1
|
Schmidt AA, David LM, Qayyum NT, Tran K, Van C, Hetta AHSHA, Shrestha RL, Varatip AO, Butenko S, Enriquez-Ochoa D, Nguyen C, Seldin MM, Liu WF, Grosberg A. Polarized macrophages modulate cardiac structure and contractility under hypoxia in novel immuno-heart on a chip. APL Bioeng 2025; 9:026114. [PMID: 40322069 PMCID: PMC12048176 DOI: 10.1063/5.0253888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cardiac adaptation to hypoxic injury is regulated by dynamic interactions between cardiomyocytes and macrophages, yet the impacts of immune phenotypes on cardiac structure and contractility remain poorly understood. To address this, we developed the immuno-heart on a chip, a novel in vitro platform to investigate cardiomyocyte-macrophage interactions under normoxic and hypoxic conditions. By integrating neonatal rat ventricular myocytes (NRVMs) and bone marrow-derived macrophages-polarized to pro-inflammatory (M1) or pro-healing (M2/M2*) phenotypes-we elucidated the dual protective and detrimental roles macrophages play in modulating cardiomyocyte cytoskeletal architecture and contractility. Pro-inflammatory stimulation reduced cardiomyocyte structural metrics (z-line length, fraction, and integrity) in normoxic co-cultures. Under hypoxia, M1-stimulated NRVM monocultures exhibited declines in cytoskeletal organization-quantified by actin and z-line orientational order parameters. Relative to monocultures, M1-stimulated co-cultures attenuated hypoxia-induced active stress declines but produced weaker normoxic stresses. In contrast, pro-healing stimulation improved normoxic z-line metrics and preserved post-hypoxia cytoskeletal organization but reduced normoxic contractility. Notably, M2-stimulated macrophages restored normoxic contractility and preserved post-hypoxia systolic stress, albeit with increased diastolic stress. RNAseq analysis of M2-stimulated co-cultures identified upregulated structural and immune pathways driving these hypoxia-induced changes. Cytokine profiles revealed stimulation-specific and density-dependent tumor necrosis factor-alpha and interleukin-10 secretion patterns. Together, these findings quantitatively link clinically relevant macrophage phenotypes and cytokines to distinct changes in cardiac structure and contractility, offering mechanistic insights into immune modulation of hypoxia-induced dysfunction. Moreover, the immuno-heart on a chip represents an innovative framework to guide the development of future therapies that integrate immune and cardiac targets to enhance patient outcomes.
Collapse
|
2
|
Lösslein AK, Henneke P. Macrophage Differentiation and Metabolic Adaptation in Mycobacterial Infections. Annu Rev Immunol 2025; 43:423-450. [PMID: 40014665 DOI: 10.1146/annurev-immunol-082323-120757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The adaptation of macrophages-the most common tissue-resident immune cells-to metabolic and microbial cues with high local variability is essential for the maintenance of organ integrity. In homeostasis, macrophages show largely predictable tissue-specific differentiation, as recently revealed by multidimensional methods. However, chronic infections with human-adapted pathogens substantially contribute to the differentiation complexity of tissue macrophages, which has been only partially resolved. Specifically, the response to mycobacterial species-which range from Mycobacterium tuberculosis (with highest specificity for humans, broad organ tropism, yet tissue-specific disease phenotypes) to environmental mycobacteria with humans as accidental hosts-may serve as a paradigm of tissue macrophage adaptation mechanisms. While mycobacterial species-specific tissue preferences are partially related to the mode of acquisition and pathogen characteristics, evolutionary convergence with macrophages driven by metabolic features of the target organ likely contributes to infection resistance and immunopathology. In this review, we unravel the mechanisms of tissue-specific macrophage differentiation and its limitations in mycobacterial infections.
Collapse
Affiliation(s)
- Anne Kathrin Lösslein
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Wang H, Liu Z, Du Y, Cheng X, Gao S, Liang W, Zhu Q, Jiang Z, Gao Y, Shang P. High expression of ARPC1B promotes the proliferation and apoptosis of clear cell renal cell carcinoma cells, leading to a poor prognosis. Mol Cell Probes 2025; 79:102011. [PMID: 39818256 DOI: 10.1016/j.mcp.2025.102011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/28/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND ARPC1B has been identified as a key regulator of malignant biological behavior in various tumors. However, its specific role in clear cell renal cell carcinoma (ccRCC) remains poorly understood. This study aims to evaluate the influence of ARPC1B on the prognosis and disease progression in ccRCC patients. METHODS Multi-omics data and clinical information from public databases were analyzed to determine the associations between ARPC1B and prognosis, clinical features, immune microenvironment, and drug sensitivity in ccRCC. Co-expression and gene set enrichment analyses were conducted to elucidate the potential role of ARPC1B in ccRCC pathogenesis. Functional assays, including RT-qPCR, CCK8 assays, colony formation assays, immunofluorescence, immunohistochemistry, and xenograft tumor formation in nude mice, were performed to assess ARPC1B's impact on cell proliferation and apoptosis. Flow cytometry and Western blotting were further employed to investigate the underlying molecular mechanisms of ARPC1B in ccRCC. RESULTS ARPC1B expression was significantly elevated in ccRCC and associated with an unfavorable prognosis. Both independent and meta-analyses confirmed that ARPC1B is an independent prognostic risk factor in ccRCC. Furthermore, ARPC1B expression significantly correlated with the immune microenvironment and drug sensitivity. In vitro, experiments demonstrated that ARPC1B knockdown suppressed ccRCC cell proliferation and induced apoptosis through the BAX-Bcl-2/c-caspase3/c-PARP axis, which was further validated by in vivo studies. CONCLUSION ARPC1B overexpression is associated with poor prognosis, altered immune status, and drug sensitivity in ccRCC. Furthermore, ARPC1B promotes the malignant behavior of ccRCC cells and holds potential as a prognostic biomarker and therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Hongbo Wang
- Department of Urology Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China; Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yuelin Du
- Department of Urology Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Shanjun Gao
- Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450003, China
| | - Wenjia Liang
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Qingyun Zhu
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhengfa Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Panfeng Shang
- Department of Urology Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
4
|
Ummarino A, Calà N, Allavena P. Extrinsic and Cell-Intrinsic Stress in the Immune Tumor Micro-Environment. Int J Mol Sci 2024; 25:12403. [PMID: 39596467 PMCID: PMC11594858 DOI: 10.3390/ijms252212403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
In continuously progressive tumor tissues, the causes of cellular stress are multiple: metabolic alterations, nutrient deprivation, chronic inflammation and hypoxia. To survive, tumor cells activate the stress response program, a highly conserved molecular reprogramming proposed to cope with challenges in a hostile environment. Not only cancer cells are affected, but stress responses in tumors also have a profound impact on their normal cellular counterparts: fibroblasts, endothelial cells and infiltrating immune cells. In recent years, there has been a growing interest in the interaction between cancer and immune cells, especially in difficult conditions of cellular stress. A growing literature indicates that knowledge of the molecular pathways activated in tumor and immune cells under stress conditions may offer new insights for possible therapeutic interventions. Counter-regulating the stress caused by the presence of a growing tumor can therefore be a weapon to limit disease progression. Here, we review the main pathways activated in cellular stress responses with a focus on immune cells present in the tumor microenvironment.
Collapse
Affiliation(s)
- Aldo Ummarino
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Nicholas Calà
- Etromapmacs Pole, Agorà Biomedical Sciences, 71010 Foggia, Italy;
| | | |
Collapse
|
5
|
Zhuang XM, Guo ZY, Zhang M, Chen YH, Qi FN, Wang RQ, Zhang L, Zhao PJ, Lu CJ, Zou CG, Ma YC, Xu J, Zhang KQ, Cao YR, Liang LM. Ethanol mediates the interaction between Caenorhabditis elegans and the nematophagous fungus Purpureocillium lavendulum. Microbiol Spectr 2023; 11:e0127023. [PMID: 37560934 PMCID: PMC10580998 DOI: 10.1128/spectrum.01270-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
Accurately recognizing pathogens by the host is vital for initiating appropriate immune response against infecting microorganisms. Caenorhabditis elegans has no known receptor to recognize pathogen-associated molecular pattern. However, recent studies showed that nematodes have a strong specificity for transcriptomes infected by different pathogens, indicating that they can identify different pathogenic microorganisms. However, the mechanism(s) for such specificity remains largely unknown. In this study, we showed that the nematophagous fungus Purpureocillium lavendulum can infect the intestinal tract of the nematode C. elegans and the infection led to the accumulation of reactive oxygen species (ROS) in the infected intestinal tract, which suppressed fungal growth. Co-transcriptional analysis revealed that fungal genes related to anaerobic respiration and ethanol production were up-regulated during infection. Meanwhile, the ethanol dehydrogenase Sodh-1 in C. elegans was also up-regulated. Together, these results suggested that the infecting fungi encounter hypoxia stress in the nematode gut and that ethanol may play a role in the host-pathogen interaction. Ethanol production in vitro during fungal cultivation in hypoxia conditions was confirmed by gas chromatography-mass spectrometry. Direct treatment of C. elegans with ethanol elevated the sodh-1 expression and ROS accumulation while repressing a series of immunity genes that were also repressed during fungal infection. Mutation of sodh-1 in C. elegans blocked ROS accumulation and increased the nematode's susceptibility to fungal infection. Our study revealed a new recognition and antifungal mechanism in C. elegans. The novel mechanism of ethanol-mediated interaction between the fungus and nematode provides new insights into fungal pathogenesis and for developing alternative biocontrol of pathogenic nematodes by nematophagous fungi. IMPORTANCE Nematodes are among the most abundant animals on our planet. Many of them are parasites in animals and plants and cause human and animal health problems as well as agricultural losses. Studying the interaction of nematodes and their microbial pathogens is of great importance for the biocontrol of animal and plant parasitic nematodes. In this study, we found that the model nematode Caenorhabditis elegans can recognize its fungal pathogen, the nematophagous fungus Purpureocillium lavendulum, through fungal-produced ethanol. Then the nematode elevated the reactive oxygen species production in the gut to inhibit fungal growth in an ethanol dehydrogenase-dependent manner. With this mechanism, novel biocontrol strategies may be developed targeting the ethanol receptor or metabolic pathway of nematodes. Meanwhile, as a volatile organic compound, ethanol should be taken seriously as a vector molecule in the microbial-host interaction in nature.
