1
|
Oliveira LFS, Wu S, Dasuri VS, Harrington AW, Olaloye O, Goldsmith J, Breault DT, Konnikova L, O’Connell AE. Cataloguing the postnatal small intestinal transcriptome during the period of susceptibility to necrotizing enterocolitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.612672. [PMID: 39386454 PMCID: PMC11463582 DOI: 10.1101/2024.09.25.612672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
In the first postnatal month, the developing mouse intestine shifts from an immature to a mature intestine that will sustain the organism throughout the lifespan. Here, we surveyed the mouse intestine in C57Bl/6 mice by RNA-Seq to evaluate the changes in gene expression over time from the day of birth through 1 month of age in both the duodenum and ileum. We analyzed gene expression for changes in gene families that correlated with the periods of NEC susceptibility or resistance. We highlight that increased expression of DNA processing genes and vacuolar structure genes, tissue development and morphogenesis genes, and cell migration genes all correlated with NEC susceptibility, while increases in immunity gene sets, intracellular transport genes, ATP production, and intracellular metabolism genes correlated with NEC resistance. Using trends identified in the RNA analyses, we further evaluated expression of cellular markers and epithelial regulators, immune cell markers, and adenosine metabolism components. We confirmed key changes with qRT-PCR and immunofluorescence. In addition, we compared some findings to humans using human intestinal biopsies and organoids. This dataset can serve as a reference for other groups considering the role of single molecules or molecular families in early intestinal and postnatal development.
Collapse
Affiliation(s)
| | - Shania Wu
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | | | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA
- Harvard Stem Cell Institute, Boston, MA
- Dept. of Pediatrics, Harvard Medical School, Boston, MA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA
- Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA
- Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Amy E. O’Connell
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Dept. of Pediatrics, Harvard Medical School, Boston, MA
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
2
|
Yamashita A, Ito Y, Osada M, Matsuda H, Hosono K, Tsujikawa K, Okamoto H, Amano H. RAMP1 Signaling Mitigates Acute Lung Injury by Distinctively Regulating Alveolar and Monocyte-Derived Macrophages. Int J Mol Sci 2024; 25:10107. [PMID: 39337592 PMCID: PMC11432488 DOI: 10.3390/ijms251810107] [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: 09/08/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury that induces cytokine hypersecretion. Receptor activity-modifying protein (RAMP) 1, a subunit of the calcitonin gene-related peptide (CGRP) receptor, regulates the production of cytokines. This study examined the role of RAMP1 signaling during lipopolysaccharide (LPS)-induced acute lung injury (ALI). LPS administration to wild-type (WT) mice depleted alveolar macrophages (AMs) and recruited monocyte-derived macrophages (MDMs) and neutrophils. RAMP1-deficient (RAMP1-/-) mice exhibited higher lung injury scores, cytokine levels, and cytokine-producing neutrophil infiltration. RAMP1-deficient AMs produced more cytokines in response to LPS than WT AMs. Adoptive transfer of RAMP1-deficient AMs to RAMP1-/- mice increased cytokine levels and neutrophil accumulation compared to the transfer of WT AMs. RAMP1-/- mice had reduced MDM recruitment and lower pro-inflammatory and reparative macrophage profiles. Cultured bone marrow (BM)-derived RAMP1-deficient macrophages stimulated with LPS showed decreased expression of pro-inflammatory and pro-repairing genes. CGRP administration to WT mice reduced cytokine production and neutrophil accumulation. These findings indicate that RAMP1 signaling mitigates LPS-induced ALI by inactivating AMs and promoting inflammatory and repair activities of MDMs. Targeting RAMP1 signaling presents a potential therapeutic approach for the treatment of ARDS.
Collapse
Affiliation(s)
- Atsushi Yamashita
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Yoshiya Ito
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Mayuko Osada
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Emergency and Critical Care Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Hiromi Matsuda
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan;
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Hideki Amano
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| |
Collapse
|
3
|
Zhang XC, Zhou YW, Wei GX, Luo YQ, Qiu M. Locoregional therapies combined with immune checkpoint inhibitors for liver metastases. Cancer Cell Int 2024; 24:302. [PMID: 39217341 PMCID: PMC11365172 DOI: 10.1186/s12935-024-03484-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have achieved remarkable success in clinical research and practice. Notably, liver metastasis is not sensitive to ICIs. Liver locoregional therapies can cause irreversible damage to tumor cells and release tumor antigens, thereby providing a rationale for immunotherapy treatments in liver metastasis. The combination therapy of ICIs with locoregional therapies is a promising option for patients with liver metastasis. Preclinical studies have demonstrated that combining ICIs with locoregional therapies produces a significantly synergistic anti-tumor effect. However, the current evidence for the efficacy of ICIs combined with locoregional therapies remains insufficient. Therefore, we review the literature on the mechanisms of locoregional therapies in treating liver metastasis and the clinical research progress of their combination with ICIs.
Collapse
Affiliation(s)
- Xing-Chen Zhang
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Gui-Xia Wei
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yi-Qiao Luo
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
4
|
Gottipati MK, D'Amato AR, Saksena J, Popovich PG, Wang Y, Gilbert RJ. Delayed administration of interleukin-4 coacervate alleviates the neurotoxic phenotype of astrocytes and promotes functional recovery after a contusion spinal cord injury. J Neural Eng 2024; 21:046052. [PMID: 39029499 DOI: 10.1088/1741-2552/ad6596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/19/2024] [Indexed: 07/21/2024]
Abstract
Objective. Macrophages and astrocytes play a crucial role in the aftermath of a traumatic spinal cord injury (SCI). Infiltrating macrophages adopt a pro-inflammatory phenotype while resident astrocytes adopt a neurotoxic phenotype at the injury site, both of which contribute to neuronal death and inhibit axonal regeneration. The cytokine interleukin-4 (IL-4) has shown significant promise in preclinical models of SCI by alleviating the macrophage-mediated inflammation and promoting functional recovery. However, its effect on neurotoxic reactive astrocytes remains to be elucidated, which we explored in this study. We also studied the beneficial effects of a sustained release of IL-4 from an injectable biomaterial compared to bolus administration of IL-4.Approach. We fabricated a heparin-based coacervate capable of anchoring and releasing bioactive IL-4 and tested its efficacyin vitroandin vivo. Main results. We show that IL-4 coacervate is biocompatible and drives a robust anti-inflammatory macrophage phenotype in culture. We also show that IL-4 and IL-4 coacervate can alleviate the reactive neurotoxic phenotype of astrocytes in culture. Finally, using a murine model of contusion SCI, we show that IL-4 and IL-4 coacervate, injected intraspinally 2 d post-injury, can reduce macrophage-mediated inflammation, and alleviate neurotoxic astrocyte phenotype, acutely and chronically, while also promoting neuroprotection with significant improvements in hindlimb locomotor recovery. We observed that IL-4 coacervate can promote a more robust regenerative macrophage phenotypein vitro, as well as match its efficacyin vivo,compared to bolus IL-4.Significance. Our work shows the promise of coacervate as a great choice for local and prolonged delivery of cytokines like IL-4. We support this by showing that the coacervate can release bioactive IL-4, which acts on macrophages and astrocytes to promote a pro-regenerative environment following a SCI leading to robust neuroprotective and functional outcomes.
Collapse
Affiliation(s)
- Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
- Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
| | - Anthony R D'Amato
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 134 Hollister Drive, 283 Kimball Hall, Ithaca, NY 14853, United States of America
| | - Jayant Saksena
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
- Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
| | - Yadong Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 134 Hollister Drive, 283 Kimball Hall, Ithaca, NY 14853, United States of America
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
| |
Collapse
|
5
|
Anitua E, Troya M, Alkhraisat MH. Immunoregulatory role of platelet derivatives in the macrophage-mediated immune response. Front Immunol 2024; 15:1399130. [PMID: 38983851 PMCID: PMC11231193 DOI: 10.3389/fimmu.2024.1399130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Background Macrophages are innate immune cells that display remarkable phenotypic heterogeneity and functional plasticity. Due to their involvement in the pathogenesis of several human conditions, macrophages are considered to be an attractive therapeutic target. In line with this, platelet derivatives have been successfully applied in many medical fields and as active participants in innate immunity, cooperation between platelets and macrophages is essential. In this context, the aim of this review is to compile the current evidence regarding the effects of platelet derivatives on the phenotype and functions of macrophages to identify the advantages and shortcomings for feasible future clinical applications. Methods A total of 669 articles were identified during the systematic literature search performed in PubMed and Web of Science databases. Results A total of 27 articles met the inclusion criteria. Based on published findings, platelet derivatives may play an important role in inducing a dynamic M1/M2 balance and promoting a timely M1-M2 shift. However, the differences in procedures regarding platelet derivatives and macrophages polarization and the occasional lack of information, makes reproducibility and comparison of results extremely challenging. Furthermore, understanding the differences between human macrophages and those derived from animal models, and taking into account the peculiarities of tissue resident macrophages and their ontogeny seem essential for the design of new therapeutic strategies. Conclusion Research on the combination of macrophages and platelet derivatives provides relevant information on the function and mechanisms of the immune response.
Collapse
Affiliation(s)
- Eduardo Anitua
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - María Troya
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mohammad H. Alkhraisat
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
6
|
Hughes HK, Moreno RJ, Ashwood P. Innate Immune Dysfunction and Neuroinflammation in Autism Spectrum Disorder (ASD). FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2024; 22:229-241. [PMID: 38680981 PMCID: PMC11046725 DOI: 10.1176/appi.focus.24022004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation. Appeared originally in Brain Behav Immun 2023; 108:245-254.
Collapse
Affiliation(s)
- H K Hughes
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA (all authors);The M.I.N.D. Institute, University of California at Davis, CA, USA (all authors)
| |
Collapse
|
7
|
Ankley LM, Conner KN, Vielma TE, Godfrey JJ, Thapa M, Olive AJ. GSK3α/β Restrain IFN-γ-Inducible Costimulatory Molecule Expression in Alveolar Macrophages, Limiting CD4+ T Cell Activation. Immunohorizons 2024; 8:147-162. [PMID: 38345473 PMCID: PMC10916365 DOI: 10.4049/immunohorizons.2300107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/15/2024] Open
Abstract
Macrophages play a crucial role in eliminating respiratory pathogens. Both pulmonary resident alveolar macrophages (AMs) and recruited macrophages contribute to detecting, responding to, and resolving infections in the lungs. Despite their distinct functions, it remains unclear how these macrophage subsets regulate their responses to infection, including how activation by the cytokine IFN-γ is regulated. This shortcoming prevents the development of therapeutics that effectively target distinct lung macrophage populations without exacerbating inflammation. We aimed to better understand the transcriptional regulation of resting and IFN-γ-activated cells using a new ex vivo model of AMs from mice, fetal liver-derived alveolar-like macrophages (FLAMs), and immortalized bone marrow-derived macrophages. Our findings reveal that IFN-γ robustly activates both macrophage types; however, the profile of activated IFN-γ-stimulated genes varies greatly between these cell types. Notably, FLAMs show limited expression of costimulatory markers essential for T cell activation upon stimulation with only IFN-γ. To understand cell type-specific differences, we examined how the inhibition of the regulatory kinases GSK3α/β alters the IFN-γ response. GSK3α/β controlled distinct IFN-γ responses, and in AM-like cells, we found that GSK3α/β restrained the induction of type I IFN and TNF, thus preventing the robust expression of costimulatory molecules and limiting CD4+ T cell activation. Together, these data suggest that the capacity of AMs to respond to IFN-γ is restricted in a GSK3α/β-dependent manner and that IFN-γ responses differ across distinct macrophage populations. These findings lay the groundwork to identify new therapeutic targets that activate protective pulmonary responses without driving deleterious inflammation.
