinical reports have been misunderstood; some others have been underappreciated because of the lack of a theoretical model that can explain them. Also, current immunotherapies are guided according to the models that are not designed to explain the functional outcomes of an immune response. The adaptation model of immunity is proposed to offer a new understanding of the existing data and galvanize a new direction for the treatment of MS. According to this model, the immune system continuously communicates with the CNS through the adaptation receptors (AdRs) and adaptation ligands (AdLs) or co-receptors, signal IV, to support cell growth and neuroplasticity. Alterations in the expression of the neuronal AdRs results in MS by shifting the cerebral inflammatory immune responses from remyelination to demyelination. Therefore, novel therapeutics for MS should be focused on the discovery and targeting of the AdR/AdL axis in the CNS rather than carrying on with immune suppressive interventions.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Virginia, USA
| |
Collapse
|
16
|
He Y, Li Y, Pan Y, Li A, Huang Y, Mi Q, Zhao S, Zhang C, Ran J, Hu H, Pan H. Correlation analysis between jejunum metabolites and immune function in Saba and Landrace piglets. Front Vet Sci 2023; 10:1069809. [PMID: 37008364 PMCID: PMC10060822 DOI: 10.3389/fvets.2023.1069809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
The immune function of the intestinal mucosa plays a crucial role in the intestinal health of hosts. As signaling molecules and precursors of metabolic reactions, intestinal chyme metabolites are instrumental in maintaining host immune homeostasis. Saba (SB) pigs, a unique local pig species in central Yunnan Province, China. However, research on jejunal metabolites in this species is limited. Here, we used immunohistochemistry and untargeted metabolomics by liquid chromatography mass spectrometry (LC-MS/MS) to study differences in jejunal immunophenotypes and metabolites between six Landrace (LA) and six SB piglets (35 days old). The results showed that the levels of the anti-inflammatory factor interleukin 10 (IL-10) were markedly higher in SB piglets than in LA piglets (P < 0.01), while the levels of the proinflammatory factors IL-6, IL-1β, and Toll-like receptor 2 (TLR-2) were markedly lower (P < 0.01). Furthermore, the levels of mucin 2 (MUC2) and zona occludens (ZO-1), which are related to mucosal barrier function, were significantly higher in SB piglets than in LA piglets (P < 0.01), as were villus height, villus height/crypt depth ratio, and goblet cell number (P < 0.05). Differences in jejunal chyme metabolic patterns were observed between the two piglets. In the negative ion mode, cholic acid metabolites ranked in the top 20 and represented 25% of the total. Taurodeoxycholic acid (TDCA) content was significantly higher in SB piglets than in LA piglets (P < 0.01). TDCA positively correlated with ZO-1, villus height, villus height/crypt depth ratio, and goblet cell number. These results suggest that SB pigs have a strong jejunal immune function and that TDCA was positively regulates jejunal immunity and mucosal barrier function. Our findings provide a reference for understanding intestinal immune function in different pig breeds and for the discovery of potential biomarkers to help solve health issues related to pig production.
Collapse
Affiliation(s)
- Yang He
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yongxiang Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | |
|