1
|
Zhang Y, Li Y, Xu Z, Xu L, Wang Y, Li N, Solek NC, Wang Y, Li B, Liu H. PPS-TLR7/8 agonist nanoparticles equip robust anticancer immunity by selectively prolonged activation of dendritic cells. Biomaterials 2025; 316:123032. [PMID: 39705927 DOI: 10.1016/j.biomaterials.2024.123032] [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: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Checkpoint inhibitor therapies do not benefit all patients, and adjuvants play a critical role in boosting immune responses for effective cancer immunotherapy. However, their systemic toxicity and suboptimal activation kinetics pose significant challenges. Here, this study presented a linker-based strategy to modulate the activation kinetics of Toll-like receptor 7/8 (TLR7/8) agonists delivered via poly (propylene sulfide) nanoparticles (PPS NPs). By covalently binding small molecule TLR7/8 agonists to PPS NPs with different linkers, enhanced therapeutic efficacy is achieved while abrogating systemic toxicity. These results showed that an alkyl linker selectively prolong the activation of DCs. It avoided the extensive activation of other APCs, favoring the limitation of immune-related toxicities. This strategy exhibited significant anti-tumor activity in alkyl linked nano-TLR7/8 agonists treatment alone, and cytokine and immune cell profiling provided evidence of prolonged immune cell activation in the tumor microenvironment, with evidence of an increase in the frequency of tumor antigen-specific CD8+ T cells. This linker-based approach offers a promising strategy to optimize the delivery of nano-TLR7/8 agonists for cancer immunotherapy, potentially advancing the field toward improved clinical outcomes.
Collapse
Affiliation(s)
- Yingxi Zhang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yicheng Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Zhaochu Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Linyi Xu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Yue Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Ning Li
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China
| | - Nicholas C Solek
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Yongjun Wang
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C1, Canada.
| | - Hongzhuo Liu
- Wuya college of innovation, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
2
|
Avitan O, Rafael T, Vreeburg M, Elst L, Bekers EM, Albersen M, Jordanova ES, Brouwer O. Penile intraepithelial neoplasia incidence, clinical classification, microenvironment and implications for imiquimod treatment. BJU Int 2024; 134:881-889. [PMID: 39030899 DOI: 10.1111/bju.16473] [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] [Indexed: 07/22/2024]
Abstract
OBJECTIVES To provide an outline of the existing data on penile intraepithelial neoplasia (PeIN), as well as a narrative review on imiquimod (IQ; a toll-like receptor 7 agonist) treatment and immune microenvironment markers that may predict response to treatment. METHODS A narrative review of the literature from 2000 to the present was conducted on PubMed, and we describe the most relevant data and cross references. RESULTS The incidence of PeIN is increasing. Local therapy with IQ may offer an easy applicable treatment with complete response rates of up to 63% but can be associated with considerable side-effects. There is no conclusive data on the optimal treatment schedule for PeIN, but evaluation of treatment results for other human papillomavirus-related pre-malignancies suggest three times a week for a duration up to 16 weeks. There are no published studies concerning the PeIN immune microenvironment. However, findings from the few studies on penile cancer and pre-cancerous vulvar and cervical lesions imply that specific immune cell subpopulations can serve as future predictors for successful immunomodulation treatments such as IQ. CONCLUSIONS Overall, limited data are available on IQ treatment for PeIN and no published data exists on the PeIN immune microenvironment. Further translational studies are warranted to gain more understanding on the pathophysiology of PeIN and potential predictors of progression and of response to topical treatments.
Collapse
Affiliation(s)
- Ofir Avitan
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Tynisha Rafael
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Manon Vreeburg
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Laura Elst
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Elise M Bekers
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Ekaterina S Jordanova
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Oscar Brouwer
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Chen O, Jiang C, Berta T, Gray B, Furutani K, Sullenger BA, Ji RR. MicroRNA let-7b enhances spinal cord nociceptive synaptic transmission and induces acute and persistent pain through neuronal and microglial signaling. Pain 2024; 165:1824-1839. [PMID: 38452223 PMCID: PMC11257826 DOI: 10.1097/j.pain.0000000000003206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024]
Abstract
ABSTRACT Secreted microRNAs (miRNAs) have been detected in various body fluids including the cerebrospinal fluid, yet their direct role in regulating synaptic transmission remains uncertain. We found that intrathecal injection of low dose of let-7b (1 μg) induced short-term (<24 hours) mechanical allodynia and heat hyperalgesia, a response that is compromised in Tlr7-/- or Trpa1-/- mice. Ex vivo and in vivo calcium imaging in GCaMP6-report mice revealed increased calcium signal in spinal cord afferent terminals and doral root ganglion/dorsal root ganglia neurons following spinal perfusion and intraplantar injection of let-7b. Patch-clamp recordings also demonstrated enhanced excitatory synaptic transmission (miniature excitatory postsynaptic currents [EPSCs]) in spinal nociceptive neurons following let-7b perfusion or optogenetic activation of axonal terminals. The elevation in spinal calcium signaling and EPSCs was dependent on the presence of toll-like receptor-7 (TLR7) and transient receptor potential ion channel subtype A1 (TRPA1). In addition, endogenous let-7b is enriched in spinal cord synaptosome, and peripheral inflammation increased let-7b in doral root ganglion/dorsal root ganglia neurons, spinal cord tissue, and the cerebrospinal fluid. Notably, let-7b antagomir inhibited inflammatory pain and inflammation-induced synaptic plasticity (EPSC increase), suggesting an endogenous role of let-7b in regulating pain and synaptic transmission. Furthermore, intrathecal injection of let-7b, at a higher dose (10 μg), induced persistent mechanical allodynia for >2 weeks, which was abolished in Tlr7-/- mice. The high dose of let-7b also induced microgliosis in the spinal cord. Of interest, intrathecal minocycline only inhibited let-7b-induced mechanical allodynia in male but not female mice. Our findings indicate that the secreted microRNA let-7b has the capacity to provoke pain through both neuronal and glial signaling, thereby establishing miRNA as an emerging neuromodulator.