Collapse
Affiliation(s)
- Xue-Mei Zhuang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Zhi-Yi Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Meng Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yong-Hong Chen
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Feng-Na Qi
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Ren-Qiao Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Ling Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Pei-Ji Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Chao-Jun Lu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Cheng-Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yi-Cheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Jianping Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Ke-Qin Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yan-Ru Cao
- College of Agriculture and Life Sciences, Kunming University, Kunming, China
| | - Lian-Ming Liang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| |
Collapse
|
6
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
7
|
Single-Cell RNA-Sequencing Reveals the Active Involvement of Macrophage Polarizations in Pulmonary Hypertension. DISEASE MARKERS 2022; 2022:5398157. [PMID: 36246557 PMCID: PMC9553540 DOI: 10.1155/2022/5398157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022]
Abstract
Background. Sustained hypoxia can trigger a progressive rise in pulmonary artery pressure and cause serious pulmonary diseases. Macrophages play important roles along the progression of pulmonary hypertension. However, the state of macrophage polarization during the early stage of pulmonary hypertension is unclear. Methods. Unlike traditional sequencing method, single-cell sequencing can accurately distinguish among cell types and better understand cell-to-cell relationships. In this study, we investigated the polarization of macrophages in pulmonary hypertension via single-cell RNA-sequencing in a mice hypoxia model, which was then validated in patients with pulmonary hypertension. Results. We identified that the intermittent exposure to hypoxic conditions could lead to the production of more M2-type macrophages than M1-type macrophages in a mouse model. Further validation analysis was performed by analyzing lung tissue of patients with pulmonary hypertension, revealing that the number of disease-associated M2 macrophages was substantially increased. Conclusions. In this study, the active anti-inflammatory response of macrophage involved in pulmonary hypertension has been identified, suggesting that intervention against the polarization of macrophages to the M2 type may be a potential way to reduce chronic pulmonary inflammation, pulmonary vascular remodeling, and artery pressure. Thus, investigation of macrophage polarization associated with hypoxia could help us better understand disease mechanism and craft effective prevention strategies and approaches.
Collapse
|
8
|
Ju H, Park KW, Kim ID, Cave JW, Cho S. Phagocytosis converts infiltrated monocytes to microglia-like phenotype in experimental brain ischemia. J Neuroinflammation 2022; 19:190. [PMID: 35850727 PMCID: PMC9295522 DOI: 10.1186/s12974-022-02552-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/02/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Monocyte-derived macrophages (MDMs) and microglia elicit neural inflammation and clear debris for subsequent tissue repair and remodeling. The role of infiltrating MDMs in the injured brain, however, has been controversial due to overlapping antigen expression with microglia. In this study, we define the origin and function of MDMs in cerebral ischemia. METHODS Using adoptive transfer of GFP+ splenocytes into adult asplenic mice subjected to transient middle cerebral artery occlusion, we compared the role of CD11b+/CD45+/NK1.1-/Ly6G- MDMs and microglia in the ischemic brain. The phagocytic activities of MDMs and microglia were measured by the uptake of fluorescent beads both in vivo with mice infused with GFP+ splenocytes and ex vivo with cultures of isolated brain immune cells. RESULTS Stroke induced an infiltration of MDMs [GFP+] into the ipsilateral hemisphere at acute (3 days) and sub-acute phases (7 days) of post-stroke. At 7 days, the infiltrating MDMs contained both CD45High and CD45Low subsets. The CD45High MDMs in the injured hemisphere exhibited a significantly higher proliferation capacity (Ki-67 expression levels) as well as higher expression levels of CD11c when compared to CD45Low MDMs. The CD45High and CD45Low MDM subsets in the injured hemisphere were approximately equal populations, indicating that CD45High MDMs infiltrating the ischemic brain changes their phenotype to CD45Low microglia-like phenotype. Studies with fluorescent beads reveal high levels of MDM phagocytic activity in the post-stroke brain, but this phagocytic activity was exclusive to post-ischemic brain tissue and was not detected in circulating monocytes. By contrast, CD45Low microglia-like cells had low levels of phagocytic activity when compared to CD45High cells. Both in vivo and ex vivo studies also show that the phagocytic activity in CD45High MDMs is associated with an increase in the CD45Low/CD45High ratio, indicating that phagocytosis promotes MDM phenotype conversion. CONCLUSIONS This study demonstrates that MDMs are the predominant phagocytes in the post-ischemic brain, with the CD45High subset having the highest phagocytic activity levels. Upon phagocytosis, CD45High MDMs in the post-ischemic brain adopt a CD45Low phenotype that is microglia-like. Together, these studies reveal key roles for MDMs and their phagocytic function in tissue repair and remodeling following cerebral ischemia.
Collapse
Affiliation(s)
- Hyunwoo Ju
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA
| | - Keun Woo Park
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Il-Doo Kim
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA
| | | | - Sunghee Cho
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA.
- Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Simulated hypoxia modulates P2X7 receptor function in mice peritoneal macrophages. Int Immunopharmacol 2022; 110:109062. [PMID: 35863257 DOI: 10.1016/j.intimp.2022.109062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
The inflammatory focus is similar to the tumor microenvironment, which contains a complex milieu with immune cells and macrophages. The accumulation of cells promotes local pH and O2 tension decline (hypoxia). Local O2 tension decline activates hypoxia-inducible factor α and β (HIF-1α and HIF-1β adenosine triphosphate (ATP) release. ATP activates the P2X7 receptor and modulates ischemic/hypoxic conditions. Similarly, α1α may regulate P2X7 receptor expression in the hypoxic microenvironment. Therefore, we investigated P2X7 receptor function under simulated hypoxic conditions by pretreating peritoneal macrophages with mitochondrial electron transport chain complex inhibitors (simulated hypoxia). Treatment with mitochondrial electron transport chain complex inhibitors until three hours of exposure did not cause LDH release. Additionally, mitochondrial electron transport chain complex inhibitors increased ATP-induced P2X7 receptor function without being able to directly activate this receptor. Other P2 receptor subtypes do not appear to participate in this mechanism. Simulated hypoxia augmented HIF-1α levels and suppressed HIF-1α and P2X7 receptor antagonists. Similarly, simulated hypoxia increased ATP-induced dye uptake and inhibited HIF-1α antagonists. Another factor activated in simulated hypoxic conditions was the intracellular P2X7 receptor regulator PIP2. Treatment with HIF-1α agonists increased PIP2 levels and reversed the effects of HIF-1α and P2X7 receptor antagonists. Additionally, the improved ATP-induced dye uptake caused by the simulated hypoxia stimulus was inhibited by P2X7 receptor and PIP2 antagonists. Therefore, simulated hypoxia may augment P2X7 receptor activity for a pathway dependent on HIF-1α and PIP2 activation.
Collapse
|
10
|
Tajaldini M, Saeedi M, Amiriani T, Amiriani AH, Sedighi S, Mohammad Zadeh F, Dehghan M, Jahanshahi M, Zanjan Ghandian M, Khalili P, Poorkhani AH, Alizadeh AM, Khori V. Cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs); where do they stand in tumorigenesis and how they can change the face of cancer therapy? Eur J Pharmacol 2022; 928:175087. [PMID: 35679891 DOI: 10.1016/j.ejphar.2022.175087] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/18/2022] [Accepted: 06/03/2022] [Indexed: 11/03/2022]
Abstract
The tumor microenvironment (TME) and its components have recently attracted tremendous attention in cancer treatment strategies, as alongside the genetic and epigenetic alterations in tumor cells, TME could also provide a fertile background for malignant cells to survive and proliferate. Interestingly, TME plays a vital role in the mediation of cancer metastasis and drug resistance even against immunotherapeutic agents. Among different cells that are presenting in TME, tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) have shown to have significant value in the regulation of angiogenesis, tumor metastasis, and drug-resistance through manipulating the composition as well as the organization of extracellular matrix (ECM). Evidence has shown that the presence of both TAMs and CAFs in TME is associated with poor prognosis and failure of chemotherapeutic agents. It seems that these cells together with ECM form a shield around tumor cells to protect them from the toxic agents and even the adaptive arm of the immune system, which is responsible for tumor surveillance. Given this, targeting TAMs and CAFs seems to be an essential approach to potentiate the cytotoxic effects of anti-cancer agents, either conventional chemotherapeutic drugs or immunotherapies. In the present review, we aimed to take a deep look at the mechanobiology of CAFs and TAMs in tumor progression and to discuss the available therapeutic approaches for harnessing these cells in TME.