Collapse
Affiliation(s)
- Laurisa M. Ankley
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Kayla N. Conner
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Taryn E. Vielma
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Jared J. Godfrey
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Mahima Thapa
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Andrew J. Olive
- Department of Microbiology, Genetics, and Immunology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
8
|
Santamarina AB, Mennitti LV, de Souza EA, Mesquita LMDS, Noronha IH, Vasconcelos JRC, Prado CM, Pisani LP. A low-carbohydrate diet with different fatty acids' sources in the treatment of obesity: Impact on insulin resistance and adipogenesis. Clin Nutr 2023; 42:2381-2394. [PMID: 37862824 DOI: 10.1016/j.clnu.2023.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/23/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND The search for nutritional intervention strategies against obesity has grown, highlighting the low-carbohydrate diet model. However, little is known about the impact of the quality of fatty acids consumed in this diet. Thus, we aim to investigate the influence of fatty acid quality on dietary strategy on obesity. METHODS Male Swiss mice were diet-induced to obesity. Afterward, mice consume a low-carb diet with different types of fat: saturated, polyunsaturated ω-3, ω-6, and monounsaturated ω-9 fatty acids. Weight gain and food consumption were monitored weekly. An oral glucose tolerance test was performed and blood and tissue samples were collected for analysis of insulin resistance markers. Protein expression of insulin signaling pathway molecules, lipid metabolism, mitochondrial function, macrophage polarization, and cytokine production were analyzed. RESULTS The high-fat diet was able to induce obesity and glucose intolerance. The switch to a low-carbohydrate dietary pattern reversed the glucose intolerance, with better results in the ω-3 and ω-9 groups. After the low-carbohydrate diet, groups ω-3 and ω-9 presented improved fasting serum glucose, insulin, and HOMA indexes. The low-carbohydrate diet also increased the activity of insulin pathway proteins such as IR, IRS1, and AKT. Furthermore, the ω-3 diet group showed greater activity of mitochondrial complexes and AMPK signaling pathway proteins. The ω-6 and ω-9 -rich diet induced M2-type macrophage polarization, as well as cytokine production modulation by the low-carbohydrate diet in the ω-3 and ω-9 groups. CONCLUSIONS Consuming a low-carbohydrate diet pattern promotes weight loss and improves glucose intolerance in obesity. Also, the quality of lipids has a direct influence, demonstrating that the consumption of ω-3 polyunsaturated and ω-9 monounsaturated lipids can lead to more favorable outcomes for the improvement of glucose intolerance, lipid metabolism, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Aline B Santamarina
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Laís V Mennitti
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Esther A de Souza
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Leonardo M de Souza Mesquita
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas, Rua Pedro Zaccaria 1300, 13484-350 Limeira, São Paulo, Brazil
| | - Isaú H Noronha
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - José Ronnie C Vasconcelos
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Carla M Prado
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil.
| |
Collapse
|
9
|
Žaloudíková M. Mechanisms and Effects of Macrophage Polarization and Its Specifics in Pulmonary Environment. Physiol Res 2023; 72:S137-S156. [PMID: 37565418 PMCID: PMC10660583 DOI: 10.33549/physiolres.935058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 06/09/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophages are a specific group of cells found in all body tissues. They have specific characteristics in each of the tissues that correspond to the functional needs of the specific environment. These cells are involved in a wide range of processes, both pro-inflammatory and anti-inflammatory ("wound healing"). This is due to their specific capacity for so-called polarization, a phenotypic change that is, moreover, partially reversible compared to other differentiated cells of the human body. This promises a wide range of possibilities for its influence and thus therapeutic use. In this article, we therefore review the mechanisms that cause polarization, the basic classification of polarized macrophages, their characteristic markers and the effects that accompany these phenotypic changes. Since the study of pulmonary (and among them mainly alveolar) macrophages is currently the focus of scientific interest of many researchers and these macrophages are found in very specific environments, given mainly by the extremely high partial pressure of oxygen compared to other locations, which specifically affects their behavior, we will focus our review on this group.
Collapse
Affiliation(s)
- M Žaloudíková
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
10
|
Sedaka R, Huang J, Yamaguchi S, Lovelady C, Hsu JS, Shinde S, Kasztan M, Crossman DK, Saigusa T. Accelerated cystogenesis by dietary protein load is dependent on, but not initiated by kidney macrophages. Front Med (Lausanne) 2023; 10:1173674. [PMID: 37538309 PMCID: PMC10394241 DOI: 10.3389/fmed.2023.1173674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023] Open
Abstract
Background Disease severity of autosomal dominant polycystic kidney disease (ADPKD) is influenced by diet. Dietary protein, a recognized cyst-accelerating factor, is catabolized into amino acids (AA) and delivered to the kidney leading to renal hypertrophy. Injury-induced hypertrophic signaling in ADPKD results in increased macrophage (MФ) activation and inflammation followed by cyst growth. We hypothesize that the cystogenesis-prompting effects of HP diet are caused by increased delivery of specific AA to the kidney, ultimately stimulating MФs to promote cyst progression. Methods Pkd1flox/flox mice with and without Cre (CAGG-ER) were given tamoxifen to induce global gene deletion (Pkd1KO). Pkd1KO mice were fed either a low (LP; 6%), normal (NP; 18%), or high (HP; 60%) protein diet for 1 week (early) or 6 weeks (chronic). Mice were then euthanized and tissues were used for histology, immunofluorescence and various biochemical assays. One week fed kidney tissue was cell sorted to isolate tubular epithelial cells for RNA sequencing. Results Chronic dietary protein load in Pkd1KO mice increased kidney weight, number of kidney infiltrating and resident MФs, chemokines, cytokines and cystic index compared to LP diet fed mice. Accelerated cyst growth induced by chronic HP were attenuated by liposomal clodronate-mediated MФ depletion. Early HP diet fed Pkd1KO mice had larger cystic kidneys compared to NP or LP fed counterparts, but without increases in the number of kidney MФs, cytokines, or markers of tubular injury. RNA sequencing of tubular epithelial cells in HP compared to NP or LP diet group revealed increased expression of sodium-glutamine transporter Snat3, chloride channel Clcnka, and gluconeogenesis marker Pepck1, accompanied by increased excretion of urinary ammonia, a byproduct of glutamine. Early glutamine supplementation in Pkd1KO mice lead to kidney hypertrophy. Conclusion Chronic dietary protein load-induced renal hypertrophy and accelerated cyst growth in Pkd1KO mice is dependent on both infiltrating and resident MФ recruitment and subsequent inflammatory response. Early cyst expansion by HP diet, however, is relient on increased delivery of glutamine to kidney epithelial cells, driving downstream metabolic changes prior to inflammatory provocation.
Collapse
Affiliation(s)
- Randee Sedaka
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jifeng Huang
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shinobu Yamaguchi
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Caleb Lovelady
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jung-Shan Hsu
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sejal Shinde
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Malgorzata Kasztan
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Takamitsu Saigusa
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
11
|
Fu M, Yang L, Wang H, Chen Y, Chen X, Hu Q, Sun H. Research progress into adipose tissue macrophages and insulin resistance. Physiol Res 2023; 72:287-299. [PMID: 37449743 PMCID: PMC10668993 DOI: 10.33549/physiolres.935046] [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: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 08/26/2023] Open
Abstract
In recent years, there has been an increasing incidence of metabolic syndrome, type 2 diabetes, and cardiovascular events related to insulin resistance. As one of the target organs for insulin, adipose tissue is essential for maintaining in vivo immune homeostasis and metabolic regulation. Currently, the specific adipose tissue mechanisms involved in insulin resistance remain incompletely understood. There is increasing evidence that the process of insulin resistance is mostly accompanied by a dramatic increase in the number and phenotypic changes of adipose tissue macrophages (ATMs). In this review, we discuss the origins and functions of ATMs, some regulatory factors of ATM phenotypes, and the mechanisms through which ATMs mediate insulin resistance. We explore how ATM phenotypes contribute to insulin resistance in adipose tissue. We expect that modulation of ATM phenotypes will provide a novel strategy for the treatment of diseases associated with insulin resistance.
Collapse
Affiliation(s)
- M Fu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | |
Collapse
|
12
|
Luo G, Zhou Z, Huang C, Zhang P, Sun N, Chen W, Deng C, Li X, Wu P, Tang J, Qing L. Itaconic acid induces angiogenesis and suppresses apoptosis via Nrf2/autophagy to prolong the survival of multi-territory perforator flaps. Heliyon 2023; 9:e17909. [PMID: 37456049 PMCID: PMC10345368 DOI: 10.1016/j.heliyon.2023.e17909] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
Background Perforator flaps are widely used in hand microsurgery to reconstruct and repair soft tissue injuries. However, ischemia and subsequent ischemia-reperfusion injury may cause distal necrosis of the flap. Itaconic acid (IA) is a modulator of macrophage function, which exerts anti-inflammatory effects in macrophage activation. Methods The necrotic area of the flap was detected by measuring the flap temperature with an infrared thermometer. Flap cell apoptosis was detected by TUNEL staining and Western blot analysis of the apoptosis-associated proteins Bcl-2 and Bax. HE staining was used to detect angiogenesis of the skin flaps. CD31 was detected to identify positive vascular expression, and the survival of choke vessels in different areas of the skin flap was assessed by arteriography. In addition, Western blot was performed to quantify the expressions of VEGF, Nrf2, LC3II, SQSTM1, and CTSD. Results Itaconic acid raises VEGF protein levels in skin flaps and the number of CD31-positive vessels. The skin flaps in the IA treatment group exhibited higher neovascularization and less necrosis than those in the control group. The results of TUNEL staining and Western blot revealed that Itaconic acid attenuated apoptosis in the ischemic area of flap. The combination of itaconic acid and Nrf2 inhibitor ML385 reversed this beneficial effect. The results revealed that itaconic acid attenuated apoptosis, enhanced angiogenesis, and enhanced autophagy. Conclusion In summary, our findings indicate that itaconic acid might be an effective treatment to reduce flap necrosis. Additionally, this study identified a novel mechanism for the effects of itaconic acid on flap survival.
Collapse
Affiliation(s)
- Gaojie Luo
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Zekun Zhou
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Chengxiong Huang
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Peiyao Zhang
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Nianzhe Sun
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Wei Chen
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Chao Deng
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Xiaoxiao Li
- Department of Pathology, Changsha Medical University, Changsha, China
| | - Panfeng Wu
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Juyu Tang
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Liming Qing
- Department of Microsurgery and Hand Surgery, Xiangya Hospital of Central South University, Changsha, 410008, China
| |
Collapse
|
13
|
Amin SN, Sakr HI, El Gazzar WB, Shaltout SA, Ghaith HS, Elberry DA. Combined saline and vildagliptin induced M2 macrophage polarization in hepatic injury induced by acute kidney injury. PeerJ 2023; 11:e14724. [PMID: 36815993 PMCID: PMC9933746 DOI: 10.7717/peerj.14724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/19/2022] [Indexed: 02/15/2023] Open
Abstract
Acute kidney injury (AKI) is a prevalent medical condition accompanied by mutual affection of other organs, including the liver resulting in complicated multiorgan malfunction. Macrophages play a vital role during tissue injury and healing; they are categorized into "classically activated macrophages" (M1) and "alternatively activated macrophages" (M2). The present study investigated and compared the conventional fluid therapy vs Dipeptidyl peptidase 4 inhibitor (DPP-4i) vildagliptin on the liver injury induced by AKI and evaluated the possible molecular mechanisms. Thirty rats comprised five groups (n = 6 rats/group): control, AKI, AKI+saline (received 1.5 mL of normal saline subcutaneous injection), AKI+vildagliptin (treated with oral vildagliptin 10 mg/kg), AKI+saline+vildagliptin. AKI was induced by intramuscular (i.m) injection of 50% glycerol (5 ml/kg). At the end of the work, we collected serum and liver samples for measurements of serum creatinine, blood urea nitrogen (BUN), alanine aminotransferase (ALT), aspartate aminotransferase (AST), tumor necrotic factor-α (TNF-α), and interleukin-10 (IL-10). Liver samples were processed for assessment of inducible nitric oxide synthase (iNOS) as a marker for M1, arginase 1 (Arg-1) as an M2 marker, c-fos, c-Jun, mitogen-activated protein kinase (MAPK), activator protein 1 (AP-1), and high-mobility-group-box1 (HMGB1) protein. The difference was insignificant regarding the relative expression of AP-1, c-Jun, c-fos, MAPK, and HMGB between the AKI+saline group and the AKI+Vildagliptin group. The difference between the same two groups concerning the hepatic content of the M1 marker (iNOS) and the M2 marker Arg-1 was insignificant. However, combined therapy produced more pronounced changes in these markers, as the difference in their relative expression between the AKI+saline+Vildagliptin group and both the AKI+saline group and the AKI+Vildagliptin group was significant. Accordingly, we suggest that the combined saline and vildagliptin hepatoprotective effect involves the downregulation of the MAPK/AP-1 signaling pathway.
Collapse
Affiliation(s)
- Shaimaa N. Amin
- Department of Anatomy, Physiology, and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan,Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hader I. Sakr
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt,Department of Medical Physiology, Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Walaa B. El Gazzar
- Department of Anatomy, Physiology, and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Sherif A. Shaltout
- Department of Pharmacology, Public health, and Clinical Skills, Faculty of Medicine, The Hashemite University, Zarqa, Jordan,Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | | | - Dalia A. Elberry
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Innate immune dysfunction and neuroinflammation in autism spectrum disorder (ASD). Brain Behav Immun 2023; 108:245-254. [PMID: 36494048 DOI: 10.1016/j.bbi.2022.12.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by communication and social behavior deficits. The presence of restricted and repetitive behaviors often accompanies these deficits, and these characteristics can range from mild to severe. The past several decades have seen a significant rise in the prevalence of ASD. The etiology of ASD remains unknown; however, genetic and environmental risk factors play a role. Multiple hypotheses converge to suggest that neuroinflammation, or at least the interaction between immune and neural systems, may be involved in the etiology of some ASD cases or groups. Repeated evidence of innate immune dysfunction has been seen in ASD, often associated with worsening behaviors. This evidence includes data from circulating myeloid cells and brain resident macrophages/microglia in both human and animal models. This comprehensive review presents recent findings of innate immune dysfunction in ASD, including aberrant innate cellular function, evidence of neuroinflammation, and microglia activation.