Collapse
Affiliation(s)
- Ouyang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Temugin Berta
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, OH 45267, USA
| | - Bethany Gray
- Department of Surgery, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Kenta Furutani
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Bruce A. Sullenger
- Department of Surgery, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| |
Collapse
|
4
|
Story ME, Ferris LK, Mathers AR. Resident memory T cells in dirty mice suppress innate cell activation and infiltration into the skin following stimulation with alarmins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602963. [PMID: 39071349 PMCID: PMC11275811 DOI: 10.1101/2024.07.11.602963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Trm cells are sequestered at barrier tissues as a swift first line defense against peripheral reinfections in both antigen dependent and antigen independent bystander modes. Trm cells are also capable of mediating autoimmune diseases, such as psoriasis, wherein autoreactive Trm cells are aberrantly activated. To quickly combat infections, activated Trm cells can stimulate the influx and activation of memory T cells and innate immune cells. However, there is significant heterogeneity in the inflammatory responses that Trm cell populations can induce, specifically in the activation of the innate profile. Most studies to date have utilized a reductionist approach to examine single Trm populations, specific pathogens, and defined tissues. Herein, we adopted a more holistic approach utilizing barrier-free 'dirty' mice to profile activated innate cells attracted to the skin in the presence of quiescent cutaneous Trm cells. Notably, dirty mice are a more human predictive model due to having a diverse microbial experience that leads to the development of a complete complement of Trm cells in the skin. We demonstrate that in the dirty mouse model mice have a significant reduction in cutaneous neutrophils and monocytes compared to SPF mice following local treatment with two separate innate stimuli. These findings reveal that cutaneous Trm cells have the capacity to temper the innate immune response and further substantiate the implication that Trm cells are heterogenous in their functions depending in large part on their tissue residency. However, in an autoimmune microenvironment Trm cells are capable of recruiting innate cells to the site of an exposure to a damage-associated molecular pattern. Likely due to the imbalance of IL-17 and IFN-γ.
Collapse
Affiliation(s)
- Meaghan E. Story
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Laura K. Ferris
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alicia R. Mathers
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
5
|
Allard RL, Mayfield J, Barchiesi R, Salem NA, Mayfield RD. Toll-like receptor 7: A novel neuroimmune target to reduce excessive alcohol consumption. Neurobiol Stress 2024; 31:100639. [PMID: 38765062 PMCID: PMC11101708 DOI: 10.1016/j.ynstr.2024.100639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Toll-like receptors (TLRs) are a family of innate immune receptors that recognize molecular patterns in foreign pathogens and intrinsic danger/damage signals from cells. TLR7 is a nucleic acid sensing endosomal TLR that is activated by single-stranded RNAs from microbes or by small noncoding RNAs that act as endogenous ligands. TLR7 signals through the MyD88 adaptor protein and activates the transcription factor interferon regulatory factor 7 (IRF7). TLR7 is found throughout the brain and is highly expressed in microglia, the main immune cells of the brain that have also been implicated in alcohol drinking in mice. Upregulation of TLR7 mRNA and protein has been identified in postmortem hippocampus and cortex from AUD subjects that correlated positively with lifetime consumption of alcohol. Similarly, Tlr7 and downstream signaling genes were upregulated in rat hippocampal and cortical slice cultures after chronic alcohol exposure and in these regions after chronic binge-like alcohol treatment in mice. In addition, repeated administration of the synthetic TLR7 agonists imiquimod (R837) or resiquimod (R848) increased voluntary alcohol drinking in different rodent models and produced sustained upregulation of IRF7 in the brain. These findings suggest that chronic TLR7 activation may drive excessive alcohol drinking. In the brain, this could occur through increased levels of endogenous TLR7 activators, like microRNAs and Y RNAs. This review explores chronic TLR7 activation as a pathway of dysregulated neuroimmune signaling in AUD and the endogenous small RNA ligands in the brain that could perpetuate innate immune responses and escalate alcohol drinking.
Collapse
Affiliation(s)
- Ruth L. Allard
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Riccardo Barchiesi
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nihal A. Salem
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
6
|
Aw E, Zhang Y, Yalcin E, Herrmann U, Lin SL, Langston K, Castrillon C, Ma M, Moffitt JR, Carroll MC. Spatial enrichment of the type 1 interferon signature in the brain of a neuropsychiatric lupus murine model. Brain Behav Immun 2023; 114:511-522. [PMID: 37369340 PMCID: PMC10918751 DOI: 10.1016/j.bbi.2023.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/01/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023] Open
Abstract
Among systemic lupus erythematosus (SLE) patients, neuropsychiatric symptoms are highly prevalent, being observed in up to 80% of adult and 95% of pediatric patients. Type 1 interferons, particularly interferon alpha (IFNα), have been implicated in the pathogenesis of SLE and its associated neuropsychiatric symptoms (NPSLE). However, it remains unclear how type 1 interferon signaling in the central nervous system (CNS) might result in neuropsychiatric sequelae. In this study, we validate an NPSLE mouse model and find an elevated peripheral type 1 interferon signature alongside clinically relevant NPSLE symptoms such as anxiety and fatigue. Unbiased single-nucleus sequencing of the hindbrain and hippocampus revealed that interferon-stimulated genes (ISGs) were among the most highly upregulated genes in both regions and that gene pathways involved in cellular interaction and neuronal development were generally repressed among astrocytes, oligodendrocytes, and neurons. Using image-based spatial transcriptomics, we found that the type 1 interferon signature is enriched as spatially distinct patches within the brain parenchyma of these mice. Our results suggest that type 1 interferon in the CNS may play an important mechanistic role in mediating NPSLE behavioral phenotypes by repressing general cellular communication pathways, and that type 1 interferon signaling modulators are a potential therapeutic option for NPSLE.
Collapse
Affiliation(s)
- Ernest Aw
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Division of Medical Sciences, Harvard Medical School, Boston, MA, United States
| | - Yingying Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Esra Yalcin
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Uli Herrmann
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Stacie L Lin
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Division of Medical Sciences, Harvard Medical School, Boston, MA, United States
| | - Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Carlos Castrillon
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Minghe Ma
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
7
|
Aw E, Lin SL, Zhang Y, Herrmann U, Yalcin E, Langston K, Castrillion C, Ma M, Moffitt JR, Carroll MC. [WITHDRAWN] Spatial enrichment of the type 1 interferon signature in the brain of a neuropsychiatric lupus murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537814. [PMID: 37131759 PMCID: PMC10153248 DOI: 10.1101/2023.04.21.537814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
bioRxiv has withdrawn this manuscript because it was posted without the consent of all authors. If you have any questions, please contact the corresponding author.