Collapse
Affiliation(s)
- Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohsen Saeedi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amir Hossein Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sima Sedighi
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Mohammad Zadeh
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Dehghan
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maziar Zanjan Ghandian
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Pedram Khalili
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
11
|
Witte HM, Gebauer N, Steinestel K. Mutational and immunologic Landscape in malignant Salivary Gland Tumors harbor the potential for novel therapeutic strategies. Crit Rev Oncol Hematol 2022; 170:103592. [PMID: 35026433 DOI: 10.1016/j.critrevonc.2022.103592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
Salivary gland carcinomas (SGC) are rare (3-6 % of all head and neck cancers) and show biological heterogeneity depending on the respective histological subtype. While complete surgical resection is the standard treatment for localized disease, chemotherapy or radiation therapy are frequently insufficient for the treatment of unresectable or metastasized SGC. Therefore, new therapeutic approaches such as molecularly targeted therapy or the application of immune checkpoint inhibition enhance the treatment repertoire. Accordingly, comprehensive analyses of the genomic landscape and the tumor-microenvironment (TME) are of crucial importance in order to optimize and individualize SGC treatment. This manuscript combines the current scientific knowledge of the composition of the mutational landscape and the TME in SGCs harboring the potential for novel (immune-) targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany; Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany; Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| |
Collapse
|
12
|
Polaryzacja mikrogleju i makrofagów w wybranych chorobach degeneracyjnych i zapalnych układu nerwowego. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Makrofagi to komórki efektorowe układu odpornościowego zdolne do polaryzacji, czyli zmiany fenotypu powiązanej ze zmianą aktywności. Można wyróżnić: polaryzację klasyczną (M1), która służy obronie przed patogenami, a makrofagi M1 mają aktywność ogólnie prozapalną, oraz polaryzację alternatywną (M2), która sprzyja wygaszaniu stanu zapalnego i regeneracji tkanki. Makrofagi zasiedlają niemal cały organizm, więc zjawisko ich polaryzacji ma wpływ na wiele procesów zachodzących w różnych tkankach. W układzie nerwowym reprezentacją osiadłych makrofagów jest mikroglej. Jednak w wielu sytuacjach patologicznych w mózgu pojawiają się także makrofagi rekrutowane z monocytów krążących we krwi. Choroby neurodegeneracyjne, urazy i choroby autoimmunologiczne są związane z reakcją układu odpornościowego, która może mieć istotny wpływ na dalszy przebieg choroby i na tempo regeneracji tkanki. Polaryzacja makrofagów ma w związku z tym znaczenie w chorobach centralnego układu nerwowego. Aktywność komórek M1 i M2 może bowiem różnie wpływać na przeżywalność neuronów i oligodendrocytów, na wzrost aksonów, na proces demielinizacji czy na szczelność bariery krew–mózg. Wynika to z różnic między fenotypami w wytwarzaniu reaktywnych form tlenu i tlenku azotu, wydzielaniu cytokin i czynników wzrostu, bezpośrednich oddziaływaniach na sąsiednie komórki i zdolnościach do fagocytozy. W artykule omówiono to zagadnienie w: udarze mózgu, urazie rdzenia kręgowego, chorobie Alzheimera, stwardnieniu zanikowym bocznym i stwardnieniu rozsianym. W wielu spośród tych patologii obserwuje się gradient czasowy lub przestrzenny rozmieszczenia w tkance poszczególnych fenotypów mikrogleju i/lub makrofagów. Wydaje się zatem, że zmiany polaryzacji makrofagów mogą potencjalnie sprzyjać regeneracji tkanki lub hamować rozwój chorób neurodegeneracyjnych.
Collapse
|
13
|
Abstract
Mitochondria are considered to be the powerhouse of the cell. Normal functioning of the mitochondria is not only essential for cellular energy production but also for several immunomodulatory processes. Macrophages operate in metabolic niches and rely on rapid adaptation to specific metabolic conditions such as hypoxia, nutrient limitations, or reactive oxygen species to neutralize pathogens. In this regard, the fast reprogramming of mitochondrial metabolism is indispensable to provide the cells with the necessary energy and intermediates to efficiently mount the inflammatory response. Moreover, mitochondria act as a physical scaffold for several proteins involved in immune signaling cascades and their dysfunction is immediately associated with a dampened immune response. In this review, we put special focus on mitochondrial function in macrophages and highlight how mitochondrial metabolism is involved in macrophage activation.
Collapse
Affiliation(s)
- Mohamed Zakaria Nassef
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Brunswick, Germany
| | - Jasmin E Hanke
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Brunswick, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Brunswick, Germany
| |
Collapse
|
14
|
Jang T, Poplawska M, Cimpeanu E, Mo G, Dutta D, Lim SH. Vaso-occlusive crisis in sickle cell disease: a vicious cycle of secondary events. J Transl Med 2021; 19:397. [PMID: 34544432 PMCID: PMC8454100 DOI: 10.1186/s12967-021-03074-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
Painful vaso-occlusive crisis (VOC) remains the most common reason for presenting to the Emergency Department and hospitalization in patients with sickle cell disease (SCD). Although two new agents have been approved by the Food and Drug Administration for treating SCD, they both target to reduce the frequency of VOC. Results from studies investigating various approaches to treat and shorten VOC have so far been generally disappointing. In this paper, we will summarize the complex pathophysiology and downstream events of VOC and discuss the likely reasons for the disappointing results using monotherapy. We will put forward the rationale for exploring some of the currently available agents to either protect erythrocytes un-involved in the hemoglobin polymerization process from sickling induced by the secondary events, or a multipronged combination approach that targets the complex downstream pathways of VOC.
Collapse
Affiliation(s)
- Tim Jang
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA
| | - Maria Poplawska
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA
| | - Emanuela Cimpeanu
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA
| | - George Mo
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA
| | - Dibyendu Dutta
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA
| | - Seah H Lim
- Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Medical Center, SUNY Downstate Health Sciences University, 450 Clarkson Avenue, MSC #20, Brooklyn, NY, 11203, USA.
| |
Collapse
|
15
|
Ludtka C, Silberman J, Moore E, Allen JB. Macrophages in microgravity: the impact of space on immune cells. NPJ Microgravity 2021; 7:13. [PMID: 33790288 PMCID: PMC8012370 DOI: 10.1038/s41526-021-00141-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The effects of a microgravity environment on the myriad types of immune cells present within the human body have been assessed both by bench-scale simulation and suborbital methods, as well as in true spaceflight. Macrophages have garnered increased research interest in this context in recent years. Their functionality in both immune response and tissue remodeling makes them a unique cell to investigate in regards to gravisensitive effects as well as parameters of interest that could impact astronaut health. Here, we review and summarize the literature investigating the effects of microgravity on macrophages and monocytes regarding the microgravity environment simulation/generation methods, cell sources, experiment durations, and parameters of interest utilized within the field. We discuss reported findings on the impacts of microgravity on macrophage/monocyte structure, adhesion and migration, proliferation, genetic expression, cytokine secretion, and reactive oxygen species production, as well as polarization. Based on this body of data, we make recommendations to the field for careful consideration of experimental design to complement existing reports, as the multitude of disparate study methods previously published can make drawing direct comparisons difficult. However, the breadth of different testing methodologies can also lend itself to attempting to identify the most robust and consistent responses to microgravity across various testing conditions.
Collapse
Affiliation(s)
- Christopher Ludtka
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Justin Silberman
- Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Erika Moore
- Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Josephine B Allen
- Materials Science and Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Alexander ET, El Naggar O, Fahey E, Mariner K, Donnelly J, Wolfgang K, Phanstiel O, Gilmour SK. Harnessing the polyamine transport system to treat BRAF inhibitor-resistant melanoma. Cancer Biol Ther 2021; 22:225-237. [PMID: 33602034 DOI: 10.1080/15384047.2021.1883185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BRAF mutations are present in over half of all melanoma tumors. Although BRAF inhibitors significantly improve survival of patients with metastatic melanoma, recurrences occur within several months. We previously reported that BRAF mutant melanoma cells are more sensitive to a novel arylmethyl-polyamine (AP) compound that exploits their increased polyamine uptake compared to that of BRAF wildtype cells. Using an animal model of BRAF inhibitor-resistant melanoma, we show that co-treatment with the BRAF inhibitor, PLX4720, and AP significantly delays the recurrence of PLX4720-resistant melanoma tumors and decreases tumor-promoting macrophages. Development of BRAF inhibitor-resistance enriches for metastatic cancer stem cells (CSC) and increases tumor-promoting macrophages. In vitro studies demonstrated that CD304+, CXCR4+ spheroid cultures of BRAF mutant melanoma cells are resistant to PLX4720 but are more sensitive to AP compared to monolayer cultures of the same cells. AP significantly inhibited YUMM1.7 melanoma cell invasiveness across a Matrigel-coated filter using the CXCR4 ligand, SDF-1α, as the chemoattractant. AP also blocked the chemotactic effect of SDF-1α on CXCR4+ macrophages and inhibited M2 polarization of macrophages. In melanoma-macrophage co-cultures, AP prevented the PLX4720-induced release of pro-tumorigenic growth factors, such as VEGF, from macrophages and prevented the macrophage rescue of BRAF mutant melanoma cells treated with PLX4720. Our study offers a novel therapy (AP) to treat chemo-resistant melanoma. AP is unique because it targets the polyamine transport system in BRAF inhibitor-resistant CSCs and also blocks CXCR4 signaling in invasive melanoma cells and pro-tumorigenic macrophages.
Collapse
Affiliation(s)
| | | | - Erin Fahey
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - Kelsey Mariner
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - Julia Donnelly
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | | | - Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, Biomolecular Research Annex, Orlando, FL, USA
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, PA, USA.,At Lankenau Institute for Medical Research, Wynnewood, PA, USA
| |
Collapse
|
17
|
Marco M, Valentina I, Daniele M, Valerio DR, Andrea P, Roberto G, Laura G, Luigi U. Peripheral Arterial Disease in Persons with Diabetic Foot Ulceration: a Current Comprehensive Overview. Curr Diabetes Rev 2021; 17:474-485. [PMID: 33023453 DOI: 10.2174/1573399816999201001203111] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
In developed countries, the prevalence of persons with diabetes and peripheral arterial disease (PAD) is approximately 50%. The presence of PAD is associated with non-healing ulcers, major amputation, cardiovascular morbidity, and mortality. It is estimated that persons with diabetes, foot ulceration and PAD have 50% of 5-years mortality rate. Therefore, subjects with ischemic diabetic foot ulcers (DFUs) should be considered a special group of patients with specific clinical characteristics, general health status and prognosis. In persons with ischemic DFUs, an early diagnosis and treatment are mandatory to reduce the risk of worse outcomes such as major amputation. Revascularization of occluded lower extremity arteries is the main treatment to restore blood flow in the foot and promote wound healing. Nonetheless, there are several unmet needs in the management of diabetic subjects with PAD and foot ulceration as medical therapy, diagnostic criteria and indications for revascularization, revascularization strategy and technical approach as well as the management of no-option critical limb ischemia patients. It is a common opinion that there is an evolution of PAD features in diabetic persons, which seems to present a more aggressive pattern. This may be related to the frequent presence of concomitant comorbidities such as renal failure which could influence the characteristics of atherosclerotic plaques and their distribution. The aim of this review is to commence a complete overview and state of the art in the treatment of patients with diabetes, PAD, and foot ulceration and to describe the current challenges and future perspectives.