Collapse
|
15
|
Fan W, Xu H, Shen C, Fang J, Li X. Nrf2 orchestrates transition from acute to chronic otitis media through inflammatory macrophages. Front Immunol 2023; 14:1170388. [PMID: 37122744 PMCID: PMC10140394 DOI: 10.3389/fimmu.2023.1170388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Acute and chronic otitis media (AOM and COM) are common middle ear infections that can lead to hearing loss and other complications. Recent research has shown that both macrophages and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway are involved in the immune response to and the resolution of otitis media. However, the specific effects of Nrf2 on macrophages in the transition of AOM to COM are not well understood, and a practical approach to prevent this transition by targeting Nrf2/macrophages has not been established. Methods In an AOM mouse model using lipopolysaccharide (LPS) injection into the middle ear, middle ear effusion (OME)-macrophages were isolated and analyzed for Nrf2 expression. M2-like polarization of macrophages was induced by Nrf2 activation and its effects on inflammatory resolution were studied by examining inflammatory neutrophils and macrophages, proinflammatory cytokines, and oxidative levels. The survival of human middle ear epithelial cells (HMMECs) co-cultured with Nrf2-modified macrophages was also evaluated. Furthermore, restoration of Nrf2 in macrophages with adeno-associated virus (AAV) vectors was performed to determine the effect on the transition of AOM to COM in experimental mice. Results Reduced Nrf2 in OME-macrophages during the recovery phase was associated with uncured AOM or its development into COM, demonstrated by persistent increases in inflammatory neutrophils and macrophages, proinflammatory cytokines, and oxidative levels. Nrf2 activation induced M2-like polarization of macrophages, which improved the survival of co-cultured HMMECs treated with LPS in vitro. Restoration of Nrf2 in OME-derived low-Nrf2-expressing macrophages with AAV vectors significantly inhibited the transition of AOM to COM in experimental mice. Discussion Nrf2 in macrophages plays a critical role in the immune response to and resolution of otitis media Restoration of Nrf2 expression in OME-macrophages could be a promising therapeutic approach to prevent the development of COM in AOM patients.
Collapse
|
16
|
Role of Zerumbone, a Phytochemical Sesquiterpenoid from Zingiber zerumbet Smith, in Maintaining Macrophage Polarization and Redox Homeostasis. Nutrients 2022; 14:nu14245402. [PMID: 36558562 PMCID: PMC9783216 DOI: 10.3390/nu14245402] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophages and microglia are highly versatile cells that can be polarized into M1 and M2 phenotypes in response to diverse environmental stimuli, thus exhibiting different biological functions. In the central nervous system, activated resident macrophages and microglial cells trigger the production of proinflammatory mediators that contribute to neurodegenerative diseases and psychiatric disorders. Therefore, modulating the activation of macrophages and microglia by optimizing the inflammatory environment is beneficial for disease management. Several naturally occurring compounds have been reported to have anti-inflammatory and neuroprotective properties. Zerumbone is a phytochemical sesquiterpenoid and also a cyclic ketone isolated from Zingiber zerumbet Smith. In this study, we found that zerumbone effectively reduced the expression of lipocalin-2 in macrophages and microglial cell lines. Lipocalin-2, also known as neutrophil gelatinase-associated lipocalin (NGAL), has been characterized as an adipokine/cytokine implicated in inflammation. Moreover, supplement with zerumbone inhibited reactive oxygen species production. Phagocytic activity was decreased following the zerumbone supplement. In addition, the zerumbone supplement remarkably reduced the production of M1-polarization-associated chemokines CXC10 and CCL-2, as well as M1-polarization-associated cytokines interleukin (IL)-6, IL-1β, and tumor necrosis factor-α. Furthermore, the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 and the production of NO were attenuated in macrophages and microglial cells supplemented with zerumbone. Notably, we discovered that zerumbone effectively promoted the production of the endogenous antioxidants heme oxygenase-1, glutamate-cysteine ligase modifier subunit, glutamate-cysteine ligase catalytic subunit, and NAD(P)H quinone oxidoreductase-1 and remarkably enhanced IL-10, a marker of M2 macrophage polarization. Endogenous antioxidant production and M2 macrophage polarization were increased through activation of the AMPK/Akt and Akt/GSK3 signaling pathways. In summary, this study demonstrated the protective role of zerumbone in maintaining M1 and M2 polarization homeostasis by decreasing inflammatory responses and enhancing the production of endogenous antioxidants in both macrophages and microglia cells. This study suggests that zerumbone can be used as a potential therapeutic drug for the supplement of neuroinflammatory diseases.
Collapse
|
17
|
Dahdah A, Jaggers RM, Sreejit G, Johnson J, Kanuri B, Murphy AJ, Nagareddy PR. Immunological Insights into Cigarette Smoking-Induced Cardiovascular Disease Risk. Cells 2022; 11:3190. [PMID: 36291057 PMCID: PMC9600209 DOI: 10.3390/cells11203190] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 01/19/2023] Open
Abstract
Smoking is one of the most prominent addictions of the modern world, and one of the leading preventable causes of death worldwide. Although the number of tobacco smokers is believed to be at a historic low, electronic cigarette use has been on a dramatic rise over the past decades. Used as a replacement for cigarette smoking, electronic cigarettes were thought to reduce the negative effects of burning tobacco. Nonetheless, the delivery of nicotine by electronic cigarettes, the most prominent component of cigarette smoke (CS) is still delivering the same negative outcomes, albeit to a lesser extent than CS. Smoking has been shown to affect both the structural and functional aspects of major organs, including the lungs and vasculature. Although the deleterious effects of smoking on these organs individually is well-known, it is likely that the adverse effects of smoking on these organs will have long-lasting effects on the cardiovascular system. In addition, smoking has been shown to play an independent role in the homeostasis of the immune system, leading to major sequela. Both the adaptive and the innate immune system have been explored regarding CS and have been demonstrated to be altered in a way that promotes inflammatory signals, leading to an increase in autoimmune diseases, inflammatory diseases, and cancer. Although the mechanism of action of CS has not been fully understood, disease pathways have been explored in both branches of the immune system. The pathophysiologically altered immune system during smoking and its correlation with cardiovascular diseases is not fully understood. Here we highlight some of the important pathological mechanisms that involve cigarette smoking and its many components on cardiovascular disease and the immune systems in order to have a better understanding of the mechanisms at play.
Collapse
Affiliation(s)
- Albert Dahdah
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Robert M. Jaggers
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jillian Johnson
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Babunageswararao Kanuri
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Andrew J. Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC 3010, Australia
| | - Prabhakara R. Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Synthesis and Assessment of the In Vitro and Ex Vivo Activity of Salicylate Synthase (Mbti) Inhibitors as New Candidates for the Treatment of Mycobacterial Infections. Pharmaceuticals (Basel) 2022; 15:ph15080992. [PMID: 36015139 PMCID: PMC9413995 DOI: 10.3390/ph15080992] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Tuberculosis (TB) causes millions of deaths every year, ranking as one of the most dangerous infectious diseases worldwide. Because several pathogenic strains of M. tuberculosis (Mtb) have developed resistance against most of the established anti-TB drugs, new therapeutic options are urgently needed. An attractive target for the development of new anti-TB agents is the salicylate synthase MbtI, the first enzyme of the mycobacterial siderophore biochemical machinery, absent in human cells. In this work, a set of analogues of 5-(3-cyanophenyl)furan-2-carboxylic acid (I), the most potent MbtI inhibitor identified to date, was synthesized, characterized, and tested to further elucidate the structural requirements for achieving an efficient MbtI inhibition and potent antitubercular activity. The structure–activity relationships (SAR) discussed herein evidenced the importance of the side chain linked to the phenyl moiety to improve the in vitro antimycobacterial activity. In detail, 1f emerged as the most effective analogue against the pathogen, acting without cytotoxicity issues. To deepen the understanding of its mechanism of action, we established a fluorescence-based screening test to quantify the pathogen infectivity within host cells, using MPI-2 murine cells, a robust surrogate for alveolar macrophages. The set-up of the new assay demonstrates significant potential to accelerate the discovery of new anti-TB drugs.
Collapse
|
19
|
Yao X, Jin G, Liu D, Zhang X, Yang Y, chen Y, Duan Z, Bi Y, Yan F, Yang Y, Zhang H, Dong G, Li S, Cheng S, Tang H, Hong F, Si C. Inducible nitric oxide synthase regulates macrophage polarization via the MAPK signals in concanavalin A-induced hepatitis. Immun Inflamm Dis 2022; 10:e643. [PMID: 35759238 PMCID: PMC9168548 DOI: 10.1002/iid3.643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 02/06/2022] [Accepted: 05/09/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Acute liver inflammatory reactions contribute to many health problems; thus, it is critical to understand the underlying pathogenic mechanisms of acute hepatitis. In this study, an experimental in vivo model of concanavalin A (ConA)-induced hepatitis was used. MATERIALS AND METHODS C57BL/6 (wild-type, WT) or inducible nitric oxide synthase-deficient (iNOS-/- ) mice were injected with PBS or 15 mg/kg ConA via tail vein. Detection of liver injury by histological examination and apoptosis, and flow cytometry to detect the effect of immune cells on liver injury. RESULTS iNOS-/- mice had lower levels of the liver enzymes aspartate aminotransferase and alanine aminotransferase, suggesting that they were protected against ConA-induced pathological liver injury and that iNOS participated in the regulation of hepatitis. Furthermore, iNOS deficiency was found to lower CD86 expression and suppressed the messenger RNA levels of inflammatory factors in the liver. In vitro experiments also demonstrated that iNOS deficiency suppressed the sequential phosphorylation of the mitogen-activated protein kinase pathway cascade, thereby inhibiting the M1 polarization of macrophages and consequently suppressing the transcription of inflammation factors. CONCLUSION iNOS may contribute to ConA-induced inflammation by promoting the activation of proinflammatory macrophages.
Collapse
Affiliation(s)
- Xiaoying Yao
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
- Institute of Immune Precision Diagnosis and Therapy & Translational MedicineAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Guiyuan Jin
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
- Institute of Immune Precision Diagnosis and Therapy & Translational MedicineAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Dong Liu
- Department of Clinical LaboratoryAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Xiaobei Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Yonghong Yang
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Yu chen
- Fourth Liver Disease Center, Beijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Zhongping Duan
- Fourth Liver Disease Center, Beijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Yanzhen Bi
- Department of Infectious DiseaseQingdao Municipal HospitalQingdaoShandongChina
| | - Fenglian Yan
- Institute of Immunology and Molecular MedicineJining Medical UniversityJiningShandongChina
| | - Yanli Yang
- Institute of Immunology and Molecular MedicineJining Medical UniversityJiningShandongChina
| | - Hui Zhang
- Institute of Immunology and Molecular MedicineJining Medical UniversityJiningShandongChina
| | - Guanjun Dong
- Institute of Immunology and Molecular MedicineJining Medical UniversityJiningShandongChina
| | - Shanshan Li
- Fourth Liver Disease Center, Beijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Shumin Cheng
- Department of GastroenterologyPeople's Hospital of Jia XiangJiningShandongChina
| | - Huixin Tang
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Feng Hong
- Medical Research Center, Affiliated Hospital of Jining Medical UniversityJiningShandongChina
- Institute of Immune Precision Diagnosis and Therapy & Translational MedicineAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Chuanping Si
- Institute of Immune Precision Diagnosis and Therapy & Translational MedicineAffiliated Hospital of Jining Medical UniversityJiningShandongChina
- Institute of Immunology and Molecular MedicineJining Medical UniversityJiningShandongChina
| |
Collapse
|
20
|
Bai W, Shi Q, Wu J, Wang K, Chen Y, Ma X, Mao D. Chinese Herb Jiedu Huayu Granules Inhibiting Immune and Inflammatory Response of Rats with Acute Liver Failure by Regulating the NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4479885. [PMID: 35601154 PMCID: PMC9117047 DOI: 10.1155/2022/4479885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022]
Abstract
Objective To research the influence of Chinese medicine Jiedu Huayu granules (JDHY) on the immune response and inflammatory response of rats with acute liver failure (ALF) and investigate its related mechanism. Methods Rats were randomly divided into 4 groups: control group (n = 6) were injected with the same amount of normal saline; ALF group (n = 10) were injected intraperitoneally with D-GaIN (700 mg/kg) and LPS (10 μg/kg); ALF+JDHY group (n = 10) were given JDHY 57.55 g/kg/d by gavage for 7 days and injected intraperitoneally with D-GaIN/LPS after the last dose; and ALF+BAY group (n = 10) were given BAY 10 mg/kg/d by gavage for 7 days and injected intraperitoneally with D-GaIN/LPS after the last dose. Changes in liver function and coagulation function were examined in rat serum; the pathological varieties of liver tissues were verified by HE staining; immunohistochemistry was utilized to determine the ratio of PCNA and F4/80 in liver tissues; the flow cytometry was applied to determine the ratio of CD4+/CD8+ cells in peripheral blood mononuclear cells (PBMCs); ELISA and qRT-PCR were utilized to check the level of IL-10, IL-6, IL-13, IL-1β, TNF-α, IFN-γ, and CD163 in serum and liver cells. Western blot was adopted to check the expression of apoptotic protein and expression and NF-κB pathway-related protein expression. Results JDHY and BAY could decline the expression of AST, ALT, ALP, and TBiL in ALF rat serum significantly (P < 0.01), increase PTA and PLT (P < 0.01), and mitigate liver tissue damage. Besides, JDHY and BAY could reduce the apoptosis and improve the proliferation of the liver cells in rats with ALF; meanwhile, the ratio of CD4+ cells and F4/80 cells was reduced while CD8+ cells were increased (P < 0.01). Further, JDHY and BAY could reduce the level of IFN-γ, IL-6, IL-1β, and TNF-α while increasing the level of IL-10 and IL-13 (P < 0.01). Additionally, the expression of sCD163 in serum and CD163 expression in liver tissues increased (P < 0.01). The result of western blot confirmed that JDHY could inhibit the phosphorylated expression of NF-κB, IKβα, and IKKβ in the ALF rat tissues. Conclusions JDHY can upregulate the level of CD163/sCD163 by the NF-κB signaling pathway, thereby regulating immune response, inhibiting inflammatory response, and ultimately improving ALF in the rats.