Collapse
|
8
|
Hawtin S, André C, Collignon-Zipfel G, Appenzeller S, Bannert B, Baumgartner L, Beck D, Betschart C, Boulay T, Brunner HI, Ceci M, Deane J, Feifel R, Ferrero E, Kyburz D, Lafossas F, Loetscher P, Merz-Stoeckle C, Michellys P, Nuesslein-Hildesheim B, Raulf F, Rush JS, Ruzzante G, Stein T, Zaharevitz S, Wieczorek G, Siegel R, Gergely P, Shisha T, Junt T. Preclinical characterization of the Toll-like receptor 7/8 antagonist MHV370 for lupus therapy. Cell Rep Med 2023; 4:101036. [PMID: 37196635 DOI: 10.1016/j.xcrm.2023.101036] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/17/2022] [Accepted: 04/12/2023] [Indexed: 05/19/2023]
Abstract
Genetic and in vivo evidence suggests that aberrant recognition of RNA-containing autoantigens by Toll-like receptors (TLRs) 7 and 8 drives autoimmune diseases. Here we report on the preclinical characterization of MHV370, a selective oral TLR7/8 inhibitor. In vitro, MHV370 inhibits TLR7/8-dependent production of cytokines in human and mouse cells, notably interferon-α, a clinically validated driver of autoimmune diseases. Moreover, MHV370 abrogates B cell, plasmacytoid dendritic cell, monocyte, and neutrophil responses downstream of TLR7/8. In vivo, prophylactic or therapeutic administration of MHV370 blocks secretion of TLR7 responses, including cytokine secretion, B cell activation, and gene expression of, e.g., interferon-stimulated genes. In the NZB/W F1 mouse model of lupus, MHV370 halts disease. Unlike hydroxychloroquine, MHV370 potently blocks interferon responses triggered by specific immune complexes from systemic lupus erythematosus patient sera, suggesting differentiation from clinical standard of care. These data support advancement of MHV370 to an ongoing phase 2 clinical trial.
Collapse
Affiliation(s)
- Stuart Hawtin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Cédric André
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | | | - Simone Appenzeller
- Department of Orthopedics, Rheumatology, and Traumatology, School of Medical Science, University of Campinas (UNICAMP), Campinas, 13083-887 São Paulo, Brazil
| | - Bettina Bannert
- Department of Rheumatology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Lea Baumgartner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Damian Beck
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Claudia Betschart
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Thomas Boulay
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Hermine I Brunner
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Melanie Ceci
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Jonathan Deane
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | - Roland Feifel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Enrico Ferrero
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Diego Kyburz
- Department of Rheumatology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Frederique Lafossas
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Pius Loetscher
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | | | - Pierre Michellys
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | | | - Friedrich Raulf
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - James S Rush
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Giulia Ruzzante
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Thomas Stein
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Samantha Zaharevitz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | - Grazyna Wieczorek
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Richard Siegel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Tamas Shisha
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland.
| |
Collapse
|
9
|
Simón-Fuentes M, Herrero C, Acero-Riaguas L, Nieto C, Lasala F, Labiod N, Luczkowiak J, Alonso B, Delgado R, Colmenares M, Corbí ÁL, Domínguez-Soto Á. TLR7 Activation in M-CSF-Dependent Monocyte-Derived Human Macrophages Potentiates Inflammatory Responses and Prompts Neutrophil Recruitment. J Innate Immun 2023; 15:517-530. [PMID: 37040733 PMCID: PMC10315069 DOI: 10.1159/000530249] [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: 11/08/2022] [Accepted: 03/15/2023] [Indexed: 04/13/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is an endosomal pathogen-associated molecular pattern (PAMP) receptor that senses single-stranded RNA (ssRNA) and whose engagement results in the production of type I IFN and pro-inflammatory cytokines upon viral exposure. Recent genetic studies have established that a dysfunctional TLR7-initiated signaling is directly linked to the development of inflammatory responses. We present evidence that TLR7 is preferentially expressed by monocyte-derived macrophages generated in the presence of M-CSF (M-MØ). We now show that TLR7 activation in M-MØ triggers a weak MAPK, NFκB, and STAT1 activation and results in low production of type I IFN. Of note, TLR7 engagement reprograms MAFB+ M-MØ towards a pro-inflammatory transcriptional profile characterized by the expression of neutrophil-attracting chemokines (CXCL1-3, CXCL5, CXCL8), whose expression is dependent on the transcription factors MAFB and AhR. Moreover, TLR7-activated M-MØ display enhanced pro-inflammatory responses and a stronger production of neutrophil-attracting chemokines upon secondary stimulation. As aberrant TLR7 signaling and enhanced pulmonary neutrophil/lymphocyte ratio associate with impaired resolution of virus-induced inflammatory responses, these results suggest that targeting macrophage TLR7 might be a therapeutic strategy for viral infections where monocyte-derived macrophages exhibit a pathogenic role.
Collapse
Affiliation(s)
- Miriam Simón-Fuentes
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Cristina Herrero
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Lucia Acero-Riaguas
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Concha Nieto
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Fatima Lasala
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Nuria Labiod
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Bárbara Alonso
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Maria Colmenares
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ángel L Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | |
Collapse
|
10
|
Yang Y, Li H, Fotopoulou C, Cunnea P, Zhao X. Toll-like receptor-targeted anti-tumor therapies: Advances and challenges. Front Immunol 2022; 13:1049340. [PMID: 36479129 PMCID: PMC9721395 DOI: 10.3389/fimmu.2022.1049340] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors, originally discovered to stimulate innate immune reactions against microbial infection. TLRs also play essential roles in bridging the innate and adaptive immune system, playing multiple roles in inflammation, autoimmune diseases, and cancer. Thanks to the immune stimulatory potential of TLRs, TLR-targeted strategies in cancer treatment have proved to be able to regulate the tumor microenvironment towards tumoricidal phenotypes. Quantities of pre-clinical studies and clinical trials using TLR-targeted strategies in treating cancer have been initiated, with some drugs already becoming part of standard care. Here we review the structure, ligand, signaling pathways, and expression of TLRs; we then provide an overview of the pre-clinical studies and an updated clinical trial watch targeting each TLR in cancer treatment; and finally, we discuss the challenges and prospects of TLR-targeted therapy.
Collapse
Affiliation(s)
- Yang Yang
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Hongyi Li
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Christina Fotopoulou
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Paula Cunnea
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xia Zhao
- Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Reyes EY, Shinohara ML. Host immune responses in the central nervous system during fungal infections. Immunol Rev 2022; 311:50-74. [PMID: 35672656 PMCID: PMC9489659 DOI: 10.1111/imr.13101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 12/19/2023]
Abstract
Fungal infections in the central nervous system (CNS) cause high morbidity and mortality. The frequency of CNS mycosis has increased over the last two decades as more individuals go through immunocompromised conditions for various reasons. Nevertheless, options for clinical interventions for CNS mycoses are still limited. Thus, there is an urgent need to understand the host-pathogen interaction mechanisms in CNS mycoses for developing novel treatments. Although the CNS has been regarded as an immune-privileged site, recent studies demonstrate the critical involvement of immune responses elicited by CNS-resident and CNS-infiltrated cells during fungal infections. In this review, we discuss mechanisms of fungal invasion in the CNS, fungal pathogen detection by CNS-resident cells (microglia, astrocytes, oligodendrocytes, neurons), roles of CNS-infiltrated leukocytes, and host immune responses. We consider that understanding host immune responses in the CNS is crucial for endeavors to develop treatments for CNS mycosis.