Collapse
Affiliation(s)
- Meloni Marco
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Izzo Valentina
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Morosetti Daniele
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Da Ros Valerio
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Panunzi Andrea
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Gandini Roberto
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Giurato Laura
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| | - Uccioli Luigi
- Department of Systems Medicine, University of Rome Tor Vegata, Rome, Italy
| |
Collapse
|
18
|
Costa DL, Amaral EP, Namasivayam S, Mittereder LR, Fisher L, Bonfim CC, Sardinha-Silva A, Thompson RW, Hieny SE, Andrade BB, Sher A. Heme oxygenase-1 inhibition promotes IFNγ- and NOS2-mediated control of Mycobacterium tuberculosis infection. Mucosal Immunol 2021; 14:253-266. [PMID: 32862202 PMCID: PMC7796944 DOI: 10.1038/s41385-020-00342-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/17/2020] [Accepted: 08/12/2020] [Indexed: 02/04/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infection induces pulmonary expression of the heme-degrading enzyme heme oxygenase-1 (HO-1). We have previously shown that pharmacological inhibition of HO-1 activity in experimental tuberculosis results in decreased bacterial loads and unexpectedly that this outcome depends on the presence of T lymphocytes. Here, we extend these findings by demonstrating that IFNγ production by T lymphocytes and NOS2 expression underlie this T-cell requirement and that HO-1 inhibition potentiates IFNγ-induced NOS2-dependent control of Mtb by macrophages in vitro. Among the products of heme degradation by HO-1 (biliverdin, carbon monoxide, and iron), only iron supplementation reverted the HO-1 inhibition-induced enhancement of bacterial control and this reversal was associated with decreased NOS2 expression and NO production. In addition, we found that HO-1 inhibition results in decreased labile iron levels in Mtb-infected macrophages in vitro and diminished iron accumulation in Mtb-infected lungs in vivo. Together these results suggest that the T-lymphocyte dependence of the therapeutic outcome of HO-1 inhibition on Mtb infection reflects the role of the enzyme in generating iron that suppresses T-cell-mediated IFNγ/NOS2-dependent bacterial control. In broader terms, our findings highlight the importance of the crosstalk between iron metabolism and adaptive immunity in determining the outcome of infection.
Collapse
Affiliation(s)
- Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,* Diego L Costa current affiliation: Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lara R Mittereder
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Logan Fisher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Caio C Bonfim
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aline Sardinha-Silva
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert W Thompson
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara E Hieny
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bruno B Andrade
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, 7925, South Africa,Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil,Curso de Medicina, Faculdade de Tecnologia e Ciências (FTC), Salvador, Brazil,Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil,Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Zhu S, Luo Z, Li X, Han X, Shi S, Zhang T. Tumor-associated macrophages: role in tumorigenesis and immunotherapy implications. J Cancer 2021; 12:54-64. [PMID: 33391402 PMCID: PMC7738842 DOI: 10.7150/jca.49692] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated macrophages (TAMs) occupy an important position in the tumor microenvironment (TME), they are a highly plastic heterogeneous population with complex effects on tumorigenesis and development. TAMs secrete a variety of cytokines, chemokines, and proteases, which promote the remodeling of extracellular matrix, tumor cell growth and metastasis, tumor vessel and lymphangiogenesis, and immunosuppression. TAMs with different phenotypes have different effects on tumor proliferation and metastasis. TAMs act a pivotal part in occurrence and development of tumors, and are very attractive target to inhibit tumor growth and metastasis in tumor immunotherapy. This article reviews the interrelationship between TAMs and tumor microenvironment and its related applications in tumor therapy.
Collapse
Affiliation(s)
- Shunyao Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyi Luo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xixi Li
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi Han
- Xiaoshan Hosptital of Traditional Chinese Medicine, Hangzhou 311201, China
| | - Senlin Shi
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ting Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
20
|
PlGF Immunological Impact during Pregnancy. Int J Mol Sci 2020; 21:ijms21228714. [PMID: 33218096 PMCID: PMC7698813 DOI: 10.3390/ijms21228714] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother’s immune system has to tolerate the persistence of paternal alloantigens without affecting the anti-infectious immune response. Consequently, several mechanisms aimed at preventing allograft rejection, occur during a pregnancy. In fact, the early stages of pregnancy are characterized by the correct balance between inflammation and immune tolerance, in which proinflammatory cytokines contribute to both the remodeling of tissues and to neo-angiogenesis, thus, favoring the correct embryo implantation. In addition to the creation of a microenvironment able to support both immunological privilege and angiogenesis, the trophoblast invades normal tissues by sharing the same behavior of invasive tumors. Next, the activation of an immunosuppressive phase, characterized by an increase in the number of regulatory T (Treg) cells prevents excessive inflammation and avoids fetal immuno-mediated rejection. When these changes do not occur or occur incompletely, early pregnancy failure follows. All these events are characterized by an increase in different growth factors and cytokines, among which one of the most important is the angiogenic growth factor, namely placental growth factor (PlGF). PlGF is initially isolated from the human placenta. It is upregulated during both pregnancy and inflammation. In this review, we summarize current knowledge on the immunomodulatory effects of PlGF during pregnancy, warranting that both innate and adaptive immune cells properly support the early events of implantation and placental development. Furthermore, we highlight how an alteration of the immune response, associated with PlGF imbalance, can induce a hypertensive state and lead to the pre-eclampsia (PE).
Collapse
|
21
|
Abstract
Traumatic brain injury is among the leading causes of death in individuals under 45 years of age. However, since trauma mechanisms and survival times differ enormously, the exact mechanisms leading to the primary and secondary injury and eventually to death after traumatic brain injury (TBI) remain unclear. Several studies showed the versatile functions of microglia, the innate macrophages of the brain, following a TBI. Earlier being characterized as rather neurotoxic, neuroprotective capacities were recently demonstrated, therefore, making microglia one of the key players following TBI. Especially in cases with only short survival times, immediate microglial reactions are of great forensic interest in questions of wound age estimation. Using standardized immunohistochemical methods, we examined 8 cases which died causatively of TBI with survival times between minutes and 7 days and 5 control cases with cardiovascular failure as the cause of death to determine acute changes in microglial morphology and antigen expression after TBI. In this pilot study, we detected highly localized changes in microglial morphology already early after traumatic damage, e.g., activated microglia and phagocyted erythrocytes in the contusion areas in cases with minute survival. Furthermore, an altered antigen expression was observed with increasing trauma wound age, showing similar effects like earlier transcriptomic studies. There is minute data on the direct impact of shear forces on microglial morphology. We were able to show localization-depending effects on microglial morphology causing localized dystrophy and adjacent activation. While rodent studies are widespread, they fail to mimic the exact mechanisms in human TBI response. Therefore, more studies focusing on cadaveric samples need to follow to thoroughly define the mechanisms leading to cell destruction and eventually evaluate their forensic value.
Collapse
|
22
|
Min Y, Yan L, Wang Q, Wang F, Hua H, Yuan Y, Jin H, Zhang M, Zhao Y, Yang J, Jiang X, Yang Y, Li F. Distinct Residential and Infiltrated Macrophage Populations and Their Phagocytic Function in Mild and Severe Neonatal Hypoxic-Ischemic Brain Damage. Front Cell Neurosci 2020; 14:244. [PMID: 32903800 PMCID: PMC7438904 DOI: 10.3389/fncel.2020.00244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/15/2020] [Indexed: 01/10/2023] Open
Abstract
Neonatal brain injury, especially severe injury induced by hypoxia-ischemia (HI), causes mortality and long-term neurological impairments. Our previous study demonstrated activation of CD11b+ myeloid cells, including residential microglial cells (MGs) and infiltrating monocyte-derived macrophages (MDMs) in a murine model of hypoxic-ischemic brain damage (HIBD), with unknown functions. Here, we study the differences in the phagocytic function of MGs and MDMs to clarify their potential roles after HIBD. HI was induced in 9-10-day postnatal mice. On days 1 and 3 after injury, pathological and neurobehavioral tests were performed to categorize the brain damage as mild or severe. Flow cytometry was applied to quantify the dynamic change in the numbers of MGs and MDMs according to the relative expression level of CD45 in CD11b+ cells. CX3CR1 GFPCCR2 RFP double-transformed mice were used to identify MGs and MDMs in the brain parenchyma after HIBD. Lysosome-associated membrane protein 1 (LAMP1), toll-like receptor 2 (TLR2), CD36, and transforming growth factor (TGF-β) expression levels were measured to assess the underlying function of phagocytes and neuroprotective factors in these cells. The FITC-dextran 40 phagocytosis assay was applied to examine the change in phagocytic function under oxygen-glucose deprivation (OGD) in vitro. We found that neonatal HI induced a different degree of brain damage: mild or severe injury. Compared with mildly injured animals, mice with severe injury had lower weight, worse neurobehavioral scores, and abnormal brain morphology. In a severely injured brain, CD11b+ cells remarkably increased, including an increase in the MDM population and a decrease in the MG population. Furthermore, MDM infiltration into the brain parenchyma was evident in CX3CR1 GFPCCR2 RFP double-transformed mice. Mild and severe brain injury caused different phagocytosis-related responses and neuroprotective functions of MDMs and MGs at 1 and 3 days following HI. The phagocytic function was activated in BV2 cells but downregulated in Raw264.7 cells under OGD in vitro. These observations indicate that neonatal HI induced different degrees of brain injury. The proportion of infiltrated macrophage MDMs was increased and they were recruited into the injured brain parenchyma in severe brain injury. The resident macrophage MGs proportion decreased and maintained activated phagocytic function in both mild and severe brain injury, and restored neuroprotective function in severe brain injury.