Collapse
Affiliation(s)
- Wenjie Bai
- Foreign Language Department, Guangxi University of Chinese Medicine, Nanning, China
| | - Qinglan Shi
- Department of Liver Diseases, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Jinyu Wu
- Department of Liver Diseases, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Kejing Wang
- Hepatobiliary Surgical Department, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Yueqiao Chen
- Department of Liver Diseases, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaocong Ma
- Foreign Language Department, Guangxi University of Chinese Medicine, Nanning, China
| | - Dewen Mao
- Department of Liver Diseases, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
21
|
Xian X, Cai LL, Li Y, Wang RC, Xu YH, Chen YJ, Xie YH, Zhu XL, Li YF. Neuron secrete exosomes containing miR-9-5p to promote polarization of M1 microglia in depression. J Nanobiotechnology 2022; 20:122. [PMID: 35264203 PMCID: PMC8905830 DOI: 10.1186/s12951-022-01332-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Background Neuroinflammation is an important component mechanism in the development of depression. Exosomal transfer of MDD-associated microRNAs (miRNAs) from neurons to microglia might exacerbate neuronal cell inflammatory injury. Results By sequence identification, we found significantly higher miR-9-5p expression levels in serum exosomes from MDD patients than healthy control (HC) subjects. Then, in cultured cell model, we observed that BV2 microglial cells internalized PC12 neuron cell-derived exosomes while successfully transferring miR-9-5p. MiR-9-5p promoted M1 polarization in microglia and led to over releasing of proinflammatory cytokines, such as interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), which exacerbated neurological damage. Furthermore, we identified suppressor of cytokine signaling 2 (SOCS2) as a direct target of miR-9-5p. Overexpression of miR-9-5p suppressed SOCS2 expression and reactivated SOCS2-repressed Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathways. Consistently, we confirmed that adeno-associated virus (AAV)-mediated overexpression of miR-9-5p polarized microglia toward the M1 phenotype and exacerbated depressive symptoms in chronic unpredictable mild stress (CUMS) mouse mode. Conclusion MiR-9-5p was transferred from neurons to microglia in an exosomal way, leading to M1 polarization of microglia and further neuronal injury. The expression and secretion of miR-9-5p might be novel therapeutic targets for MDD. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01332-w.
Collapse
Affiliation(s)
- Xian Xian
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Li-Li Cai
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Yang Li
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Ran-Chao Wang
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Yu-Hao Xu
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Ya-Jie Chen
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Yu-Hang Xie
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China
| | - Xiao-Lan Zhu
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang, 212001, Jiangsu, China.
| | - Yue-Feng Li
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, 212001, Jiangsu, China. .,Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
22
|
Vafaee T, Walker F, Thomas D, Roderjan JG, Veiga Lopes S, da Costa FDA, Desai A, Rooney P, Jennings LM, Fisher J, Berry HE, Ingham E. Repopulation of decellularised porcine pulmonary valves in the right ventricular outflow tract of sheep: Role of macrophages. J Tissue Eng 2022; 13:20417314221102680. [PMID: 35782993 PMCID: PMC9243591 DOI: 10.1177/20417314221102680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
The primary objective was to evaluate performance of low concentration SDS decellularised porcine pulmonary roots in the right ventricular outflow tract of juvenile sheep. Secondary objectives were to explore the cellular population of the roots over time. Animals were monitored by echocardiography and roots explanted at 1, 3, 6 (n = 4) and 12 months (n = 8) for gross analysis. Explanted roots were subject to histological, immunohistochemical and quantitative calcium analysis (n = 4 at 1, 3 and 12 months) and determination of material properties (n = 4; 12 months). Cryopreserved ovine pulmonary root allografts (n = 4) implanted for 12 months, and non-implanted cellular ovine roots were analysed for comparative purposes. Decellularised porcine pulmonary roots functioned well and were in very good condition with soft, thin and pliable leaflets. Morphometric analysis showed cellular population by 1 month. However, by 12 months the total number of cells was less than 50% of the total cells in non-implanted native ovine roots. Repopulation of the decellularised porcine tissues with stromal (α-SMA+; vimentin+) and progenitor cells (CD34+; CD271+) appeared to be orchestrated by macrophages (MAC 387+/ CD163low and CD163+/MAC 387-). The calcium content of the decellularised porcine pulmonary root tissues increased over the 12-month period but remained low (except suture points) at 401 ppm (wet weight) or below. The material properties of the decellularised porcine pulmonary root wall were unchanged compared to pre-implantation. There were some changes in the leaflets but importantly, the porcine tissues did not become stiffer. The decellularised porcine pulmonary roots showed good functional performance in vivo and were repopulated with ovine cells of the appropriate phenotype in a process orchestrated by M2 macrophages, highlighting the importance of these cells in the constructive tissue remodelling of cardiac root tissues.
Collapse
Affiliation(s)
- Tayyebeh Vafaee
- Institute of Medical and Biological
Engineering, School of Biomedical Sciences, Faculty of Biological Sciences,
University of Leeds, Leeds, UK
| | - Fiona Walker
- Institute of Medical and Biological
Engineering, School of Biomedical Sciences, Faculty of Biological Sciences,
University of Leeds, Leeds, UK
| | - Dan Thomas
- Institute of Medical and Biological
Engineering, School of Biomedical Sciences, Faculty of Biological Sciences,
University of Leeds, Leeds, UK
| | - João Gabriel Roderjan
- Department of Cardiac Surgery, Santa
Casa de Curitiba, Pontifica Universidade Catolica do Parana, Curitiba, Brazil
| | - Sergio Veiga Lopes
- Department of Cardiac Surgery, Santa
Casa de Curitiba, Pontifica Universidade Catolica do Parana, Curitiba, Brazil
| | - Francisco DA da Costa
- Department of Cardiac Surgery, Santa
Casa de Curitiba, Pontifica Universidade Catolica do Parana, Curitiba, Brazil
| | - Amisha Desai
- Institute of Medical and Biological
Engineering, School of Mechanical Engineering, University of Leeds, Leeds, UK
| | - Paul Rooney
- NHS Blood and Transplant, Tissue and
Eye Services, Estuary Banks, Liverpool, UK
| | - Louise M Jennings
- Institute of Medical and Biological
Engineering, School of Mechanical Engineering, University of Leeds, Leeds, UK
| | - John Fisher
- Institute of Medical and Biological
Engineering, School of Mechanical Engineering, University of Leeds, Leeds, UK
| | - Helen E Berry
- Institute of Medical and Biological
Engineering, School of Biomedical Sciences, Faculty of Biological Sciences,
University of Leeds, Leeds, UK
| | - Eileen Ingham
- Institute of Medical and Biological
Engineering, School of Biomedical Sciences, Faculty of Biological Sciences,
University of Leeds, Leeds, UK
| |
Collapse
|
23
|
Xu Y, Yan H, Zhang X, Zhuo J, Han Y, Zhang H, Xie D, Lan X, Cai W, Wang X, Wang S, Li X. Roles of Altered Macrophages and Cytokines: Implications for Pathological Mechanisms of Postmenopausal Osteoporosis, Rheumatoid Arthritis, and Alzheimer's Disease. Front Endocrinol (Lausanne) 2022; 13:876269. [PMID: 35757427 PMCID: PMC9226340 DOI: 10.3389/fendo.2022.876269] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Postmenopausal osteoporosis (PMOP) is characterized by the uncoupling of bone resorption and bone formation induced by estrogen deficiency, which is a complex outcome related to estrogen and the immune system. The interaction between bone and immune cells is regarded as the context of PMOP. Macrophages act differently on bone cells, depending on their polarization profile and secreted paracrine factors, which may have implications for the development of PMOP. PMOP, rheumatoid arthritis (RA), and Alzheimer's disease (AD) might have pathophysiological links, and the similarity of their pathological mechanisms is partially visible in altered macrophages and cytokines in the immune system. This review focuses on exploring the pathological mechanisms of PMOP, RA, and AD through the roles of altered macrophages and cytokines secretion. First, the multiple effects on cytokines secretion by bone-bone marrow (BM) macrophages in the pathological mechanism of PMOP are reviewed. Then, based on the thought of "different tissue-same cell type-common pathological molecules-disease pathological links-drug targets" and the methodologies of "molecular network" in bioinformatics, highlight that multiple cytokines overlap in the pathological molecules associated with PMOP vs. RA and PMOP vs. AD, and propose that these overlaps may lead to a pathological synergy in PMOP, RA, and AD. It provides a novel strategy for understanding the pathogenesis of PMOP and potential drug targets for the treatment of PMOP.
Collapse
Affiliation(s)
- Yunteng Xu
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hui Yan
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Basic Discipline Laboratory of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xin Zhang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Junkuan Zhuo
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yidan Han
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haifeng Zhang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dingbang Xie
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xin Lan
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wanping Cai
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaoning Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shanshan Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xihai Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Fujian University of Traditional Chinese Medicine, Fuzhou, China
- *Correspondence: Xihai Li,
| |
Collapse
|
24
|
Li N, Guo X, Li R, Zhou J, Yu F, Yan X. p-Coumaric acid regulates macrophage polarization in myocardial ischemia/reperfusion by promoting the expression of indoleamine 2, 3-dioxygenase. Bioengineered 2021; 12:10971-10981. [PMID: 34738873 PMCID: PMC8810143 DOI: 10.1080/21655979.2021.2001924] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophage infiltration is a hallmark pathological change observed in early stage myocardial ischemia/reperfusion (MI/R) injury and one of the main causes of myocardial damage. Here, we investigated the effects of p-Coumaric acid (p-CA) on macrophage polarization following MI/R injury and its mechanisms. In vitro, p-CA decreases the expression of LPS/IFN-γ-induced M1 macrophage markers (TNF-α, IL-6, iNOS and CCL2) and increases IL-4-induced M2 macrophage markers (IL-10, CD206, Arg1 and Mrc) in mouse bone marrow-derived macrophages (BMDMs). Additionally, p-CA elevated indoleamine 2, 3-dioxygenase (IDO) protein expression levels, M2 macrophage polarization and M2 macrophage markers through IL-4. In contrast, repression of IDO attenuated p-CA functions regulating BMDMs through IL-4. In vivo, IDO expression was downregulated in mouse hearts subjected to MI/R injury. Treatment of p-CA increased IDO expression in the hearts of MI/R mice. Functionally, p-CA decreases M1 macrophage markers, the number of M1 macrophages and inflammation around heart tissue following MI/R injury. Importantly, p-CA reduces cardiomyocyte apoptosis caused by MI/R. Altogether, our study identified that p-CA modulates macrophage polarization by promoting IDO expression and that p-CA attenuates macrophage-mediated inflammation following MI/R by promoting M2 macrophage polarization through IDO.