Collapse
Affiliation(s)
- Estefany Y. Reyes
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Mari L. Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27705, USA
| |
Collapse
|
12
|
Schapovalova O, Gorlova A, de Munter J, Sheveleva E, Eropkin M, Gorbunov N, Sicker M, Umriukhin A, Lyubchyk S, Lesch KP, Strekalova T, Schroeter CA. Immunomodulatory effects of new phytotherapy on human macrophages and TLR4- and TLR7/8-mediated viral-like inflammation in mice. Front Med (Lausanne) 2022; 9:952977. [PMID: 36091684 PMCID: PMC9450044 DOI: 10.3389/fmed.2022.952977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background While all efforts have been undertaken to propagate the vaccination and develop remedies against SARS-CoV-2, no satisfactory management of this infection is available yet. Moreover, poor availability of any preventive and treatment measures of SARS-CoV-2 in economically disadvantageous communities aggravates the course of the pandemic. Here, we studied a new immunomodulatory phytotherapy (IP), an extract of blackberry, chamomile, garlic, cloves, and elderberry as a potential low-cost solution for these problems given the reported efficacy of herbal medicine during the previous SARS virus outbreak. Methods The key feature of SARS-CoV-2 infection, excessive inflammation, was studied in in vitro and in vivo assays under the application of the IP. First, changes in tumor-necrosis factor (TNF) and lnteurleukin-1 beta (IL-1β) concentrations were measured in a culture of human macrophages following the lipopolysaccharide (LPS) challenge and treatment with IP or prednisolone. Second, chronically IP-pre-treated CD-1 mice received an agonist of Toll-like receptors (TLR)-7/8 resiquimod and were examined for lung and spleen expression of pro-inflammatory cytokines and blood formula. Finally, chronically IP-pre-treated mice challenged with LPS injection were studied for "sickness" behavior. Additionally, the IP was analyzed using high-potency-liquid chromatography (HPLC)-high-resolution-mass-spectrometry (HRMS). Results LPS-induced in vitro release of TNF and IL-1β was reduced by both treatments. The IP-treated mice displayed blunted over-expression of SAA-2, ACE-2, CXCL1, and CXCL10 and decreased changes in blood formula in response to an injection with resiquimod. The IP-treated mice injected with LPS showed normalized locomotion, anxiety, and exploration behaviors but not abnormal forced swimming. Isoquercitrin, choline, leucine, chlorogenic acid, and other constituents were identified by HPLC-HRMS and likely underlie the IP immunomodulatory effects. Conclusions Herbal IP-therapy decreases inflammation and, partly, "sickness behavior," suggesting its potency to combat SARS-CoV-2 infection first of all via its preventive effects.
Collapse
Affiliation(s)
- Olesia Schapovalova
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Anna Gorlova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Johannes de Munter
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Elisaveta Sheveleva
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Mikhail Eropkin
- Department of Etiology and Epidemiology, Smorodintsev Research Institute of Influenza, St. Petersburg State University, Saint Petersburg, Russia
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Michail Sicker
- Rehabilitation Research Unit of Clinic of Bad Kreuzbach, Bad Kreuzbach, Germany
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergiy Lyubchyk
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- EIGES Center, Universidade Lusofona, Lisboa, Portugal
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
13
|
Lovelock DF, Liu W, Langston SE, Liu J, Van Voorhies K, Giffin KA, Vetreno RP, Crews FT, Besheer J. The Toll-like receptor 7 agonist imiquimod increases ethanol self-administration and induces expression of Toll-like receptor related genes. Addict Biol 2022; 27:e13176. [PMID: 35470561 PMCID: PMC9286850 DOI: 10.1111/adb.13176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/24/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022]
Abstract
There is growing evidence that immune signalling may be involved in both the causes and consequences of alcohol abuse. Toll-like receptor (TLR) expression is increased by alcohol consumption and is implicated in AUD, and specifically TLR7 may play an important role in ethanol consumption. We administered the TLR7-specific agonist imiquimod in male and female Long-Evans rats to determine (1) gene expression changes in brain regions involved in alcohol reinforcement, the nucleus accumbens core and anterior insular cortex, in rats with and without an alcohol history, and (2) whether TLR7 activation could modulate operant alcohol self-administration. Interferon regulatory factor 7 (IRF7) was dramatically increased in both sexes at both 2- and 24-h post-injection regardless of alcohol history and TLR3 and 7 gene expression was increased as well. The proinflammatory cytokine TNFα was increased 24-h post-injection in rats with an alcohol self-administration history, but this effect did not persist after four injections, suggesting molecular tolerance. Ethanol consumption was increased 24 h after imiquimod injections but did not occur until the third injection, suggesting adaptation to repeated TLR7 activation is necessary for increased drinking to occur. Notably, imiquimod reliably induced weight loss, indicating that sickness behaviour persisted across repeated injections. These findings show that TLR7 activation can modulate alcohol drinking in an operant self-administration paradigm and suggest that TLR7 and IRF7 signalling pathways may be a viable druggable target for treatment of AUD.
Collapse
Affiliation(s)
- Dennis F. Lovelock
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Wen Liu
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Sarah E. Langston
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Jiaqi Liu
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kalynn Van Voorhies
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kaitlin A. Giffin
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
14
|
Di Zazzo A, De Piano M, Coassin M, Mori T, Balzamino BO, Micera A. Ocular surface toll like receptors in ageing. BMC Ophthalmol 2022; 22:185. [PMID: 35459112 PMCID: PMC9027701 DOI: 10.1186/s12886-022-02398-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/12/2022] [Indexed: 12/27/2022] Open
Abstract
Background To evaluate changes in Toll Like Receptors (TLRs) expression at the ocular surface of healthy volunteers within different age groups. Methods Fifty-one healthy volunteers were enrolled in a pilot observational study. Clinical function tests (OSDI questionnaire, Schirmer test type I and Break Up time) were assessed in all subjects. Temporal Conjunctival imprints were performed for molecular and immunohistochemical analysis to measure TLRs expression (TLR2, 4, 3, 5, 7, 8, 9 and MyD88). Results Immunofluorescence data showed an increased TLR2 and decreased TLR7 and TLR8 immunoreactivity in old conjunctival imprints. Up-regulation of TLR2 and down-regulation of TLR7, TLR8 and MyD88 transcripts expression corroborated the data. A direct correlation was showed between increasing ICAM-1 and increasing TLR2 changes with age. Within the age OSDI score increases, T-BUT values decrease, and goblet cells showed a decreasing trend. Conclusion Changes in TLRs expression are associated with ageing, suggesting physiological role of TLRs in modulating ocular surface immunity. TLRs age related changes may participate to the changes of ocular surface homeostatic mechanisms which lead to inflammAging.