Collapse
Affiliation(s)
- Yingjun Min
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Lin Yan
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Qian Wang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Fang Wang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Hairong Hua
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yun Yuan
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Huiyan Jin
- Department of Functional Experiment, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Ming Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China
| | - Yaling Zhao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianzhong Yang
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiangning Jiang
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Yuan Yang
- Department of Physiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Fan Li
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| |
Collapse
|
23
|
Transcriptional, Epigenetic and Metabolic Programming of Tumor-Associated Macrophages. Cancers (Basel) 2020; 12:cancers12061411. [PMID: 32486098 PMCID: PMC7352439 DOI: 10.3390/cancers12061411] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/16/2020] [Accepted: 05/17/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages are key innate immune cells in the tumor microenvironment (TME) that regulate primary tumor growth, vascularization, metastatic spread and tumor response to various types of therapies. The present review highlights the mechanisms of macrophage programming in tumor microenvironments that act on the transcriptional, epigenetic and metabolic levels. We summarize the latest knowledge on the types of transcriptional factors and epigenetic enzymes that control the direction of macrophage functional polarization and their pro- and anti-tumor activities. We also focus on the major types of metabolic programs of macrophages (glycolysis and fatty acid oxidation), and their interaction with cancer cells and complex TME. We have discussed how the regulation of macrophage polarization on the transcriptional, epigenetic and metabolic levels can be used for the efficient therapeutic manipulation of macrophage functions in cancer.
Collapse
|
24
|
Lier J, Ondruschka B, Bechmann I, Dreßler J. Fast microglial activation after severe traumatic brain injuries. Int J Legal Med 2020; 134:2187-2193. [PMID: 32372233 PMCID: PMC7578125 DOI: 10.1007/s00414-020-02308-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury is among the leading causes of death in individuals under 45 years of age. However, since trauma mechanisms and survival times differ enormously, the exact mechanisms leading to the primary and secondary injury and eventually to death after traumatic brain injury (TBI) remain unclear. Several studies showed the versatile functions of microglia, the innate macrophages of the brain, following a TBI. Earlier being characterized as rather neurotoxic, neuroprotective capacities were recently demonstrated, therefore, making microglia one of the key players following TBI. Especially in cases with only short survival times, immediate microglial reactions are of great forensic interest in questions of wound age estimation. Using standardized immunohistochemical methods, we examined 8 cases which died causatively of TBI with survival times between minutes and 7 days and 5 control cases with cardiovascular failure as the cause of death to determine acute changes in microglial morphology and antigen expression after TBI. In this pilot study, we detected highly localized changes in microglial morphology already early after traumatic damage, e.g., activated microglia and phagocyted erythrocytes in the contusion areas in cases with minute survival. Furthermore, an altered antigen expression was observed with increasing trauma wound age, showing similar effects like earlier transcriptomic studies. There is minute data on the direct impact of shear forces on microglial morphology. We were able to show localization-depending effects on microglial morphology causing localized dystrophy and adjacent activation. While rodent studies are widespread, they fail to mimic the exact mechanisms in human TBI response. Therefore, more studies focusing on cadaveric samples need to follow to thoroughly define the mechanisms leading to cell destruction and eventually evaluate their forensic value.
Collapse
Affiliation(s)
- Julia Lier
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, D-04103, Leipzig, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University of Leipzig, Johannisallee 28, D-04103, Leipzig, Germany.
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, D-04103, Leipzig, Germany
| | - Jan Dreßler
- Institute of Legal Medicine, University of Leipzig, Johannisallee 28, D-04103, Leipzig, Germany
| |
Collapse
|
25
|
Feng Z, Yu Q, Zhang T, Tie W, Li J, Zhou X. Updates on mechanistic insights and targeting of tumour metastasis. J Cell Mol Med 2020; 24:2076-2086. [PMID: 31957271 PMCID: PMC7011147 DOI: 10.1111/jcmm.14931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/15/2019] [Accepted: 12/16/2019] [Indexed: 02/05/2023] Open
Abstract
Malignant tumours are one of the major diseases that seriously endanger human health. The characteristics of their invasion and metastasis are one of the main causes of death in cancer patients, and these features cannot be separated from the participation of various molecules-related cells living in the tumour microenvironment and specific structures. Tumour invasion can approximately be divided into several specific steps according to the movement of tumour cells. In each step, there are different actions in the tumour microenvironment that mediate the interactions among substances. Researchers are attempting to clarify every mechanism of the tumour dissemination. However, there is still a long way to the final determination. Here, we review these interactions in tumour invasion and metastasis at the structural, molecular and cellular levels. We also discuss the ongoing studies and the promise of targeting metastasis in tumour therapy.
Collapse
Affiliation(s)
- Zeru Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Qiuxuan Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Ting Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Wanpeng Tie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Zhao C, Mirando AC, Sové RJ, Medeiros TX, Annex BH, Popel AS. A mechanistic integrative computational model of macrophage polarization: Implications in human pathophysiology. PLoS Comput Biol 2019; 15:e1007468. [PMID: 31738746 PMCID: PMC6860420 DOI: 10.1371/journal.pcbi.1007468] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
Macrophages respond to signals in the microenvironment by changing their functional phenotypes, a process known as polarization. Depending on the context, they acquire different patterns of transcriptional activation, cytokine expression and cellular metabolism which collectively constitute a continuous spectrum of phenotypes, of which the two extremes are denoted as classical (M1) and alternative (M2) activation. To quantitatively decode the underlying principles governing macrophage phenotypic polarization and thereby harness its therapeutic potential in human diseases, a systems-level approach is needed given the multitude of signaling pathways and intracellular regulation involved. Here we develop the first mechanism-based, multi-pathway computational model that describes the integrated signal transduction and macrophage programming under M1 (IFN-γ), M2 (IL-4) and cell stress (hypoxia) stimulation. Our model was calibrated extensively against experimental data, and we mechanistically elucidated several signature feedbacks behind the M1-M2 antagonism and investigated the dynamical shaping of macrophage phenotypes within the M1-M2 spectrum. Model sensitivity analysis also revealed key molecular nodes and interactions as targets with potential therapeutic values for the pathophysiology of peripheral arterial disease and cancer. Through simulations that dynamically capture the signal integration and phenotypic marker expression in the differential macrophage polarization responses, our model provides an important computational basis toward a more quantitative and network-centric understanding of the complex physiology and versatile functions of macrophages in human diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| | - Adam C. Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard J. Sové
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Thalyta X. Medeiros
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- Divison of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Brian H. Annex
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- Divison of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
27
|
Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, Fang W, Xiong X. The Involvement and Therapy Target of Immune Cells After Ischemic Stroke. Front Immunol 2019; 10:2167. [PMID: 31572378 PMCID: PMC6749156 DOI: 10.3389/fimmu.2019.02167] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
After ischemic stroke, the integrity of the blood-brain barrier is compromised. Peripheral immune cells, including neutrophils, T cells, B cells, dendritic cells, and macrophages, infiltrate into the ischemic brain tissue and play an important role in regulating the progression of ischemic brain injury. In this review, we will discuss the role of different immune cells after stroke in the secondary inflammatory reaction and focus on the phenotypes and functions of macrophages in ischemic stroke, as well as briefly introduce the anti-ischemic stroke therapy targeting macrophages.
Collapse
Affiliation(s)
- Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, China
| | - Xiqun Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daniel Smerin
- Department of Neurosurgery, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Jiang S, Xu Y. Annexin A2 upregulation protects human retinal endothelial cells from oxygen-glucose deprivation injury by activating autophagy. Exp Ther Med 2019; 18:2901-2908. [PMID: 31572534 PMCID: PMC6755473 DOI: 10.3892/etm.2019.7909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
Retinal neovascularization is a common pathological change in multiple diseases of the eyes and the upregulation of annexin A2 (A2) under a hypoxic and ischemic microenvironment has been demonstrated to be a key factor in the pathological process. However, the underlying mechanism by which A2 regulates retinal neovascularization remains unclear. In the present study, oxygen-glucose deprivation (OGD) was used to mimic the hypoxic and ischemic microenvironment, to observe the role of A2 in retinal neovascularization regulation by focusing on autophagy. The results showed that OGD treatment significantly increased the mRNA and protein levels of A2 in human retinal endothelial cells (HRECs), which was dependent on activation of hypoxia inducible factor (HIF)-1α signaling. The OGD-induced activation of autophagy was attenuated when A2 was silenced, but increased when A2 was overexpressed, suggesting that A2 upregulation contributed to OGD-induced cell autophagy activation. Furthermore, knockdown of A2 decreased cell viability and promoted cell apoptosis under OGD conditions. Overexpression of A2 increased cell viability and reduced cell apoptosis under OGD conditions, and inhibiting autophagy using an inhibitor, reversed these changes, suggesting that upregulation of A2 by OGD serves a cytoprotective role by inducing cell autophagy in HRECs. Taken together, the results of the present study suggested that promoting retinal endothelial cell survival by autophagy activation via the HIF-1α signaling pathway in a hypoxic and ischemic microenvironment may underlie the mechanism by which A2 regulates retinal neovascularization. The present study is the first study to demonstrate the novel role of A2 during retinal neovascularization under pathological conditions, to the best of our knowledge. Therefore, A2 may serve as a potential therapeutic target for treating neovascularization-associated conditions of the eye.
Collapse
Affiliation(s)
- Shule Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yile Xu
- Department of Ophthalmology, The Hangzhou First People's Hospital, Hangzhou, Zhejiang 310001, P.R. China
| |
Collapse
|
29
|
Albonici L, Giganti MG, Modesti A, Manzari V, Bei R. Multifaceted Role of the Placental Growth Factor (PlGF) in the Antitumor Immune Response and Cancer Progression. Int J Mol Sci 2019; 20:ijms20122970. [PMID: 31216652 PMCID: PMC6627047 DOI: 10.3390/ijms20122970] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022] Open
Abstract
The sharing of molecules function that affects both tumor growth and neoangiogenesis with cells of the immune system creates a mutual interplay that impairs the host’s immune response against tumor progression. Increasing evidence shows that tumors are able to create an immunosuppressive microenvironment by recruiting specific immune cells. Moreover, molecules produced by tumor and inflammatory cells in the tumor microenvironment create an immunosuppressive milieu able to inhibit the development of an efficient immune response against cancer cells and thus fostering tumor growth and progression. In addition, the immunoediting could select cancer cells that are less immunogenic or more resistant to lysis. In this review, we summarize recent findings regarding the immunomodulatory effects and cancer progression of the angiogenic growth factor namely placental growth factor (PlGF) and address the biological complex effects of this cytokine. Different pathways of the innate and adaptive immune response in which, directly or indirectly, PlGF is involved in promoting tumor immune escape and metastasis will be described. PlGF is important for building up vascular structures and functions. Although PlGF effects on vascular and tumor growth have been widely summarized, its functions in modulating the immune intra-tumoral microenvironment have been less highlighted. In agreement with PlGF functions, different antitumor strategies can be envisioned.