Collapse
Affiliation(s)
- Na Li
- Department of Cardiology, Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, China
| | - Xueyuan Guo
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases
| | - Rui Li
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China
| | - Jian Zhou
- Department of Cardiology, Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, China
| | - Fangfang Yu
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University
| | - Xianliang Yan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases
| |
Collapse
|
25
|
Estrada McDermott J, Pezzanite L, Goodrich L, Santangelo K, Chow L, Dow S, Wheat W. Role of Innate Immunity in Initiation and Progression of Osteoarthritis, with Emphasis on Horses. Animals (Basel) 2021; 11:3247. [PMID: 34827979 PMCID: PMC8614551 DOI: 10.3390/ani11113247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common condition with diverse etiologies, affecting horses, humans, and companion animals. Importantly, OA is not a single disease, but rather a disease process initiated by different events, including acute trauma, irregular or repetitive overload of articular structures, and spontaneous development with aging. Our understanding of the pathogenesis of OA is still evolving, and OA is increasingly considered a multifactorial disease in which the innate immune system plays a key role in regulating and perpetuating low-grade inflammation, resulting in sustained cartilage injury and destruction. Macrophages within the synovium and synovial fluid are considered the key regulators of immune processes in OA and are capable of both stimulating and suppressing joint inflammation, by responding to local and systemic cues. The purpose of this review is to examine the role of the innate immune system in the overall pathogenesis of OA, drawing on insights from studies in humans, animal models of OA, and from clinical and research studies in horses. This review also discusses the various therapeutic immune modulatory options currently available for managing OA and their mechanisms of action.
Collapse
Affiliation(s)
- Juan Estrada McDermott
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
| | - Lynn Pezzanite
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
| | - Laurie Goodrich
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
| | - Kelly Santangelo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - Lyndah Chow
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
| | - Steven Dow
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - William Wheat
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (J.E.M.); (L.P.); (L.G.); (L.C.); (S.D.)
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| |
Collapse
|
26
|
Xie C, Wan L, Li C, Feng Y, Kang YJ. Selective suppression of M1 macrophages is involved in zinc inhibition of liver fibrosis in mice. J Nutr Biochem 2021; 97:108802. [PMID: 34119631 DOI: 10.1016/j.jnutbio.2021.108802] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/23/2021] [Accepted: 05/31/2021] [Indexed: 02/08/2023]
Abstract
Zinc deficiency is common in the liver of patients with chronic liver disease. Zinc supplementation suppresses the progression of liver fibrosis induced by bile duct ligation (BDL) in mice. The present study was undertaken to specifically investigate a possible mechanism by which zinc plays this role in the liver. Kunming mice were subjected to BDL for 4 weeks to induce liver fibrosis, and concomitantly treated with zinc sulfite or saline as control by gavage once a day. The results showed that zinc supplementation significantly suppressed liver fibrosis and inflammation along with inhibition of hepatic stellate cells activation induced by BDL. These inhibitory effects were accompanied by the reduction of collagen deposition and a significant reduction of macrophage infiltration affected livers. Importantly, zinc selectively inhibited M1 macrophage polarization and M1-related inflammatory cytokines. This inhibitory effect was further confirmed by the reduction of relevant biomarkers of M1 macrophages including inducible NO synthase (iNOS), monocyte chemotactic protein-1 (MCP-1/CCL2), and tumor necrosis factor-α in the zinc supplemented BDL livers. In addition, zinc inhibition of M1 macrophages was associated with a decrease of Notch1 expression. Taken together, these data indicated that zinc supplementation inhibited liver inflammation and fibrosis in BDL mice through selective suppression of M1 macrophages, which is associated with inhibition of Notch1 pathway in M1 macrophage polarization.
Collapse
Affiliation(s)
- Chengxia Xie
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinrui Feng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China; Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
27
|
De Muynck K, Vanderborght B, Van Vlierberghe H, Devisscher L. The Gut-Liver Axis in Chronic Liver Disease: A Macrophage Perspective. Cells 2021; 10:2959. [PMID: 34831182 PMCID: PMC8616442 DOI: 10.3390/cells10112959] [Citation(s) in RCA: 15] [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: 10/05/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic liver disease (CLD) is a growing health concern which accounts for two million deaths per year. Obesity, alcohol overconsumption, and progressive cholestasis are commonly characterized by persistent low-grade inflammation and advancing fibrosis, which form the basis for development of end-stage liver disease complications, including hepatocellular carcinoma. CLD pathophysiology extends to the intestinal tract and is characterized by intestinal dysbiosis, bile acid dysregulation, and gut barrier disruption. In addition, macrophages are key players in CLD progression and intestinal barrier breakdown. Emerging studies are unveiling macrophage heterogeneity and driving factors of their plasticity in health and disease. To date, in-depth investigation of how gut-liver axis disruption impacts the hepatic and intestinal macrophage pool in CLD pathogenesis is scarce. In this review, we give an overview of the role of intestinal and hepatic macrophages in homeostasis and gut-liver axis disruption in progressive stages of CLD.
Collapse
Affiliation(s)
- Kevin De Muynck
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
| |
Collapse
|
28
|
Junior, Lai YS, Nguyen HT, Salmanida FP, Chang KT. MERTK +/hi M2c Macrophages Induced by Baicalin Alleviate Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:10604. [PMID: 34638941 PMCID: PMC8508959 DOI: 10.3390/ijms221910604] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/19/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide. An accumulation of fat, followed by inflammation, is the major cause of NAFLD progression. During inflammation, macrophages are the most abundant immune cells recruited to the site of injury. Macrophages are classified into "proinflammatory" M1 macrophages, and "anti-inflammatory" M2 macrophages. In NAFLD, M1 macrophages are the most prominent macrophages that lead to an excessive inflammatory response. Previously, we found that baicalin could polarize macrophages into anti-inflammatory M2c subtype macrophages with an increased level of MERTK expression. Several studies have also shown a strong correlation between MERTK expression and cholesterol efflux, efferocytosis, as well as phagocytosis capability. Therefore, in this study, we aim to elucidate the potential and efficacy of mononuclear-cell (MNC)-derived MERTK+/hi M2c macrophages induced by baicalin as a cell-based therapy for NAFLD treatment. In our results, we have demonstrated that a MERTK+/hi M2c macrophage injection to NAFLD mice contributes to an increased level of serum HDL secretion in the liver, a decline in the circulating CD4+CD25- and CD8+CD25- T cells and lowers the total NAFLD pathological score by lessening the inflammation, necrosis, and fibrosis. In the liver, profibrotic COL1A1 and FN, proinflammation TNFα, as well as the regulator of lipid metabolism PPARɣ expression, were also downregulated after injection. In parallel, the transcriptomic profiles of the injected MERTK+/hi M2c macrophages showed that the various genes directly or indirectly involved in NAFLD progression (e.g., SERPINE1, FADS2) were also suppressed. Downregulation of cytokines and inflammation-associated genes, such as CCR5, may promote a pro-resolving milieu in the NAFLD liver. Altogether, cell-based therapy using MERTK+/hi M2c macrophages is promising, as it ameliorates NAFLD in mice.
Collapse
Affiliation(s)
- Junior
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (J.); (H.T.N.); (F.P.S.)
| | - Yin-Siew Lai
- Research Center for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan;
| | - Huyen Thi Nguyen
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (J.); (H.T.N.); (F.P.S.)
| | - Farrah P. Salmanida
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (J.); (H.T.N.); (F.P.S.)
| | - Ko-Tung Chang
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; (J.); (H.T.N.); (F.P.S.)
- Research Center for Animal Biologics, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan;
- Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| |
Collapse
|
29
|
Debuque RJ, Hart AJ, Johnson GH, Rosenthal NA, Godwin JW. Identification of the Adult Hematopoietic Liver as the Primary Reservoir for the Recruitment of Pro-regenerative Macrophages Required for Salamander Limb Regeneration. Front Cell Dev Biol 2021; 9:750587. [PMID: 34568347 PMCID: PMC8456783 DOI: 10.3389/fcell.2021.750587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/02/2021] [Indexed: 12/30/2022] Open
Abstract
The lack of scar-free healing and regeneration in many adult human tissues imposes severe limitations on the recovery of function after injury. In stark contrast, salamanders can functionally repair a range of clinically relevant tissues throughout adult life. The impressive ability to regenerate whole limbs after amputation, or regenerate following cardiac injury, is critically dependent on the recruitment of (myeloid) macrophage white blood cells to the site of injury. Amputation in the absence of macrophages results in regeneration failure and scar tissue induction. Identifying the exact hematopoietic source or reservoir of myeloid cells supporting regeneration is a necessary step in characterizing differences in macrophage phenotypes regulating scarring or regeneration across species. Mammalian wounds are dominated by splenic-derived monocytes that originate in the bone marrow and differentiate into macrophages within the wound. Unlike mammals, adult axolotls do not have functional bone marrow but instead utilize liver and spleen tissues as major sites for adult hematopoiesis. To interrogate leukocyte identity, tissue origins, and modes of recruitment, we established several transgenic axolotl hematopoietic tissue transplant models and flow cytometry protocols to study cell migration and identify the source of pro-regenerative macrophages. We identified that although bidirectional trafficking of leukocytes can occur between spleen and liver tissues, the liver is the major source of leukocytes recruited to regenerating limbs. Recruitment of leukocytes and limb regeneration occurs in the absence of the spleen, thus confirming the dependence of liver-derived myeloid cells in regeneration and that splenic maturation is dispensable for the education of pro-regenerative macrophages. This work provides an important foundation for understanding the hematopoietic origins and education of myeloid cells recruited to, and essential for, adult tissue regeneration.
Collapse
Affiliation(s)
- Ryan J Debuque
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Andrew J Hart
- The MDI Biological Laboratory (MDIBL), Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME, United States
| | - Gabriela H Johnson
- The MDI Biological Laboratory (MDIBL), Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME, United States
| | - Nadia A Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.,The Jackson Laboratory, Bar Harbor, ME, United States
| | - James W Godwin
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.,The MDI Biological Laboratory (MDIBL), Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME, United States.,The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
30
|
Aschbacher K, Hagan M, Steine IM, Rivera L, Cole S, Baccarella A, Epel ES, Lieberman A, Bush NR. Adversity in early life and pregnancy are immunologically distinct from total life adversity: macrophage-associated phenotypes in women exposed to interpersonal violence. Transl Psychiatry 2021; 11:391. [PMID: 34282132 PMCID: PMC8289995 DOI: 10.1038/s41398-021-01498-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Early childhood and pregnancy are two sensitive periods of heightened immune plasticity, when exposure to adversity may disproportionately increase health risks. However, we need deeper phenotyping to disentangle the impact of adversity during sensitive periods from that across the total lifespan. This study examined whether retrospective reports of adversity during childhood or pregnancy were associated with inflammatory imbalance, in an ethnically diverse cohort of 53 low-income women seeking family-based trauma treatment following exposure to interpersonal violence. Structured interviews assessed early life adversity (trauma exposure ≤ age 5), pregnancy adversity, and total lifetime adversity. Blood serum was assayed for pro-inflammatory (TNF-a, IL-1ß, IL-6, and CRP) and anti-inflammatory (IL-1RA, IL-4, and IL-10) cytokines. CD14+ monocytes were isolated in a subsample (n = 42) and gene expression assayed by RNA sequencing (Illumina HiSeq 4000; TruSeq cDNA library). The primary outcome was a macrophage-associated M1/M2 gene expression phenotype. To evaluate sensitivity and specificity, we contrasted M1/M2 gene expression with a second, clinically-validated macrophage-associated immunosuppressive phenotype (endotoxin tolerance) and with pro-inflammatory and anti-inflammatory cytokine levels. Adjusting for demographics, socioeconomic status, and psychopathology, higher adversity in early life (ß = .337, p = 0.029) and pregnancy (ß = .332, p = 0.032) were each associated with higher M1/M2 gene expression, whereas higher lifetime adversity (ß = -.341, p = 0.031) was associated with lower immunosuppressive gene expression. Adversity during sensitive periods was uniquely associated with M1/M2 imbalance, among low-income women with interpersonal violence exposure. Given that M1/M2 imbalance is found in sepsis, severe COVID-19 and myriad chronic diseases, these findings implicate novel immune mechanisms underlying the impact of adversity on health.
Collapse
Affiliation(s)
- Kirstin Aschbacher
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, USA.
- Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, USA.