Collapse
Affiliation(s)
- Antonio Di Zazzo
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Maria De Piano
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Rome, Italy
| | - Marco Coassin
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Tommaso Mori
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Rome, Italy
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Rome, Italy.
| |
Collapse
|
15
|
Systemic Administration of the TLR7/8 Agonist Resiquimod (R848) to Mice Is Associated with Transient, In Vivo-Detectable Brain Swelling. BIOLOGY 2022; 11:biology11020274. [PMID: 35205140 PMCID: PMC8869423 DOI: 10.3390/biology11020274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
Peripheral administration of the E. coli endotoxin lipopolysaccharide (LPS) to rats promotes secretion of pro-inflammatory cytokines and in previous studies was associated with transient enlargement of cortical volumes. Here, resiquimod (R848) was administered to mice to stimulate peripheral immune activation, and the effects on brain volumes and neurometabolites determined. After baseline scans, 24 male, wild-type C57BL mice were triaged into three groups including R848 at low (50 μg) and high (100 μg) doses and saline controls. Animals were scanned again at 3 h and 24 h following treatment. Sickness indices of elevated temperature and body weight loss were observed in all R848 animals. Animals that received 50 μg R848 exhibited decreases in hippocampal N-acetylaspartate and phosphocreatine at the 3 h time point that returned to baseline levels at 24 h. Animals that received the 100 μg R848 dose demonstrated transient, localized, volume expansion (~5%) detectable at 3 h in motor, somatosensory, and olfactory cortices; and pons. A metabolic response evident at the lower dose and a volumetric change at the higher dose suggests a temporal evolution of the effect wherein the neurochemical change is demonstrable earlier than neurostructural change. Transient volume expansion in response to peripheral immune stimulation corresponds with previous results and is consistent with brain swelling that may reflect CNS edema.
Collapse
|
16
|
Englmeier L, Subburayalu J. What's happening where when SARS-CoV-2 infects: are TLR7 and MAFB sufficient to explain patient vulnerability? Immun Ageing 2022; 19:6. [PMID: 35065665 PMCID: PMC8783172 DOI: 10.1186/s12979-022-00262-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/02/2022] [Indexed: 12/04/2022]
Abstract
The present COVID-19 pandemic has revealed that several characteristics render patients especially prone to developing severe COVID-19 disease, i.e., the male sex, obesity, and old age. An explanation for the observed pattern of vulnerability has been proposed which is based on the concept of low sensitivity of the TLR7-signaling pathway at the time of infection as a common denominator of vulnerable patient groups. We will discuss whether the concept of established TLR-tolerance in macrophages and dendritic cells of the obese and elderly prior to infection can explain not only the vulnerability of these two demographic groups towards development of a severe infection with SARS-CoV-2, but also the observed cytokine response in these vulnerable patients, which is skewed towards pro-inflammatory cytokines with a missing interferon signature.
Collapse
Affiliation(s)
- Ludwig Englmeier
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany. .,Patent Attorney Dr. Ludwig Englmeier, scrIPtum, Erlenaustrasse 11, 83080, Oberaudorf, Germany. .,Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany.
| | - Julien Subburayalu
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany.,Mildred Scheel Early Career Center, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
17
|
Yates AG, Weglinski CM, Ying Y, Dunstan IK, Strekalova T, Anthony DC. Nafamostat reduces systemic inflammation in TLR7-mediated virus-like illness. J Neuroinflammation 2022; 19:8. [PMID: 34991643 PMCID: PMC8734544 DOI: 10.1186/s12974-021-02357-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The serine protease inhibitor nafamostat has been proposed as a treatment for COVID-19, by inhibiting TMPRSS2-mediated viral cell entry. Nafamostat has been shown to have other, immunomodulatory effects, which may be beneficial for treatment, however animal models of ssRNA virus infection are lacking. In this study, we examined the potential of the dual TLR7/8 agonist R848 to mimic the host response to an ssRNA virus infection and the associated behavioural response. In addition, we evaluated the anti-inflammatory effects of nafamostat in this model. METHODS CD-1 mice received an intraperitoneal injection of R848 (200 μg, prepared in DMSO, diluted 1:10 in saline) or diluted DMSO alone, and an intravenous injection of either nafamostat (100 μL, 3 mg/kg in 5% dextrose) or 5% dextrose alone. Sickness behaviour was determined by temperature, food intake, sucrose preference test, open field and forced swim test. Blood and fresh liver, lung and brain were collected 6 h post-challenge to measure markers of peripheral and central inflammation by blood analysis, immunohistochemistry and qPCR. RESULTS R848 induced a robust inflammatory response, as evidenced by increased expression of TNF, IFN-γ, CXCL1 and CXCL10 in the liver, lung and brain, as well as a sickness behaviour phenotype. Exogenous administration of nafamostat suppressed the hepatic inflammatory response, significantly reducing TNF and IFN-γ expression, but had no effect on lung or brain cytokine production. R848 administration depleted circulating leukocytes, which was restored by nafamostat treatment. CONCLUSIONS Our data indicate that R848 administration provides a useful model of ssRNA virus infection, which induces inflammation in the periphery and CNS, and virus infection-like illness. In turn, we show that nafamostat has a systemic anti-inflammatory effect in the presence of the TLR7/8 agonist. Therefore, the results indicate that nafamostat has anti-inflammatory actions, beyond its ability to inhibit TMPRSS2, that might potentiate its anti-viral actions in pathologies such as COVID-19.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Caroline M Weglinski
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Yuxin Ying
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Isobel K Dunstan
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Tatyana Strekalova
- Sechenov First Moscow State Medical University, Moscow, Russia
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Daniel C Anthony
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russia.