Collapse
Affiliation(s)
- Loredana Albonici
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
30
|
IL-1 beta-mediated macrophage-hepatocyte crosstalk upregulates hepcidin under physiological low oxygen levels. Redox Biol 2019; 24:101209. [PMID: 31108461 PMCID: PMC6526398 DOI: 10.1016/j.redox.2019.101209] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/19/2022] Open
Abstract
In mammals, the iron masterswitch hepcidin efficiently controls iron recycling by the macrophage-liver axis but the exact interplay between macrophages and hepatocytes remains poorly understood. We here study hepcidin response during macrophage differentiation as well as the macrophage-hepatocyte crosstalk and its subsequent effects on hepatocyte hepcidin using an in vitro co-culture model that mimics the physiological liver microenvironment. We show that macrophage differentiation strongly induces hepcidin by 60-fold both in THP1 macrophages and primary isolated monocyte-derived macrophages. Removal of H2O2 by catalase or inhibition of NOX2 efficiently blocked hepcidin induction. After differentiation, macrophage hepcidin accounted for 10% of total hepatocyte hepcidin and did not respond to low oxygen levels. In contrast, co-culture of differentiated macrophages with Huh7 cells significantly induced hepatocyte hepcidin, which was further potentiated under low oxygen levels. Hepatocyte hepcidin was also upregulated when Huh7 cells were solely exposed to macrophage-conditioned hypoxic medium. A cytokine screen identified macrophage secreted IL-1β as major inducer of hepcidin in hepatocytes. In confirmation, treatment of Huh7 cells with the IL-1 receptor antagonist (anakinra) completely blunted macrophage-mediated hepcidin transcription in hepatocytes. Finally, detailed analysis of potentially involved signaling pathways points toward STAT3 and CEBPδ-mediated hepcidin induction independent of IL-6. In conclusion, our study demonstrates a strong NOX2-mediated hepcidin induction during macrophage differentiation. These differentiated macrophages are able to efficiently induce hepatocyte hepcidin mainly through secretion of IL-1β. Our data highlight a hitherto unrecognized role of macrophage-hepatocyte crosstalk for a joint and oxygen-dependent hepcidin production through STAT3 and CEBPδ.
Hepcidin is strongly induced during NOX2-mediated macrophage differentiation in a H2O2-dependent manner. In contrast to hepatocyte hepcidin, macrophage hepcidin transcription is not modulated by low O2 level. Macrophage released IL1-β strongly induces hepatocyte hepcidin via STAT3 signaling. IL1-β mediated hepatocyte hepcidin induction is independent of IL-6. Despite the mandatory requirement of STAT3, CEBPδ also involved in IL1-β induced hepatocyte hepcidin transcription.
Collapse
|
31
|
Expression of Hypoxia-Inducible Factor 1α (HIF-1α) and Genes of Related Pathways in Altered Gravity. Int J Mol Sci 2019; 20:ijms20020436. [PMID: 30669540 PMCID: PMC6358763 DOI: 10.3390/ijms20020436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 12/28/2022] Open
Abstract
Immune system deterioration in space represents a major risk, which has to be mitigated for exploration-class missions into the solar system. Altered gravitational forces have been shown to regulate adaptation processes in cells of the immune system, which are important for appropriate risk management, monitoring and development of countermeasures. T lymphocytes and cells of the monocyte-macrophage system are highly migratory cell types that frequently encounter a wide range of oxygen tensions in human tissues and in hypoxic areas, even under homeostatic conditions. Hypoxia-inducible factor 1 and 2 (HIF's) might have an important role in activation of T cells and cells of the monocyte-macrophages system. Thus, we investigated the regulation of HIF-dependent and, therefore, hypoxia-signaling systems in both cell types in altered gravity and performed transcript and protein analysis from parabolic flight and suborbital ballistic rocket experiments. We found that HIF-1α and HIF-1-dependent transcripts were differently regulated in altered gravity, whereas HIF-1α-dependent gene expression adapted after 5 min microgravity. Inter-platform comparisons identified PDK1 as highly responsive to gravitational changes in human U937 myelomonocytic cells and in Jurkat T cells. We suggest HIF-1 as a potential pharmacological target for counteracting immune system deterioration during space flight.
Collapse
|
32
|
Lin W, Hsuan YCY, Lin MT, Kuo TW, Lin CH, Su YC, Niu KC, Chang CP, Lin HJ. Human Umbilical Cord Mesenchymal Stem Cells Preserve Adult Newborn Neurons and Reduce Neurological Injury after Cerebral Ischemia by Reducing the Number of Hypertrophic Microglia/Macrophages. Cell Transplant 2018; 26:1798-1810. [PMID: 29338384 PMCID: PMC5784525 DOI: 10.1177/0963689717728936] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Microglia are the first source of a neuroinflammatory cascade, which seems to be involved in every phase of stroke-related neuronal damage. Two weeks after transient middle cerebral artery occlusion (MCAO), vehicle-treated rats displayed higher numbers of total ionized calcium-binding adaptor molecule 1 (Iba-1)-positive cells, greater cell body areas of Iba-1-positive cells, and higher numbers of hypertrophic Iba-1-positive cells (with a cell body area over 80 μm2) in the ipsilateral ischemic brain regions including the frontal cortex, striatum, and parietal cortex. In addition, MCAO decreased the number of migrating neuroblasts (or DCX- and 5-ethynyl-2′-deoxyuridine-positive cells) in the cortex, subventricular zone, and hippocampus of the ischemic brain, followed by neurological injury (including brain infarct and neurological deficits). Intravenous administration of human umbilical cord–derived mesenchymal stem cells (hUC-MSCs; 1 × 106 or 4 × 106) at 24 h after MCAO reduced neurological injury, decreased the number of hypertrophic microglia/macrophages, and increased the number of newborn neurons in rat brains. Thus, the accumulation of hypertrophic microglia/macrophages seems to be detrimental to neurogenesis after stroke. Treatment with hUC-MSCs preserved adult newborn neurons and reduced functional impairment after transient cerebral ischemia by reducing the number of hypertrophic microglia/macrophages.
Collapse
Affiliation(s)
- Willie Lin
- 1 Meridigen Biotech Co., Ltd., Neihu, Taipei City, Taiwan
| | | | - Mao-Tsun Lin
- 2 Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ting-Wei Kuo
- 3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan
| | | | - Yu-Chin Su
- 1 Meridigen Biotech Co., Ltd., Neihu, Taipei City, Taiwan
| | - Ko-Chi Niu
- 4 Department of Hyperbaric Oxygen, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ching-Ping Chang
- 2 Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan.,3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan.,5 The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Hung-Jung Lin
- 3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan.,6 Department of Emergency Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| |
Collapse
|
33
|
Billin AN, Honeycutt SE, McDougal AV, Kerr JP, Chen Z, Freudenberg JM, Rajpal DK, Luo G, Kramer HF, Geske RS, Fang F, Yao B, Clark RV, Lepore J, Cobitz A, Miller R, Nosaka K, Hinken AC, Russell AJ. HIF prolyl hydroxylase inhibition protects skeletal muscle from eccentric contraction-induced injury. Skelet Muscle 2018; 8:35. [PMID: 30424786 PMCID: PMC6234580 DOI: 10.1186/s13395-018-0179-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/14/2018] [Indexed: 12/23/2022] Open
Abstract
Background In muscular dystrophy and old age, skeletal muscle repair is compromised leading to fibrosis and fatty tissue accumulation. Therefore, therapies that protect skeletal muscle or enhance repair would be valuable medical treatments. Hypoxia-inducible factors (HIFs) regulate gene transcription under conditions of low oxygen, and HIF target genes EPO and VEGF have been associated with muscle protection and repair. We tested the importance of HIF activation following skeletal muscle injury, in both a murine model and human volunteers, using prolyl hydroxylase inhibitors that stabilize and activate HIF. Methods Using a mouse eccentric limb injury model, we characterized the protective effects of prolyl hydroxylase inhibitor, GSK1120360A. We then extended these studies to examine the impact of EPO modulation and infiltrating immune cell populations on muscle protection. Finally, we extended this study with an experimental medicine approach using eccentric arm exercise in untrained volunteers to measure the muscle-protective effects of a clinical prolyl hydroxylase inhibitor, daprodustat. Results GSK1120360A dramatically prevented functional deficits and histological damage, while accelerating recovery after eccentric limb injury in mice. Surprisingly, this effect was independent of EPO, but required myeloid HIF1α-mediated iNOS activity. Treatment of healthy human volunteers with high-dose daprodustat reduced accumulation of circulating damage markers following eccentric arm exercise, although we did not observe any diminution of functional deficits with compound treatment. Conclusion The results of these experiments highlight a novel skeletal muscle protective effect of prolyl hydroxylase inhibition via HIF-mediated expression of iNOS in macrophages. Partial recapitulation of these findings in healthy volunteers suggests elements of consistent pharmacology compared to responses in mice although there are clear differences between these two systems. Electronic supplementary material The online version of this article (10.1186/s13395-018-0179-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew N Billin
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Samuel E Honeycutt
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Alan V McDougal
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Jaclyn P Kerr
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Zhe Chen
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | | | | | - Guizhen Luo
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Henning Fritz Kramer
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Robert S Geske
- Target Sciences, GlaxoSmithKline, King of Prussia, PA, USA
| | - Frank Fang
- Clinical Statistics, GlaxoSmithKline, King of Prussia, PA, USA
| | - Bert Yao
- Metabolic Pathways and Cardiovascular Therapy Area, GlaxoSmithKline, King of Prussia, PA, USA
| | - Richard V Clark
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - John Lepore
- Metabolic Pathways and Cardiovascular Therapy Area, GlaxoSmithKline, King of Prussia, PA, USA
| | - Alex Cobitz
- Metabolic Pathways and Cardiovascular Therapy Area, GlaxoSmithKline, King of Prussia, PA, USA
| | - Ram Miller
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Kazunori Nosaka
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Aaron C Hinken
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA
| | - Alan J Russell
- Muscle Metabolism Discovery Performance Unit, GlaxoSmithKline, King of Prussia, PA, USA.