- The Institute for Integrative Health, San Francisco, USA.
| | - Melissa Hagan
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, USA
- Department of Psychology, College of Science & Engineering, San Francisco State University, San Francisco, USA
| | - Iris M Steine
- Department of Psychology, University of California, Berkeley, USA
- Department of Psychosocial Science, University of Bergen, Bergen, Norway
| | - Luisa Rivera
- Department of Anthropology, Emory University, Atlanta, Georgia
| | - Steve Cole
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, USA
| | | | - Elissa S Epel
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Alicia Lieberman
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Nicole R Bush
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, USA
- Center for Health and Community, University of California, San Francisco, USA
- Department of Pediatrics, Division of Developmental Medicine, University of California, San Francisco, USA
| |
Collapse
|
31
|
Santamarina AB, Pisani LP, Baker EJ, Marat AD, Valenzuela CA, Miles EA, Calder PC. Anti-inflammatory effects of oleic acid and the anthocyanin keracyanin alone and in combination: effects on monocyte and macrophage responses and the NF-κB pathway. Food Funct 2021; 12:7909-7922. [PMID: 34250536 DOI: 10.1039/d1fo01304a] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Monocyte recruitment and activation of macrophages are essential for homeostasis but are also related to the development and progression of cardiometabolic diseases. The management of inflammation with dietary components has been widely investigated. Two components that may influence inflammation are unsaturated fatty acids such as oleic acid (OA; 18:1cis-9) and antioxidant compounds like anthocyanins. Molecular and metabolic effects of such bioactive compounds are usually investigated in isolation, whereas they may be present in combination in foods or the diet. Considering this, we aimed to analyze the effects of OA and the anthocyanin keracyanin (AC) alone and in combination on toll-like receptor-mediated inflammatory responses in monocytes and macrophages. For this, THP-1-derived macrophages and monocytes were exposed to 3 treatments: OA, AC, or the combination (OAAC) and then stimulated with lipopolysaccharide. Inflammation-related gene expression and protein concentrations of IL-1β, TNF-α, IL-6, MCP-1, and IL-10 were assessed. Also, NFκBp65, IκBα, and PPAR-γ protein expression were determined. OA, AC, and OAAC decreased pNFκBp65, PPARγ, IκBα, TNF-α, IL-1β, IL-6, and MCP-1 and increased IL-10. MCP-1 protein expression was lower with OAAC than with either OA and AC alone. Compared to control, OAAC decreased mRNA for TLR4, IκKα, IκBα, NFκB1, MCP-1, TNF-α, IL-6, and IL-1β more than OA or AC did alone. Also, IL-10 mRNA was increased by OAAC compared with control, OA, and AC. In summary, OA and AC have anti-inflammatory effects individually but their combination (OAAC) exerts a greater effect.
Collapse
Affiliation(s)
- Aline B Santamarina
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, 11015-020, Brazil
| | | | | | | | | | | | | |
Collapse
|
32
|
Vallejo AN, Mroczkowski HJ, Michel JJ, Woolford M, Blair HC, Griffin P, McCracken E, Mihalik SJ, Reyes‐Mugica M, Vockley J. Pervasive inflammatory activation in patients with deficiency in very-long-chain acyl-coA dehydrogenase (VLCADD). Clin Transl Immunology 2021; 10:e1304. [PMID: 34194748 PMCID: PMC8236555 DOI: 10.1002/cti2.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 05/06/2021] [Accepted: 06/03/2021] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVES Very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a disorder of fatty acid oxidation. Symptoms are managed by dietary supplementation with medium-chain fatty acids that bypass the metabolic block. However, patients remain vulnerable to hospitalisations because of rhabdomyolysis, suggesting pathologic processes other than energy deficit. Since rhabdomyolysis is a self-destructive process that can signal inflammatory/immune cascades, we tested the hypothesis that inflammation is a physiologic dimension of VLCADD. METHODS All subjects (n = 18) underwent informed consent/assent. Plasma cytokine and cytometry analyses were performed. A prospective case analysis was carried out on a patient with recurrent hospitalisation. Health data were extracted from patient medical records. RESULTS Patients showed systemic upregulation of nine inflammatory mediators during symptomatic and asymptomatic periods. There was also overall abundance of immune cells with high intracellular expression of IFNγ, IL-6, MIP-1β (CCL4) and TNFα, and the transcription factors p65-NFκB and STAT1 linked to inflammatory pathways. A case analysis of a patient exhibited already elevated plasma cytokine levels during diagnosis in early infancy, evolving into sustained high systemic levels during recurrent rhabdomyolysis-related hospitalisations. There were corresponding activated leukocytes, with higher intracellular stores of inflammatory molecules in monocytes compared to T cells. Exposure of monocytes to long-chain free fatty acids recapitulated the cytokine signature of patients. CONCLUSION Pervasive plasma cytokine upregulation and pre-activated immune cells indicate chronic inflammatory state in VLCADD. Thus, there is rationale for practical implementation of clinical assessment of inflammation and/or translational testing, or adoption, of anti-inflammatory intervention(s) for personalised disease management.
Collapse
Affiliation(s)
- Abbe N Vallejo
- Division of Pediatric Rheumatology, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Department of ImmunologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Children's Hospital of PittsburghUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Henry J Mroczkowski
- Children's Hospital of PittsburghUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of Genetic and Genomic Medicine, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Present address:
Department of PediatricsUniversity of Tennessee Health Sciences CenterMemphisTNUSA
| | - Joshua J Michel
- Division of Pediatric Rheumatology, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Michael Woolford
- Division of Pediatric Rheumatology, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Harry C Blair
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Pittsburgh Veterans Administration Medical CenterPittsburghPAUSA
| | - Patricia Griffin
- Division of Pediatric Rheumatology, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Elizabeth McCracken
- Children's Hospital of PittsburghUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of Genetic and Genomic Medicine, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Center for Rare Disease and TherapyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Stephanie J Mihalik
- Division of Genetic and Genomic Medicine, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Miguel Reyes‐Mugica
- Children's Hospital of PittsburghUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Jerry Vockley
- Children's Hospital of PittsburghUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of Genetic and Genomic Medicine, Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Center for Rare Disease and TherapyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Department of Human GeneticsUniversity of Pittsburgh Graduate School of Public HealthPittsburghPAUSA
| |
Collapse
|
33
|
Albarrak SM. Antioxidant and immune responses of broiler chickens supplemented with Rhazya stricta extract in drinking water. Vet World 2021; 14:1437-1449. [PMID: 34316190 PMCID: PMC8304433 DOI: 10.14202/vetworld.2021.1437-1449] [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: 01/02/2021] [Accepted: 04/19/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Aim Rhazya stricta is a herbal plant widely used in traditional medicine due to its proficiency and naturalness with few side effects. In this study, we investigated the impact of using an R. stricta extract supplement on broiler chickens' performance, especially the immune system. Materials and Methods In addition to the control group, one group received the methanol extract of R. stricta in drinking water for the first 2 weeks before being challenged with sheep erythrocytes (SRBCs), while the other group was challenged with SRBCs without receiving the R. stricta treatment. We evaluated cellular immunity by determining the phagocytic activity and lymphocyte (L) proliferation and assessed humoral immunity by quantification of the serum total IgM and IgG. We measured the serum levels of antioxidant enzymes and performed a histological examination of the spleen and the bursa of Fabricius (BF). Results Our results indicate a significant enhancement in cellular immunity in the group supplemented with R. stricta as demonstrated by a significant increase in the phagocytic activity, L proliferation, and percentages of circulating L (p<0.05). The chickens treated with R. stricta exhibit an enhanced humoral response shown by a significant elevation in the serum levels of the total antibodies of the IgM and IgG isotypes, along with a notable increase in BF activity. Furthermore, R. stricta supplementation is associated with a significant increase in the serum levels of catalase and superoxide dismutase (p<0.05), along with a significant improvement in broilers' general performance, body weight, and feed efficiency. Conclusion Our results suggest an immunomodulatory effect for the methanol extract of R. stricta and highlight the potential use of this plant in preventive and therapeutic medicine.
Collapse
Affiliation(s)
- Saleh M Albarrak
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
34
|
Hu Q, Lyon CJ, Fletcher JK, Tang W, Wan M, Hu TY. Extracellular vesicle activities regulating macrophage- and tissue-mediated injury and repair responses. Acta Pharm Sin B 2021; 11:1493-1512. [PMID: 34221864 PMCID: PMC8245807 DOI: 10.1016/j.apsb.2020.12.014] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Abstract
Macrophages are typically identified as classically activated (M1) macrophages and alternatively activated (M2) macrophages, which respectively exhibit pro- and anti-inflammatory phenotypes, and the balance between these two subtypes plays a critical role in the regulation of tissue inflammation, injury, and repair processes. Recent studies indicate that tissue cells and macrophages interact via the release of small extracellular vesicles (EVs) in processes where EVs released by stressed tissue cells can promote the activation and polarization of adjacent macrophages which can in turn release EVs and factors that can promote cell stress and tissue inflammation and injury, and vice versa. This review discusses the roles of such EVs in regulating such interactions to influence tissue inflammation and injury in a number of acute and chronic inflammatory disease conditions, and the potential applications, advantage and concerns for using EV-based therapeutic approaches to treat such conditions, including their potential role of drug carriers for the treatment of infectious diseases.
Collapse
Key Words
- ADSCs, adipose-derived stem cells
- AKI, acute kidney injury
- ALI, acute lung injury
- AMs, alveolar macrophages
- BMSCs, bone marrow stromal cells
- CLP, cecal ligation and puncture
- DSS, dextran sodium sulphate
- EVs, extracellular vesicles
- Extracellular vesicles
- HSPA12B, heat shock protein A12B
- HUCMSCs, human umbilical cord mesenchymal stem cells
- IBD, inflammatory bowel disease
- ICAM-1, intercellular adhesion molecule 1
- IL-1β, interleukin-1β
- Inflammatory disease
- Interaction loop
- KCs, Kupffer cells
- KLF4, krüppel-like factor 4
- LPS, lipopolysaccharides
- MHC, major histocompatibility complex
- MSCs, mesenchymal stromal cells
- MVs, microvesicles
- Macrophage
- PEG, polyethylene glycol
- PMFA, 5,7,30,40,50-pentamethoxyflavanone
- PPARγ, peroxisome proliferator-activated receptor γ
- SIRPα, signal regulatory protein α
- Sepsis
- Stem cell
- TECs, tubular epithelial cells
- TNF, tumor necrosis factor
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
- Targeted therapy
- Tissue injury
- iNOS, inducible nitrogen oxide synthase
Collapse
|
35
|
Peng L, Zhang Y, Wang Z. Immune Responses against Disseminated Tumor Cells. Cancers (Basel) 2021; 13:2515. [PMID: 34063848 PMCID: PMC8196619 DOI: 10.3390/cancers13112515] [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: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Most cancer-related deaths are a consequence of metastases, a series of linear events, notably the invasion-metastasis cascade. The current understanding of cancer immune surveillance derives from studies in primary tumors, but disseminated cancer cells acquire mutations and, in some cases, appear to progress independently after spreading from primary sites. An early step in this process is micrometastatic dissemination. As such, the equilibrium between the immune system and disseminated cancer cells controls the fate of the cancer. Immune checkpoint inhibitors (ICIs) exhibit significant clinical activity in patients, but the efficacy of ICIs depends on both the tumor and its microenvironment. Data often suggest that disseminated cancer cells are not adequately targeted by the immune system. In this review, we summarize the main basic findings of immune responses against disseminated tumor cells and their organ-specific characteristics. Such studies may provide new directions for cancer immune therapy.
Collapse
Affiliation(s)
- Ling Peng
- Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Yongchang Zhang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha 410013, China;
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
36
|
Abstract
Liver metastases are commonly detected in a range of malignancies including colorectal cancer (CRC), pancreatic cancer, melanoma, lung cancer and breast cancer, although CRC is the most common primary cancer that metastasizes to the liver. Interactions between tumour cells and the tumour microenvironment play an important part in the engraftment, survival and progression of the metastases. Various cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, parenchymal hepatocytes, dendritic cells, resident natural killer cells as well as other immune cells such as monocytes, macrophages and neutrophils are implicated in promoting and sustaining metastases in the liver. Four key phases (microvascular, pre-angiogenic, angiogenic and growth phases) have been identified in the process of liver metastasis. Imaging modalities such as ultrasonography, CT, MRI and PET scans are typically used for the diagnosis of liver metastases. Surgical resection remains the main potentially curative treatment among patients with resectable liver metastases. The role of liver transplantation in the management of liver metastasis remains controversial. Systemic therapies, newer biologic agents (for example, bevacizumab and cetuximab) and immunotherapeutic agents have revolutionized the treatment options for liver metastases. Moving forward, incorporation of genetic tests can provide more accurate information to guide clinical decision-making and predict prognosis among patients with liver metastases.
Collapse
|
37
|
Lee J, Park S, Oh N, Park J, Kwon M, Seo J, Roh S. Oral intake of Lactobacillus plantarum L-14 extract alleviates TLR2- and AMPK-mediated obesity-associated disorders in high-fat-diet-induced obese C57BL/6J mice. Cell Prolif 2021; 54:e13039. [PMID: 33830560 PMCID: PMC8168423 DOI: 10.1111/cpr.13039] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/11/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Whether periodic oral intake of postbiotics positively affects weight regulation and prevents obesity-associated diseases in vivo is unclear. This study evaluated the action mechanism of Lactobacillus plantarum L-14 (KTCT13497BP) extract and the effects of its periodic oral intake in a high-fat-diet (HFD) mouse model. MATERIALS AND METHODS Mouse pre-adipocyte 3T3-L1 cells and human bone marrow mesenchymal stem cells (hBM-MSC) were treated with L-14 extract every 2 days during adipogenic differentiation, and the mechanism underlying anti-adipogenic effects was analysed at cellular and molecular levels. L-14 extract was orally administrated to HFD-feeding C57BL/6J mice every 2 days for 7 weeks. White adipose tissue was collected and weighed, and liver and blood serum were analysed. The anti-adipogenic mechanism of exopolysaccharide (EPS) isolated from L-14 extract was also analysed using Toll-like receptor 2 (TLR2) inhibitor C29. RESULTS L-14 extract inhibited 3T3-L1 and hBM-MSC differentiation into mature adipocytes by upregulating AMPK signalling pathway in the early stage of adipogenic differentiation. The weight of the HFD + L-14 group (31.51 ± 1.96 g) was significantly different from that of the HFD group (35.14 ± 3.18 g). L-14 extract also significantly decreased the serum triacylglycerol/high-density lipoprotein cholesterol ratio (an insulin resistance marker) and steatohepatitis. In addition, EPS activated the AMPK signalling pathway by interacting with TLR2, consequently inhibiting adipogenesis. CONCLUSIONS EPS from L-14 extract inhibits adipogenesis via TLR2 and AMPK signalling pathways, and oral intake of L-14 extract improves obesity and obesity-associated diseases in vivo. Therefore, EPS can be used to prevent and treat obesity and metabolic disorders.