- University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
18
|
Zhang S, Zeng J, Zhou Y, Gao R, Rice S, Guo X, Liu Y, Feng P, Zhao Z. Simultaneous Detection of Herpes Simplex Virus Type 1 Latent and Lytic Transcripts in Brain Tissue. ASN Neuro 2022; 14:17590914211053505. [PMID: 35164537 PMCID: PMC9171132 DOI: 10.1177/17590914211053505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022] Open
Abstract
Neurotrophic herpes simplex virus type 1 (HSV-1) establishes lifelong latent infection in humans. Accumulating studies indicate that HSV-1, a risk factor of neurodegenerative diseases, exacerbates the sporadic Alzheimer's disease (AD). The analysis of viral genetic materials via genomic sequencing and quantitative PCR (qPCR) is the current approach used for the detection of HSV-1; however, this approach is limited because of its difficulty in detecting both latent and lytic phases of the HSV-1 life cycle in infected hosts. RNAscope, a novel in situ RNA hybridization assay, enables visualized detection of multiple RNA targets on tissue sections. Here, we developed a fluorescent multiplex RNAscope assay in combination with immunofluorescence to detect neuronal HSV-1 transcripts in various types of mouse brain samples and human brain tissues. Specifically, the RNA probes were designed to separately recognize two transcripts in the same brain section: (1) the HSV-1 latency-associated transcript (LAT) and (2) the lytic-associated transcript, the tegument protein gene of the unique long region 37 (UL37). As a result, both LAT and UL37 signals were detectable in neurons in the hippocampus and trigeminal ganglia (TG). The quantifications of HSV-1 transcripts in the TG and CNS neurons are correlated with the viral loads during lytic and latent infection. Collectively, the development of combinational detection of neuronal HSV-1 transcripts in mouse brains can serve as a valuable tool to visualize HSV-1 infection phases in various types of samples from AD patients and facilitate our understanding of the infectious origin of neurodegeneration and dementia.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jianxiong Zeng
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yuzheng Zhou
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Ruoyun Gao
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Stephanie Rice
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Xinying Guo
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yongzhen Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Zhen Zhao
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Kwon J, Suessmilch M, McColl A, Cavanagh J, Morris BJ. Distinct trans-placental effects of maternal immune activation by TLR3 and TLR7 agonists: implications for schizophrenia risk. Sci Rep 2021; 11:23841. [PMID: 34903784 PMCID: PMC8668921 DOI: 10.1038/s41598-021-03216-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
Exposure to infection in utero predisposes towards psychiatric diseases such as autism, depression and schizophrenia in later life. The mechanisms involved are typically studied by administering mimetics of double-stranded (ds) virus or bacterial infection to pregnant rats or mice. The effect of single-stranded (ss) virus mimetics has been largely ignored, despite evidence linking prenatal ss virus exposure with psychiatric disease. Understanding the effects of gestational ss virus exposure has become even more important with recent events. In this study, in pregnant mice, we compare directly the effects, on the maternal blood, placenta and the embryonic brain, of maternal administration of ds-virus mimetic poly I:C (to activate Toll-like receptor 3, TLR3) and ss-virus mimetic resiquimod (to activate TLR7/8). We find that, 4 h after the administration, both poly I:C and resiquimod elevated the levels of IL-6, TNFα, and chemokines including CCL2 and CCL5, in maternal plasma. Both agents also increased placental mRNA levels of IL-6 and IL-10, but only resiquimod increased placental TNFα mRNA. In foetal brain, poly I:C produced no detectable immune-response-related increases, whereas pronounced increases in cytokine (e.g. Il-6, Tnfα) and chemokine (e.g. Ccl2, Ccl5) expression were observed with maternal resiquimod administration. The data show substantial differences between the effect of maternal exposure to a TLR7/8 activator as compared to a TLR3 activator. There are significant implications for future modelling of diseases where maternal ss virus exposure contributes to environmental disease risk in offspring.
Collapse
Affiliation(s)
- Jaedeok Kwon
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Maria Suessmilch
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Alison McColl
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
20
|
Potential value of pharmacological agents acting on toll-like receptor (TLR) 7 and/or TLR8 in COVID-19. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100068. [PMID: 34870161 PMCID: PMC8562070 DOI: 10.1016/j.crphar.2021.100068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic is an ongoing global pandemic of coronavirus disease 2019 as an atypical type of viral pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many potential pharmacotherapies are currently being investigated against this disease. This article points to and justifies, the importance of investigating the potential therapeutic value of pharmacological agents acting on Toll-like Receptor (TLR) 7 and/or TLR8 as double-edged swords combating COVID-19. Induction of TLR7 and/or TLR8 may be investigated as a strategy to stimulate immunity and may be added to anti-COVID19 vaccines to cope with their current viral escape challenge. TLR7 stimulation may not only help viral clearance through Th1 antiviral responses but may also provide beneficial broncho- and vaso-dilatory, as well as, anti-inflammatory effects. Pharmacological compounds acting as TLR7 and/or TLR8 agonists may be of value if used by frontline healthcare workers with comorbidities who demonstrate mild symptoms of the disease. On the other hand, TLR7 and/or TLR8 antagonists may be used in combination with immune-modulatory/anti-inflammatory drugs in severe cases of the disease, with potential synergistic effects that could also help in reducing the doses of such therapies, and consequently their adverse effects. There is potential value in development of TLR7/8 agonists/antagonists as double-edged swords combating COVID-19. TLR7/8 agonists, as immune-stimulants in mild disease, may also be added to preventive vaccines acting against viral escape. TLR7/8 antagonists may be combined with immune-suppressants in severe cases for potential synergism and minimized toxicity.
Collapse
|
21
|
Carloni E, Ramos A, Hayes LN. Developmental Stressors Induce Innate Immune Memory in Microglia and Contribute to Disease Risk. Int J Mol Sci 2021; 22:13035. [PMID: 34884841 PMCID: PMC8657756 DOI: 10.3390/ijms222313035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Many types of stressors have an impact on brain development, function, and disease susceptibility including immune stressors, psychosocial stressors, and exposure to drugs of abuse. We propose that these diverse developmental stressors may utilize a common mechanism that underlies impaired cognitive function and neurodevelopmental disorders such as schizophrenia, autism, and mood disorders that can develop in later life as a result of developmental stressors. While these stressors are directed at critical developmental windows, their impacts are long-lasting. Immune activation is a shared pathophysiology across several different developmental stressors and may thus be a targetable treatment to mitigate the later behavioral deficits. In this review, we explore different types of prenatal and perinatal stressors and their contribution to disease risk and underlying molecular mechanisms. We highlight the impact of developmental stressors on microglia biology because of their early infiltration into the brain, their critical role in brain development and function, and their long-lived status in the brain throughout life. Furthermore, we introduce innate immune memory as a potential underlying mechanism for developmental stressors' impact on disease. Finally, we highlight the molecular and epigenetic reprogramming that is known to underlie innate immune memory and explain how similar molecular mechanisms may be at work for cells to retain a long-term perturbation after exposure to developmental stressors.