| |
Collapse
|
34
|
Inhibition of prolyl hydroxylase domain proteins selectively enhances venous thrombus neovascularisation. Thromb Res 2018; 169:105-112. [PMID: 30031289 DOI: 10.1016/j.thromres.2018.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/30/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Hypoxia within acute venous thrombi is thought to drive resolution through stabilisation of hypoxia inducible factor 1 alpha (HIF1α). Prolyl hydroxylase domain (PHD) isoforms are critical regulators of HIF1α stability. Non-selective inhibition of PHD isoforms with l-mimosine has been shown to increase HIF1α stabilisation and promote thrombus resolution. OBJECTIVE The aim of this study was to investigate the therapeutic potential of PHD inhibition in venous thrombus resolution. METHODS Thrombosis was induced in the inferior vena cava of mice using a combination of flow restriction and endothelial activation. Gene and protein expression of PHD isoforms in the resolving thrombus was measured by RT-PCR and immunohistochemistry. Thrombus resolution was quantified in mice treated with pan PHD inhibitors AKB-4924 and JNJ-42041935 or inducible all-cell Phd2 knockouts by micro-computed tomography, 3D high frequency ultrasound or endpoint histology. RESULTS Resolving venous thrombi demonstrated significant temporal gene expression profiles for PHD2 and PHD3 (P < 0.05), but not for PHD1. PHD isoform protein expression was localised to early and late inflammatory cell infiltrates. Treatment with selective pan PHD inhibitors, AKB-4924 and JNJ-42041935, enhanced thrombus neovascularisation (P < 0.05), but had no significant effect on overall thrombus resolution. Thrombus resolution or its markers, macrophage accumulation and neovascularisation, did not differ significantly in inducible all-cell homozygous Phd2 knockouts compared with littermate controls (P > 0.05). CONCLUSIONS This data suggests that PHD-mediated thrombus neovascularisation has a limited role in the resolution of venous thrombi. Directly targeting angiogenesis alone may not be a viable therapeutic strategy to enhance venous thrombus resolution.
Collapse
|
35
|
Tuo Y, Xiang M. mTOR: A double‐edged sword for diabetes. J Leukoc Biol 2018; 106:385-395. [DOI: 10.1002/jlb.3mr0317-095rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/05/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yali Tuo
- Department of PharmacologySchool of PharmacyTongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Ming Xiang
- Department of PharmacologySchool of PharmacyTongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| |
Collapse
|
36
|
Lee JH, Kim K, Jo YH, Hwang JE, Chung HJ, Yang C. Reoxygenation speed and its implication for cellular injury responses in hypoxic RAW 264.7 cells. J Surg Res 2018; 227:88-94. [PMID: 29804868 DOI: 10.1016/j.jss.2017.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/12/2017] [Accepted: 11/03/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Ischemia/reperfusion injury is characterized by excess generation of reactive oxygen species (ROS). The purpose of this study is to test the effect of reoxygenation speed on ROS production and the cellular injury responses in hypoxic macrophages RAW 264.7 cells and its potential mechanisms for the generation of ROS. MATERIALS AND METHODS After hypoxic exposure of RAW 264.7 cells for 20 h, reoxygenation was performed for 6 h by stepwise increase in oxygen concentration (0.8% increase of oxygen every 15 min) in the slow reoxygenation (SRox) group or by moving the culture flasks quickly to a normoxic incubator in the rapid reoxygenation (RRox) group. To identify the potential effect of reoxygenation speed on the generation of ROS, the cells were pretreated with apocynin, VAS2870, and MitoTEMPO before the induction of hypoxia. RESULTS SRox significantly decreased cell death and cytotoxicity compared with RRox (P < 0.05). RRox resulted in significantly more generation of ROS, interleukin-1β, interleukin-6, and nitric oxide than SRox (P < 0.05). SRox also increased the expression of prosurvival proteins and decreased apoptosis. In cells pretreated with VAS2870 or MitoTEMPO, the reduced ROS generation by SRox was maintained. However, pretreatment with apocynin abolished the effect of reoxygenation speed on ROS generation. CONCLUSIONS SRox compared with RRox decreased cellular injury in hypoxic RAW 264.7 cells by decreasing ROS and inflammatory cytokine production and decreasing apoptosis.
Collapse
Affiliation(s)
- Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Kyuseok Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea.
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Ji Eun Hwang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Hea Jin Chung
- Department of Emergency Medicine, Emergency Care Center, Soonchunhyang University Hospital, Seoul, Republic of Korea
| | - Chungmi Yang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| |
Collapse
|
37
|
Ebersole JL, Novak MJ, Orraca L, Martinez-Gonzalez J, Kirakodu S, Chen KC, Stromberg A, Gonzalez OA. Hypoxia-inducible transcription factors, HIF1A and HIF2A, increase in aging mucosal tissues. Immunology 2018; 154:452-464. [PMID: 29338076 DOI: 10.1111/imm.12894] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023] Open
Abstract
Hypoxia (i.e. oxygen deprivation) activates the hypoxia-signalling pathway, primarily via hypoxia-inducible transcription factors (HIF) for numerous target genes, which mediate angiogenesis, metabolism and coagulation, among other processes to try to replenish tissues with blood and oxygen. Hypoxia signalling dysregulation also commonly occurs during chronic inflammation. We sampled gingival tissues from rhesus monkeys (Macaca mulatta; 3-25 years old) and total RNA was isolated for microarray analysis. HIF1A, HIF1B and HIF2A were significantly different in healthy aged tissues, and both HIF1A and HIF3A were positively correlated with aging. Beyond these transcription factor alterations, analysis of patterns of gene expression involved in hypoxic changes in tissues showed specific increases in metabolic pathway hypoxia-inducible genes, whereas angiogenesis pathway gene changes were more variable in healthy aging tissues across the animals. With periodontitis, aging tissues showed decreases in metabolic gene expression related to carbohydrate/lipid utilization (GBE1, PGAP1, TPI1), energy metabolism and cell cycle regulation (IER3, CCNG2, PER1), with up-regulation of transcription genes and cellular proliferation genes (FOS, EGR1, MET, JMJD6) that are hypoxia-inducible. The potential clinical implications of these results are related to the epidemiological findings of increased susceptibility and expression of periodontitis with aging. More specifically the findings describe that hypoxic stress may exist in aging gingival tissues before documentation of clinical changes of periodontitis and, so, may provide an explanatory molecular risk factor for an elevated capacity of the tissues to express destructive processes in response to changes in the microbial biofilms characteristic of a more pathogenic microbial challenge.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Michael John Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Luis Orraca
- School of Dentistry, University of Puerto Rico, Sabana Seca, PR, USA
| | | | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Kuey C Chen
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Arnold Stromberg
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Octavio A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
38
|
Hwang GS, Chen CC, Chou JC, Chang LL, Kan SF, Lai WH, Lieu FK, Hu S, Wang PS, Wang SW. Stimulatory Effect of Intermittent Hypoxia on the Production of Corticosterone by Zona Fasciculata-Reticularis Cells in Rats. Sci Rep 2017; 7:9035. [PMID: 28831034 PMCID: PMC5567345 DOI: 10.1038/s41598-017-07054-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
Hypoxia or intermittent hypoxia (IH) have known to alter both synthesis and secretion of hormones. However, the effect of IH on the production of adrenal cortical steroid hormones is still unclear. The aim of present study was to explore the mechanism involved in the effect of IH on the production of corticosterone by rat ZFR cells. Male rats were exposed at 12% O2 and 88% N2 (8 hours per day) for 1, 2, or 4 days. The ZFR cells were incubated at 37 °C for 1 hour with or without ACTH, 8-Br-cAMP, calcium ion channel blockers, or steroidogenic precursors. The concentration of plasma corticosterone was increased time-dependently by administration of IH hypoxia. The basal levels of corticosterone production in cells were higher in the IH groups than in normoxic group. IH resulted in a time-dependent increase of corticosterone production in response to ACTH, 8-Br-cAMP, progesterone and deoxycorticosterone. The production of pregnenolone in response to 25-OH-C and that of progesterone in response to pregnenolone in ZFR cells were enhanced by 4-day IH. These results suggest that IH in rats increases the secretion of corticosterone via a mechanism at least in part associated with the activation of cAMP pathway and steroidogenic enzymes.