Collapse
Affiliation(s)
- Jaehoon Lee
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seoul, Korea
| | - Sangkyu Park
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seoul, Korea.,Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Naeun Oh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Jaehyun Park
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Mijin Kwon
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Jeongmin Seo
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seoul, Korea.,Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| |
Collapse
|
38
|
Chumakova S, Urazova O, Shipulin V, Vins M, Pryakhin A, Sukhodolo I, Stelmashenko A, Litvinova L, Kolobovnikova Y, Churina E, Novitskiy V. Galectin 3 and non-classical monocytes of blood as myocardial remodeling factors at ischemic cardiomyopathy. IJC HEART & VASCULATURE 2021; 33:100766. [PMID: 33869726 PMCID: PMC8047163 DOI: 10.1016/j.ijcha.2021.100766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/19/2022]
Abstract
Aims To identify an imbalance of cardiac remodeling mediators and monocytes subpopulation in blood, distribution of myocardium macrophages in patients with ischemic cardiomyopathy (ICMP). Methods The study engaged 30 patients with ICMP, 26 patients with coronary heart disease (CHD) without ICMP, 15 healthy donors. Concentrations of TGFβ, MMP-9, MCP-1, galectin-3 were measured in plasma of blood from the coronary sinus and peripheral blood in CHD patients, as well as in peripheral blood in healthy donors, by enzyme immunoassay method. The ration of classical, intermediate, non-classical, transitional monocytes in peripheral blood of patients and healthy donors was assessed by flow cytometry (expression CD14, CD16); the content of CD68+ macrophages in myocardium – by immunohistochemistry method. Results In both samples of blood, the content of galectin-3 in patients with ICMP was higher than in CHD patients without ICMP and the level of TGFβ was comparable between the groups. At ICMP, the concentration of MMP-9 in sinus blood was higher than that in CHD patients without ICMP in whom an excess of MCP-1 in the general blood flow was determined. The density of distribution of CD68+ cells in the myocardium in patients with ICMP was higher in the perianeurysmal zone than in the right atrium appendage. ICMP was characterized by a deficiency of non-classical monocytes, and CHD without ICMP – by an excess of intermediate cells in peripheral blood. Conclusion Myocardium remodeling at ICMP is mediated by not so much TGFβ but intracardiac galectin-3, which determines the subpopulation composition of blood monocytes.
Collapse
Affiliation(s)
- S Chumakova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - O Urazova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - V Shipulin
- Cardiovascular Surgery Unit of CardiologyResearchInstitute, Tomsk National Medical Research Center of Russian Academy of Sciences, 111A Kievskaya Street, Tomsk 634012, Russia
| | - M Vins
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - A Pryakhin
- Cardiovascular Surgery Unit of CardiologyResearchInstitute, Tomsk National Medical Research Center of Russian Academy of Sciences, 111A Kievskaya Street, Tomsk 634012, Russia
| | - I Sukhodolo
- Morphology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - A Stelmashenko
- Morphology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - L Litvinova
- Immunology and Cell Biotechnology Center of Immanuel Kant, Baltic Federal University, 14 A. Nevskogo Street, Kaliningrad 236041, Russia
| | - Yu Kolobovnikova
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| | - E Churina
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia.,National Research Tomsk State University, 36 Lenina Ave, Tomsk 634050, Russia
| | - V Novitskiy
- Pathophysiology Division of Siberian State Medical University, 2 Moskovsky Trakt, Tomsk 634050, Russia
| |
Collapse
|
39
|
Furuta K, Guo Q, Pavelko KD, Lee JH, Robertson KD, Nakao Y, Melek J, Shah VH, Hirsova P, Ibrahim SH. Lipid-induced endothelial vascular cell adhesion molecule 1 promotes nonalcoholic steatohepatitis pathogenesis. J Clin Invest 2021; 131:143690. [PMID: 33476308 PMCID: PMC7954604 DOI: 10.1172/jci143690] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Monocyte homing to the liver and adhesion to the liver sinusoidal endothelial cells (LSECs) are key elements in nonalcoholic steatohepatitis (NASH) pathogenesis. We reported previously that VCAM-1 mediates monocyte adhesion to LSECs. However, the pathogenic role of VCAM-1 in NASH is unclear. Herein, we report that VCAM-1 was a top upregulated adhesion molecule in the NASH mouse liver transcriptome. Open chromatin landscape profiling combined with genome-wide transcriptome analysis showed robust transcriptional upregulation of LSEC VCAM-1 in murine NASH. Moreover, LSEC VCAM-1 expression was significantly increased in human NASH. LSEC VCAM-1 expression was upregulated by palmitate treatment in vitro and reduced with inhibition of the mitogen-activated protein 3 kinase (MAP3K) mixed lineage kinase 3 (MLK3). Likewise, LSEC VCAM-1 expression was reduced in the Mlk3-/- mice with diet-induced NASH. Furthermore, VCAM-1 neutralizing Ab or pharmacological inhibition attenuated diet-induced NASH in mice, mainly via reducing the proinflammatory monocyte hepatic population as examined by mass cytometry by time of flight (CyTOF). Moreover, endothelium-specific Vcam1 knockout mice were also protected against NASH. In summary, lipotoxic stress enhances the expression of LSEC VCAM-1, in part, through MLK3 signaling. Inhibition of VCAM-1 was salutary in murine NASH and might serve as a potential therapeutic strategy for human NASH.
Collapse
Affiliation(s)
| | | | | | - Jeong-Heon Lee
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, and
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jan Melek
- Department of Pediatrics, Charles University in Prague, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové, Czechia
| | | | | | - Samar H Ibrahim
- Division of Gastroenterology and Hepatology.,Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
40
|
Abstract
Pancreatic cancer (PC) is a malignant tumour of the human digestive system that has a poor prognosis. Exosomes contain proteins and nucleic acids, and constitute a class of extracellular vesicles defined as membrane-bound nanovesicles of endocytic origin, with a diameter of 40-150 nm. Exosomes are potential diagnostic markers of PC; however, their roles in cancer initiation and progression remain unclear. Previous studies have focused on the molecular mechanisms and functions of exosomes that allow them to accelerate PC cell proliferation, migration and invasion. The present review discusses the interactions between exosomes and the pathophysiology of PC. The potential clinical applications of exosomes are also discussed.
Collapse
|
41
|
Bacteria and Host Interplay in Staphylococcus aureus Septic Arthritis and Sepsis. Pathogens 2021; 10:pathogens10020158. [PMID: 33546401 PMCID: PMC7913561 DOI: 10.3390/pathogens10020158] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus (S. aureus) infections are a major healthcare challenge and new treatment alternatives are needed. S. aureus septic arthritis, a debilitating joint disease, causes permanent joint dysfunction in almost 50% of the patients. S. aureus bacteremia is associated with higher mortalities than bacteremia caused by most other microbes and can develop to severe sepsis and death. The key to new therapies is understanding the interplay between bacterial virulence factors and host immune response, which decides the disease outcome. S. aureus produces numerous virulence factors that facilitate bacterial dissemination, invasion into joint cavity, and cause septic arthritis. Monocytes, activated by several components of S. aureus such as lipoproteins, are responsible for bone destructions. In S. aureus sepsis, cytokine storm induced by S. aureus components leads to the hyperinflammatory status, DIC, multiple organ failure, and later death. The immune suppressive therapies at the very early time point might be protective. However, the timing of treatment is crucial, as late treatment may aggravate the immune paralysis and lead to uncontrolled infection and death.
Collapse
|
42
|
Marti-Aguado D, Rodríguez-Ortega A, Mestre-Alagarda C, Bauza M, Valero-Pérez E, Alfaro-Cervello C, Benlloch S, Pérez-Rojas J, Ferrández A, Alemany-Monraval P, Escudero-García D, Monton C, Aguilera V, Alberich-Bayarri Á, Serra MÁ, Marti-Bonmati L. Digital pathology: accurate technique for quantitative assessment of histological features in metabolic-associated fatty liver disease. Aliment Pharmacol Ther 2021; 53:160-171. [PMID: 32981113 DOI: 10.1111/apt.16100] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/24/2020] [Accepted: 09/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Histological evaluation of metabolic-associated fatty liver disease (MAFLD) biopsies is subjective, descriptive and with interobserver variability. AIMS To examine the relationship between different histological features (fibrosis, steatosis, inflammation and iron) measured with automated whole-slide quantitative digital pathology and corresponding semiquantitative scoring systems, and the distribution of digital pathology measurements across Fatty Liver Inhibition of Progression (FLIP) algorithm and Steatosis, Activity and Fibrosis (SAF) scoring system METHODS: We prospectively included 136 consecutive patients who underwent liver biopsy for MAFLD at three Spanish centres (January 2017-January 2020). Biopsies were scored by two blinded pathologists according to the Non-alcoholic Steatohepatitis (NASH) Clinical Research Network system for fibrosis staging, the FLIP/SAF classification for steatosis and inflammation grading and Deugnier score for iron grading. Proportionate areas of collagen, fat, inflammatory cells and iron deposits were measured with computer-assisted digital image analysis. A test-retest experiment was performed for precision repeatability evaluation. RESULTS Digital pathology showed strong correlation with fibrosis (r = 0.79; P < 0.001), steatosis (r = 0.85; P < 0.001) and iron (r = 0.70; P < 0.001). Performance was lower when assessing the degree of inflammation (r = 0.35; P < 0.001). NASH cases had a higher proportion of collagen and fat compared to non-NASH cases (P < 0.005), whereas inflammation and iron quantification did not show significant differences between categories. Repeatability evaluation showed that all the coefficients of variation were ≤1.1% and all intraclass correlation coefficient values were ≥0.99, except those of collagen. CONCLUSION Digital pathology allows an automated, precise, objective and quantitative assessment of MAFLD histological features. Digital analysis measurements show good concordance with pathologists´ scores.
Collapse
|
43
|
Boulter L, Bullock E, Mabruk Z, Brunton VG. The fibrotic and immune microenvironments as targetable drivers of metastasis. Br J Cancer 2021; 124:27-36. [PMID: 33239677 PMCID: PMC7782519 DOI: 10.1038/s41416-020-01172-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Although substantial progress has been made over the past 40 years in treating patients with cancer, effective therapies for those who are diagnosed with advanced metastatic disease are still few and far between. Cancer cells do not exist in isolation: rather, they exist within a complex microenvironment composed of stromal cells and extracellular matrix. Within this tumour microenvironment exists an interplay between the two main stromal cell subtypes, cancer-associated fibroblasts (CAFs) and immune cells, that are important in controlling metastasis. A complex network of paracrine signalling pathways between CAFs, immune cells and tumour cells are involved at multiple stages of the metastatic process, from invasion and intravasation at the primary tumour site to extravasation and colonisation in the metastatic site. Heterogeneity and plasticity within stromal cell populations also contribute to the complexity. Although many of these processes are likely to be common to a number of metastatic sites, we will describe in detail the interplay within the liver, a preferred site of metastasis for many tumours. A greater understanding of these networks provides opportunities for the design of new therapeutic approaches for targeting the metastatic disease.