Collapse
Affiliation(s)
- Elisa Carloni
- Department of Molecular and Cellular Biology, Dartmouth College, Hanover, NH 03755, USA;
| | - Adriana Ramos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Lindsay N. Hayes
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Chand S, Gowen A, Savine M, Moore D, Clark A, Huynh W, Wu N, Odegaard K, Weyrich L, Bevins RA, Fox HS, Pendyala G, Yelamanchili SV. A comprehensive study to delineate the role of an extracellular vesicle-associated microRNA-29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021; 10:e12177. [PMID: 34913274 PMCID: PMC8674191 DOI: 10.1002/jev2.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), which express a repertoire of cargo molecules (cf. proteins, microRNA, lipids, etc.), have been garnering a prominent role in the modulation of several cellular processes. Here, using both non-human primate and rodent model systems, we provide evidence that brain-derived EV (BDE) miRNA, miR-29a-3p (mir-29a), is significantly increased during chronic methamphetamine (MA) exposure. Further, miR-29a levels show significant increase both with drug-seeking and reinstatement in a rat MA self-administration model. We also show that EV-associated miR-29a is enriched in EV pool comprising of small EVs and exomeres and further plays a critical role in MA-induced inflammation and synaptodendritic damage. Furthermore, treatment with the anti-inflammatory drug ibudilast (AV411), which is known to reduce MA relapse, decreased the expression of miR-29a and subsequently attenuated inflammation and rescued synaptodendritic injury. Finally, using plasma from MUD subjects, we provide translational evidence that EV-miR29a could potentially serve as a biomarker to detect neuronal damage in humans diagnosed with MA use disorder (MUD). In summary, our work suggests that EV-associated miR-29a-3p plays a crucial role in MUD and might be used as a potential blood-based biomarker for detecting chronic inflammation and synaptic damage.
Collapse
Affiliation(s)
- Subhash Chand
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Austin Gowen
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Mason Savine
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Dalia Moore
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Alexander Clark
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Wendy Huynh
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Niming Wu
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Katherine Odegaard
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | | | - Rick A. Bevins
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Howard S. Fox
- Department of Neurological SciencesUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Gurudutt Pendyala
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Sowmya V. Yelamanchili
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
23
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
24
|
Varshney D, Qiu SY, Graf TP, McHugh KJ. Employing Drug Delivery Strategies to Overcome Challenges Using TLR7/8 Agonists for Cancer Immunotherapy. AAPS JOURNAL 2021; 23:90. [PMID: 34181117 DOI: 10.1208/s12248-021-00620-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) are a potential target for cancer immunotherapy due to their role in the activation of the innate immune system. More specifically, TLR7 and TLR8, two structurally similar pattern recognition receptors that trigger interferon and cytokine responses, have proven to be therapeutically relevant targets for cancer in numerous preclinical and clinical studies. When triggered by an agonist, such as imiquimod or resiquimod, the TLR7/8 activation pathway induces cellular and humoral immune responses that can kill cancer cells with high specificity. Unfortunately, TLR7/8 agonists also present a number of issues that must be overcome prior to broad clinical implementation, such as poor drug solubility and systemic toxic effects. To overcome the key limitations of TLR7/8 agonists as a cancer therapy, biomaterial-based drug delivery systems have been developed. These delivery devices are highly diverse in their design and include systems that can be directly administered to the tumor, passively accumulated in relevant cancerous and lymph tissues, triggered by environmental stimuli, or actively targeted to specific physiological areas and cellular populations. In addition to improved delivery systems, recent studies have also demonstrated the potential benefits of TLR7/8 agonist co-delivery with other types of therapies, particularly checkpoint inhibitors, cancer vaccines, and chemotherapeutics, which can yield impressive anti-cancer effects. In this review, we discuss recent advances in the development of TLR7/8 agonist delivery systems and provide perspective on promising future directions.
Collapse
Affiliation(s)
- Dhruv Varshney
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Sherry Yue Qiu
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Tyler P Graf
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.
| |
Collapse
|
25
|
Lin W, Li C, Xu N, Watanabe M, Xue R, Xu A, Araki M, Sun R, Liu C, Nasu Y, Huang P. Dual-Functional PLGA Nanoparticles Co-Loaded with Indocyanine Green and Resiquimod for Prostate Cancer Treatment. Int J Nanomedicine 2021; 16:2775-2787. [PMID: 33880023 PMCID: PMC8052122 DOI: 10.2147/ijn.s301552] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE With the advance of screening techniques, there is a growing number of low-risk or intermediate-risk prostate cancer (PCa) cases, remaining a serious threat to men's health. To obtain better efficacy, a growing interest has been attracted to develop such emerging treatments as immunotherapy and focal therapy. However, few studies offer guidance on whether and how to combine these modalities against PCa. This study was designed to develop dual-functional nanoparticles (NPs) which combined photothermal therapy (PTT) with immunotherapy and determine the anti-tumor efficacy for PCa treatment. METHODS By a double emulsion technique, the drug nanocarrier, poly(lactic-co-glycolic acid) or PLGA, was applied for co-loading of a fluorescent dye, indocyanine green (ICG) and a toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) to synthesize PLGA-ICG-R848 NPs. Next, we determined their characteristic features and evaluated whether they inhibited the cell viability in multiple PCa cell lines. After treatment with PLGA-ICG-R848, the maturation markers of bone marrow-derived dendritic cells (BMDCs) were detected by flow cytometry. By establishing a subcutaneous xenograft model of mouse PCa, we explored both the anti-tumor effect and immune response following the NPs-based laser ablation. RESULTS With a mean diameter of 157.7 nm, PLGA-ICG-R848 exhibited no cytotoxic effect in PCa cells, but they significantly decreased RM9 cell viability to (3.9±1.0)% after laser irradiation. Moreover, PLGA-ICG-R848 promoted BMDCs maturation with the significantly elevated proportions of CD11c+CD86+ and CD11c+CD80+ cells. Following PLGA-ICG-R848-based laser ablation in vivo, the decreased bioluminescent signals indicated a significant inhibition of PCa growth, while the ratio of splenic natural killer (NK) cells in PLGA-ICG-R848 was (3.96±1.88)% compared with (0.99±0.10)% in PBS group, revealing the enhanced immune response against PCa. CONCLUSION The dual-functional PLGA-ICG-R848 NPs under laser irradiation exhibit the anti-tumor efficacy for PCa treatment by combining PTT with immunotherapy.
Collapse
Affiliation(s)
- Wenfeng Lin
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chaoming Li
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Naijin Xu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Ruizhi Xue
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ruifen Sun
- Center for Scientific Research, Yunnan University of Chinese Traditional Medicine, Kunming, People’s Republic of China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Peng Huang
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Okayama Medical Innovation Center, Okayama University, Okayama, Japan
| |
Collapse
|
26
|
Grantham EK, Warden AS, McCarthy GS, DaCosta A, Mason S, Blednov Y, Mayfield RD, Harris RA. Role of toll-like receptor 7 (TLR7) in voluntary alcohol consumption. Brain Behav Immun 2020; 89:423-432. [PMID: 32726684 PMCID: PMC7572874 DOI: 10.1016/j.bbi.2020.07.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/09/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
Overactivation of neuroimmune signaling has been linked to excessive ethanol consumption. Toll-like receptors (TLRs) are a major component of innate immune signaling and initiate anti- and pro-inflammatory responses via intracellular signal transduction cascades. TLR7 is upregulated in post-mortem brain tissue from humans with alcohol use disorder (AUD) and animals with prior exposure to ethanol. Despite this evidence, the role of TLR7 in the regulation of voluntary ethanol consumption has not been studied. We test the hypothesis that TLR7 activation regulates voluntary ethanol drinking behavior by administering a TLR7 agonist (R848) during an intermittent access drinking procedure in mice. Acute activation of TLR7 reduced ethanol intake, preference, and total fluid intake due, at least in part, to an acute sickness response. However, chronic pre-treatment with R848 resulted in tolerance to the adverse effects of the drug and a subsequent increase in ethanol consumption. To determine the molecular machinery that mediates these behavioral changes, we evaluated gene expression after acute and chronic TLR7 activation. We found that acute TLR7 activation produces brain region specific changes in expression of immune pathway genes, whereas chronic TLR7 activation causes downregulation of TLRs and blunted cytokine induction, suggesting molecular tolerance. Our results demonstrate a novel role for TLR7 signaling in regulating voluntary ethanol consumption. Taken together, our findings suggest TLR7 may be a viable target for development of therapies to treat AUD.
Collapse
Affiliation(s)
- E K Grantham
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | - A S Warden
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - G S McCarthy
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - A DaCosta
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - S Mason
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - Y Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA
| | - R D Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2500 Speedway, Stop 14800, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
27
|
Lovasz RM, Marks DL, Chan BK, Saunders KE. Effects on Mouse Food Consumption After Exposure to Bedding from Sick Mice or Healthy Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2020; 59:687-694. [PMID: 32859281 DOI: 10.30802/aalas-jaalas-19-000154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Control mice housed in the same room as mice with pancreatic ductal adenocarcinoma (PDAC) demonstrate decreased food intake coincident with the cachexia experienced by the mice with PDAC. Mice are considered an empathetic species, and we hypothesized that the reduced food intake in normal mice was an "empathy state" that was mediated by olfactory cues. Naïve male and female C57BL/6 mice were exposed to soiled bedding from mice experiencing PDAC induced cachexia or from control mice in the PDAC study. Body weight, food intake, and food spillage were measured across 48 h. Statistically significant differences in food consumption were found at various time points in both positive and negative directions for the 2 bedding conditions, and the direction of effect was opposite for males and females. Although analysis of data from previous PDAC studies showed differences in food spillage between PDAC mice and their controls, in this study we found no correlation between food consumption and food spillage based on bedding type. Disruption of food intake due to the "empathy state" requires testing larger numbers of animals to attain appropriate statistical power, which is contrary to the goal of using fewer animals. Empathy effects require careful consideration of sample size and cautious interpretation of results. This study also highlights the importance of sex as a biologic variable and why quantifying food spillage is important in studies of food intake.
Collapse
Affiliation(s)
- Rebecca M Lovasz
- Department of Comparative Medicine, Oregon Health & Science University, Portland, Oregon;,
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon
| | - Benjamin K Chan
- Biostatistics and Design Program, Oregon Health & Science University, Portland, Oregon
| | - Kim E Saunders
- Department of Comparative Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
28
|
Almarzooq S, Kwon J, Willis A, Craig J, Morris BJ. Novel alternatively-spliced exons of the VRK2 gene in mouse brain and microglial cells. Mol Biol Rep 2020; 47:5127-5136. [PMID: 32583282 PMCID: PMC7417415 DOI: 10.1007/s11033-020-05584-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
Abstract
Common sequence variations in the VRK2 gene contribute to genetic risk for various psychiatric diseases including schizophrenia and major depressive disorder. Despite the clear importance of studying the regulation and function of VRK2 for understanding the causes of these diseases, the organisation and expression of the gene remain poorly characterised. Using reverse-transcriptase-PCR, we have amplifed exons of Vrk2 mRNA from regions of mouse brain, and from different cell classes comprising neurones, astrocytes and microglial cells. We find that Vrk2 mRNA is expressed in all cell types, and that the splicing of the mouse Vrk2 gene is much more complex than previously appreciated. In addition to the predicted alternative splicing (absence/presence) of the penultimate 3 prime exon, we also detected a variety of 5 prime structures, including two novel exons spanning the first characterised exon (exon 1), which we term exons 1a and 1b. While expressed in neurones and astrocytes, exon 1b was not expressed in microglial cells. Expression of transcripts containing exon 1a in microglia was increased by immune stimulation. An additional truncated transcript lacking 7 central exons was also identified. As with the human gene, the results confirm complex patterns of alternative splicing which are likely to be relevant for understanding the physiological and pathological function of the gene in the CNS.
Collapse
Affiliation(s)
- Salsabil Almarzooq
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, G12 8QQ, Glasgow, UK
| | - Jaedeok Kwon
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, G12 8QQ, Glasgow, UK
| | - Ashleigh Willis
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, G12 8QQ, Glasgow, UK
| | - John Craig
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, G12 8QQ, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, G12 8QQ, Glasgow, UK.
| |
Collapse
|
29
|
Shi Z, Pelletier NE, Wong J, Li B, Sdrulla AD, Madden CJ, Marks DL, Brooks VL. Leptin increases sympathetic nerve activity via induction of its own receptor in the paraventricular nucleus. eLife 2020; 9:e55357. [PMID: 32538782 PMCID: PMC7316512 DOI: 10.7554/elife.55357] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Whether leptin acts in the paraventricular nucleus (PVN) to increase sympathetic nerve activity (SNA) is unclear, since PVN leptin receptors (LepR) are sparse. We show in rats that PVN leptin slowly increases SNA to muscle and brown adipose tissue, because it induces the expression of its own receptor and synergizes with local glutamatergic neurons. PVN LepR are not expressed in astroglia and rarely in microglia; instead, glutamatergic neurons express LepR, some of which project to a key presympathetic hub, the rostral ventrolateral medulla (RVLM). In PVN slices from mice expressing GCaMP6, leptin excites glutamatergic neurons. LepR are expressed mainly in thyrotropin-releasing hormone (TRH) neurons, some of which project to the RVLM. Injections of TRH into the RVLM and dorsomedial hypothalamus increase SNA, highlighting these nuclei as likely targets. We suggest that this neuropathway becomes important in obesity, in which elevated leptin maintains the hypothalamic pituitary thyroid axis, despite leptin resistance.
Collapse
Affiliation(s)
- Zhigang Shi
- Department of Physiology and PharmacologyPortlandUnited States
| | | | - Jennifer Wong
- Department of Physiology and PharmacologyPortlandUnited States
| | - Baoxin Li
- Department of Physiology and PharmacologyPortlandUnited States
| | - Andrei D Sdrulla
- Department of Anesthesiology and Perioperative MedicinePortlandUnited States
| | | | - Daniel L Marks
- Department of Pediatrics, Pape Family Pediatric Research Institute, Brenden-Colson Center for Pancreatic Care Oregon Health & Science UniversityPortlandUnited States
| | | |
Collapse
|