Collapse
Affiliation(s)
- Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Esme Bullock
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Zeanap Mabruk
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
44
|
Stevens HY, Bowles AC, Yeago C, Roy K. Molecular Crosstalk Between Macrophages and Mesenchymal Stromal Cells. Front Cell Dev Biol 2020; 8:600160. [PMID: 33363157 PMCID: PMC7755599 DOI: 10.3389/fcell.2020.600160] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been widely investigated for regenerative medicine applications, from treating various inflammatory diseases as a cell therapy to generating engineered tissue constructs. Numerous studies have evaluated the potential effects of MSCs following therapeutic administration. By responding to their surrounding microenvironment, MSCs may mediate immunomodulatory effects through various mechanisms that directly (i.e., contact-dependent) or indirectly (i.e., paracrine activity) alter the physiology of endogenous cells in various disease pathologies. More specifically, a pivotal crosstalk between MSCs and tissue-resident macrophages and monocytes (TMφ) has been elucidated using in vitro and in vivo preclinical studies. An improved understanding of this crosstalk could help elucidate potential mechanisms of action (MOAs) of therapeutically administered MSCs. TMφ, by nature of their remarkable functional plasticity and prevalence within the body, are uniquely positioned as critical modulators of the immune system - not only in maintaining homeostasis but also during pathogenesis. This has prompted further exploration into the cellular and molecular alterations to TMφ mediated by MSCs. In vitro assays and in vivo preclinical trials have identified key interactions mediated by MSCs that polarize the responses of TMφ from a pro-inflammatory (i.e., classical activation) to a more anti-inflammatory/reparative (i.e., alternative activation) phenotype and function. In this review, we describe physiological and pathological TMφ functions in response to various stimuli and discuss the evidence that suggest specific mechanisms through which MSCs may modulate TMφ phenotypes and functions, including paracrine interactions (e.g., secretome and extracellular vesicles), nanotube-mediated intercellular exchange, bioenergetics, and engulfment by macrophages. Continued efforts to elucidate this pivotal crosstalk may offer an improved understanding of the immunomodulatory capacity of MSCs and inform the development and testing of potential MOAs to support the therapeutic use of MSCs and MSC-derived products in various diseases.
Collapse
Affiliation(s)
- Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annie C. Bowles
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Carolyn Yeago
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
- Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
45
|
Forrester JV, Kuffova L, Delibegovic M. The Role of Inflammation in Diabetic Retinopathy. Front Immunol 2020; 11:583687. [PMID: 33240272 PMCID: PMC7677305 DOI: 10.3389/fimmu.2020.583687] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is central to pathogenic processes in diabetes mellitus and the metabolic syndrome and particularly implicates innate immunity in the development of complications. Inflammation is a primary event in Type 1 diabetes where infectious (viral) and/or autoimmune processes initiate disease; in contrast, chronic inflammation is typical in Type 2 diabetes and is considered a sequel to increasing insulin resistance and disturbed glucose metabolism. Diabetic retinopathy (DR) is perceived as a vascular and neurodegenerative disease which occurs after some years of poorly controlled diabetes. However, many of the clinical features of DR are late events and reflect the nature of the retinal architecture and its cellular composition. Retinal microvascular disease is, in fact, an early event pathogenetically, induced by low grade, persistent leukocyte activation which causes repeated episodes of capillary occlusion and, progressive, attritional retinal ischemia. The later, overt clinical signs of DR are a consequence of the retinal ischemia. Metabolic dysregulation involving both lipid and glucose metabolism may lead to leukocyte activation. On a molecular level, we have shown that macrophage-restricted protein tyrosine phosphatase 1B (PTP1B) is a key regulator of inflammation in the metabolic syndrome involving insulin resistance and it is possible that PTP1B dysregulation may underlie retinal microvascular disease. We have also shown that adherent CCR5+CD11b+ monocyte macrophages appear to be selectively involved in retinal microvascular occlusion. In this review, we discuss the relationship between early leukocyte activation and the later features of DR, common pathogenetic processes between diabetic microvascular disease and other vascular retinopathies, the mechanisms whereby leukocyte activation is induced in hyperglycemia and dyslipidemia, the signaling mechanisms involved in diabetic microvascular disease, and possible interventions which may prevent these retinopathies. We also address a possible role for adaptive immunity in DR. Although significant improvements in treatment of DR have been made with intravitreal anti-VEGF therapy, a sizeable proportion of patients, particularly with sight-threatening macular edema, fail to respond. Alternative therapies targeting inflammatory processes may offer an advantage.
Collapse
Affiliation(s)
- John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom
| | - Lucia Kuffova
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom.,Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Mirela Delibegovic
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom
| |
Collapse
|
46
|
Ye H, Minhajuddin M, Krug A, Pei S, Chou CH, Culp-Hill R, Ponder J, De Bloois E, Schniedewind B, Amaya ML, Inguva A, Stevens BM, Pollyea DA, Christians U, Grimes HL, D'Alessandro A, Jordan CT. The Hepatic Microenvironment Uniquely Protects Leukemia Cells through Induction of Growth and Survival Pathways Mediated by LIPG. Cancer Discov 2020; 11:500-519. [PMID: 33028621 DOI: 10.1158/2159-8290.cd-20-0318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/11/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Due to the disseminated nature of leukemia, malignant cells are exposed to many different tissue microenvironments, including a variety of extramedullary sites. In the present study, we demonstrate that leukemic cells residing in the liver display unique biological properties and also contribute to systemic changes that influence physiologic responses to chemotherapy. Specifically, the liver microenvironment induces metabolic adaptations via upregulating expression of endothelial lipase in leukemia cells, which not only stimulates tumor cell proliferation through polyunsaturated fatty acid-mediated pathways, but also promotes survival by stabilizing antiapoptotic proteins. Additionally, hepatic infiltration and tissue damage caused by malignant cells induces release of liver-derived enzymes capable of degrading chemotherapy drugs, an event that further protects leukemia cells from conventional therapies. Together, these studies demonstrate a unique role for liver in modulating the pathogenesis of leukemic disease and suggest that the hepatic microenvironment may protect leukemia cells from chemotherapeutic challenge. SIGNIFICANCE: The studies presented herein demonstrate that the liver provides a microenvironment in which leukemia cells acquire unique metabolic properties. The adaptations that occur in the liver confer increased resistance to chemotherapy. Therefore, we propose that therapies designed to overcome liver-specific metabolic changes will yield improved outcomes for patients with leukemia.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Haobin Ye
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Mohammad Minhajuddin
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anna Krug
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shanshan Pei
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Chih-Hsing Chou
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica Ponder
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Erik De Bloois
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Björn Schniedewind
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Maria L Amaya
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anagha Inguva
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Angelo D'Alessandro
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
47
|
Cacicedo ML, Medina-Montano C, Kaps L, Kappel C, Gehring S, Bros M. Role of Liver-Mediated Tolerance in Nanoparticle-Based Tumor Therapy. Cells 2020; 9:E1985. [PMID: 32872352 PMCID: PMC7563539 DOI: 10.3390/cells9091985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decades, the use of nanocarriers for immunotherapeutic purposes has gained a lot of attention, especially in the field of tumor therapy. However, most types of nanocarriers accumulate strongly in the liver after systemic application. Due to the default tolerance-promoting role of liver non-parenchymal cells (NPCs), Kupffer cells (KCs), liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs), their potential role on the immunological outcome of systemic nano-vaccination approaches for therapy of tumors in the liver and in other organs needs to be considered. Concerning immunological functions, KCs have been the focus until now, but recent studies have elucidated an important role of LSECs and HSCs as well. Therefore, this review aims to summarize current knowledge on the employment of nanocarriers for immunotherapeutic therapy of liver diseases and the overall role of liver NPCs in the context of nano-vaccination approaches. With regard to the latter, we discuss strategies on how to address liver NPCs, aiming to exploit and modulate their immunological properties, and alternatively how to avoid unwanted engagement of nano-vaccines by liver NPCs for tumor therapy.
Collapse
Affiliation(s)
- Maximiliano L. Cacicedo
- Children’s Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.L.C.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| | - Leonard Kaps
- Department of Medicine, University Medical Center Mainz, I. Langenbeckstrasse 1, 55131 Mainz, Germany;
| | - Cinja Kappel
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| | - Stephan Gehring
- Children’s Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.L.C.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| |
Collapse
|
48
|
Tavallaee G, Rockel JS, Lively S, Kapoor M. MicroRNAs in Synovial Pathology Associated With Osteoarthritis. Front Med (Lausanne) 2020; 7:376. [PMID: 32850892 PMCID: PMC7431695 DOI: 10.3389/fmed.2020.00376] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis, a disease that affects the entire joint. The relative involvement of each tissue, and their interactions, add to the complexity of OA, hampering our understanding of the underlying molecular mechanisms, and the generation of a disease modifying therapy. The synovium is essential in maintaining joint homeostasis, and pathologies associated with the synovium contribute to joint destruction, pain and stiffness in OA. MicroRNAs (miRNAs) are post-transcriptional regulators dysregulated in OA tissues including the synovium. MiRNAs are important contributors to OA synovial changes that have the potential to improve our understanding of OA and to act as novel therapeutic targets. The purpose of this review is to summarize and integrate current published literature investigating the roles that miRNAs play in OA-related synovial pathologies including inflammation, matrix deposition and cell proliferation.
Collapse
Affiliation(s)
- Ghazaleh Tavallaee
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jason S. Rockel
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Starlee Lively
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mohit Kapoor
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
da Silva CO, Gicquel T, Daniel Y, Bártholo T, Vène E, Loyer P, Pôrto LC, Lagente V, Victoni T. Alteration of immunophenotype of human macrophages and monocytes after exposure to cigarette smoke. Sci Rep 2020; 10:12796. [PMID: 32732964 PMCID: PMC7393094 DOI: 10.1038/s41598-020-68753-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/29/2020] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke exposure (CS) is the main risk factor for chronic obstructive pulmonary disease (COPD). Macrophages have an important role in COPD because they release pro-inflammatory and anti-inflammatory cytokines. The present study's we investigate the functional changes in macrophages and monocytes exposed to cigarette smoke extract (CSE). Herein, using human monocyte-derived macrophages (MDMs) from healthy donors and we found that CSE was not associated with significant changes in the production of pro inflammatory cytokines by MDMs. In contrast, exposure to CSE suppressed the production of IL-6 and Gro-a/CXCL1 by LPS-stimulated-MDMs, but had an additive effect on the release of IL-8/CXCL8 and MCP1/CCL2. However, CSE exposure was associated with greater production, TARC/CCL-17 and CCL22/MDC. Moreover, MDMs displayed a lower uptake capacity after CSE exposure. We identify, for what is to our knowledge the first time that monocytes from patients with COPD produced less IL-8/CXCL8 and Gro-α/CXCL1 after LPS stimulation and produced higher levels of TARC/CCL17 and MDC/CCL-22 after IL-4 stimulation. Our present results highlighted a skewed immune response, with an imbalance in M1 vs. M2 cytokine production. In conclusion, exposure to CS has contrasting, multifaceted effects on macrophages and monocytes. Our data may provide a better understanding of the mechanisms underlying COPD.
Collapse
Affiliation(s)
- Camila Oliveira da Silva
- Laboratory of Histocompatibility and Cryopresevation, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thomas Gicquel
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France
- Forensic and Toxicology Laboratory, Pontchaillou University Hospital, Rennes, France
| | - Yoann Daniel
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France
| | - Thiago Bártholo
- Laboratory of Histocompatibility and Cryopresevation, Rio de Janeiro State University, Rio de Janeiro, Brazil
- Department of Pulmonology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Elise Vène
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France
| | - Pascal Loyer
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France
| | - Luís Cristóvão Pôrto
- Laboratory of Histocompatibility and Cryopresevation, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vincent Lagente
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France
| | - Tatiana Victoni
- Laboratory of Histocompatibility and Cryopresevation, Rio de Janeiro State University, Rio de Janeiro, Brazil.
- NuMeCan Institute (Nutrition, Metabolism and Cancer), INSERM, INRAE, CHU Rennes, Hôpital Pontchaillou, Univ Rennes, 35033, Rennes Cedex, France.
- University of Lyon, VetAgro Sup, APCSe, Marcy l'Étoile, France.
| |
Collapse
|
50
|
Luque-Martin R, Mander PK, Leenen PJM, Winther MPJ. Classic and new mediators for in vitro modelling of human macrophages. J Leukoc Biol 2020; 109:549-560. [PMID: 32592421 PMCID: PMC7984372 DOI: 10.1002/jlb.1ru0620-018r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages are key immune cells in the activation and regulation of immune responses. These cells are present in all tissues under homeostatic conditions and in many disease settings. Macrophages can exhibit a wide range of phenotypes depending on local and systemic cues that drive the differentiation and activation process. Macrophage heterogeneity is also defined by their ontogeny. Tissue macrophages can either derive from circulating blood monocytes or are seeded as tissue-resident macrophages during embryonic development. In humans, the study of in vivo-generated macrophages is often difficult with laborious and cell-changing isolation procedures. Therefore, translatable, reproducible, and robust in vitro models for human macrophages in health and disease are necessary. Most of the methods for studying monocyte-derived macrophages are based on the use of limited factors to differentiate the monocytes into macrophages. Current knowledge shows that the in vivo situation is more complex, and a wide range of molecules in the tissue microenvironment promote and impact on monocyte to macrophage differentiation as well as activation. In this review, macrophage heterogeneity is discussed and the human in vitro models that can be applied for research, especially for monocyte-derived macrophages. We also focus on new molecules (IL-34, platelet factor 4, etc.) used to generate macrophages expressing different phenotypes.
Collapse
Affiliation(s)
- Rosario Luque-Martin
- Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | | | - Pieter J M Leenen
- Erasmus University Medical Center, Department of Immunology, Rotterdam, The Netherlands
| | - Menno P J Winther
- Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| |
Collapse
|