Collapse
Affiliation(s)
- Guey-Shyang Hwang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan.,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, 33378, Taiwan
| | - Chih-Chieh Chen
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Jou-Chun Chou
- Medical Center of Aging Research, China Medical University Hospital, Taichung, 40402, Taiwan.,Department of Life Sciences, National Chung Hsing University, Taichung, 40254, Taiwan
| | - Ling-Ling Chang
- Department of Chemical and Materials Engineering, Chinese Culture University, Taipei, 11114, Taiwan
| | - Shu-Fen Kan
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Wei-Ho Lai
- Department of Rehabilitation, Cheng Hsin General Hospital, Taipei, 11212, Taiwan
| | - Fu-Kong Lieu
- Department of Rehabilitation, Cheng Hsin General Hospital, Taipei, 11212, Taiwan
| | - Sindy Hu
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan.,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, 33378, Taiwan
| | - Paulus S Wang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.,Medical Center of Aging Research, China Medical University Hospital, Taichung, 40402, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan.,Graduate Institute of Basic Medical Science,College of Medicine, China Medical University, Taichung, 40402, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, 41354, Taiwan, Republic of China
| | - Shyi-Wu Wang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan. .,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, 33378, Taiwan.
| |
Collapse
|
39
|
McLeod DS, Bhutto I, Edwards MM, Silver RE, Seddon JM, Lutty GA. Distribution and Quantification of Choroidal Macrophages in Human Eyes With Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2017; 57:5843-5855. [PMID: 27802514 PMCID: PMC5098452 DOI: 10.1167/iovs.16-20049] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Increasing evidence suggests a role for macrophages in the pathogenesis of age-related macular degeneration (AMD). This study examined choroidal macrophages and their activation in postmortem eyes from subjects with and without AMD. Methods Choroids were incubated with anti-ionized calcium-binding adapter molecule 1 (anti-IBA1) to label macrophages, anti-human leukocyte antigen-antigen D-related (anti-HLA-DR) as a macrophage activation marker, and Ulex europaeus agglutinin lectin to label blood vessels. Whole mounts were imaged using confocal microscopy. IBA1- and HLA-DR–positive (activated) cells were counted in submacula, paramacula, and nonmacula, and cell volume and sphericity were determined using computer-assisted image analysis. Results In aged control eyes, the mean number of submacular IBA1+ and HLA-DR+ macrophages was 433/mm2 and 152/mm2, respectively. In early AMD eyes, there was a significant increase in IBA1+ and HLA-DR+ cells in submacula compared to those in controls (P = 0.0015 and P = 0.008, respectively). In eyes with neovascular AMD, there were significantly more HLA-DR+ cells associated with submacular choroidal neovascularization (P = 0.001). Mean cell volume was significantly lower (P ≤ 0.02), and sphericity was significantly higher (P ≤ 0.005) in all AMD groups compared to controls. Conclusions The average number of IBA1+ macrophages in submacular and paramacular choroid was significantly higher in early/intermediate AMD compared to that in aged controls. HLA-DR+ submacular macrophages were significantly increased in all stages of AMD, and they were significantly more round and smaller in size in the submacular AMD choroid, suggesting their activation. These findings support the concept that AMD is an inflammatory disease.
Collapse
Affiliation(s)
- D Scott McLeod
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland
| | - Imran Bhutto
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland
| | - Malia M Edwards
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland
| | - Rachel E Silver
- Ophthalmic Epidemiology and Genetics Service, New England Eye Center, Tufts Medical Center, Boston, Massachusetts
| | - Johanna M Seddon
- Ophthalmic Epidemiology and Genetics Service, New England Eye Center, Tufts Medical Center, Boston, Massachusetts 3Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts
| | - Gerard A Lutty
- Wilmer Ophthalmological Institute, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
40
|
Gerri C, Marín-Juez R, Marass M, Marks A, Maischein HM, Stainier DYR. Hif-1α regulates macrophage-endothelial interactions during blood vessel development in zebrafish. Nat Commun 2017; 8:15492. [PMID: 28524872 PMCID: PMC5493593 DOI: 10.1038/ncomms15492] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 04/01/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages are known to interact with endothelial cells during developmental and pathological angiogenesis but the molecular mechanisms modulating these interactions remain unclear. Here, we show a role for the Hif-1α transcription factor in this cellular communication. We generated hif-1aa;hif-1ab double mutants in zebrafish, hereafter referred to as hif-1α mutants, and find that they exhibit impaired macrophage mobilization from the aorta-gonad-mesonephros (AGM) region as well as angiogenic defects and defective vascular repair. Importantly, macrophage ablation is sufficient to recapitulate the vascular phenotypes observed in hif-1α mutants, revealing for the first time a macrophage-dependent angiogenic process during development. Further substantiating our observations of vascular repair, we find that most macrophages closely associated with ruptured blood vessels are Tnfα-positive, a key feature of classically activated macrophages. Altogether, our data provide genetic evidence that Hif-1α regulates interactions between macrophages and endothelial cells starting with the mobilization of macrophages from the AGM. The molecular mechanism regulating macrophage interaction with endothelial cells during development is unclear. Here, the authors show that in zebrafish mutation of hypoxia-inducible factor-1α impairs macrophage mobilization from the aorta-gonad-mesonephros, causing defects in angiogenesis and vessel repair.
Collapse
Affiliation(s)
- Claudia Gerri
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Michele Marass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Alora Marks
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
41
|
Petrovic-Djergovic D, Goonewardena SN, Pinsky DJ. Inflammatory Disequilibrium in Stroke. Circ Res 2017; 119:142-58. [PMID: 27340273 DOI: 10.1161/circresaha.116.308022] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/25/2016] [Indexed: 01/01/2023]
Abstract
Over the past several decades, there have been substantial advances in our knowledge of the pathophysiology of stroke. Understanding the benefits of timely reperfusion has led to the development of thrombolytic therapy as the cornerstone of current management of ischemic stroke, but there remains much to be learned about mechanisms of neuronal ischemic and reperfusion injury and associated inflammation. For ischemic stroke, novel therapeutic targets have continued to remain elusive. When considering modern molecular biological techniques, advanced translational stroke models, and clinical studies, a consistent pattern emerges, implicating perturbation of the immune equilibrium by stroke in both central nervous system injury and repair responses. Stroke triggers activation of the neuroimmune axis, comprised of multiple cellular constituents of the immune system resident within the parenchyma of the brain, leptomeninges, and vascular beds, as well as through secretion of biological response modifiers and recruitment of immune effector cells. This neuroimmune activation can directly impact the initiation, propagation, and resolution phases of ischemic brain injury. To leverage a potential opportunity to modulate local and systemic immune responses to favorably affect the stroke disease curve, it is necessary to expand our mechanistic understanding of the neuroimmune axis in ischemic stroke. This review explores the frontiers of current knowledge of innate and adaptive immune responses in the brain and how these responses together shape the course of ischemic stroke.
Collapse
Affiliation(s)
- Danica Petrovic-Djergovic
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor
| | - Sascha N Goonewardena
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor
| | - David J Pinsky
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor.
| |
Collapse
|
42
|
Inflammation and Cancer: Extra- and Intracellular Determinants of Tumor-Associated Macrophages as Tumor Promoters. Mediators Inflamm 2017; 2017:9294018. [PMID: 28197019 PMCID: PMC5286482 DOI: 10.1155/2017/9294018] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/26/2016] [Indexed: 02/08/2023] Open
Abstract
One of the hallmarks of cancer-related inflammation is the recruitment of monocyte-macrophage lineage cells to the tumor microenvironment. These tumor infiltrating myeloid cells are educated by the tumor milieu, rich in cancer cells and stroma components, to exert functions such as promotion of tumor growth, immunosuppression, angiogenesis, and cancer cell dissemination. Our review highlights the ontogenetic diversity of tumor-associated macrophages (TAMs) and describes their main phenotypic markers. We cover fundamental molecular players in the tumor microenvironment including extra- (CCL2, CSF-1, CXCL12, IL-4, IL-13, semaphorins, WNT5A, and WNT7B) and intracellular signals. We discuss how these factors converge on intracellular determinants (STAT3, STAT6, STAT1, NF-κB, RORC1, and HIF-1α) of cell functions and drive the recruitment and polarization of TAMs. Since microRNAs (miRNAs) modulate macrophage polarization key miRNAs (miR-146a, miR-155, miR-125a, miR-511, and miR-223) are also discussed in the context of the inflammatory myeloid tumor compartment. Accumulating evidence suggests that high TAM infiltration correlates with disease progression and overall poor survival of cancer patients. Identification of molecular targets to develop new therapeutic interventions targeting these harmful tumor infiltrating myeloid cells is emerging nowadays.
Collapse
|
43
|
Takikawa A, Mahmood A, Nawaz A, Kado T, Okabe K, Yamamoto S, Aminuddin A, Senda S, Tsuneyama K, Ikutani M, Watanabe Y, Igarashi Y, Nagai Y, Takatsu K, Koizumi K, Imura J, Goda N, Sasahara M, Matsumoto M, Saeki K, Nakagawa T, Fujisaka S, Usui I, Tobe K. HIF-1α in Myeloid Cells Promotes Adipose Tissue Remodeling Toward Insulin Resistance. Diabetes 2016; 65:3649-3659. [PMID: 27625023 DOI: 10.2337/db16-0012] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 09/08/2016] [Indexed: 01/05/2023]
Abstract
Adipose tissue hypoxia is an important feature of pathological adipose tissue expansion. Hypoxia-inducible factor-1α (HIF-1α) in adipocytes reportedly induces oxidative stress and fibrosis, rather than neoangiogenesis via vascular endothelial growth factor (VEGF)-A. We previously reported that macrophages in crown-like structures (CLSs) are both hypoxic and inflammatory. In the current study, we examined how macrophage HIF-1α is involved in high-fat diet (HFD)-induced inflammation, neovascularization, hypoxia, and insulin resistance using mice with myeloid cell-specific HIF-1α deletion that were fed an HFD. Myeloid cell-specific HIF-1α gene deletion protected against HFD-induced inflammation, CLS formation, poor vasculature development in the adipose tissue, and systemic insulin resistance. Despite a reduced expression of Vegfa in epididymal white adipose tissue (eWAT), the preadipocytes and endothelial cells of HIF-1α-deficient mice expressed higher levels of angiogenic factors, including Vegfa, Angpt1, Fgf1, and Fgf10 in accordance with preferable eWAT remodeling. Our in vitro study revealed that lipopolysaccharide-treated bone marrow-derived macrophages directly inhibited the expression of angiogenic factors in 3T3-L1 preadipocytes. Thus, macrophage HIF-1α is involved not only in the formation of CLSs, further enhancing the inflammatory responses, but also in the inhibition of neoangiogenesis in preadipocytes. We concluded that these two pathways contribute to the obesity-related physiology of pathological adipose tissue expansion, thus causing systemic insulin resistance.
Collapse
Affiliation(s)
- Akiko Takikawa
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Arshad Mahmood
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Allah Nawaz
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Keisuke Okabe
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, Toyama, Japan
| | | | - Satoko Senda
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Koichi Tsuneyama
- Department of Diagnostic Pathology, University of Toyama, Toyama, Japan
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, Advanced Biomedicine Genome Pharmaceutical Science, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Yasuharu Watanabe
- Department of Immunobiology and Pharmacological Genetics, Advanced Biomedicine Genome Pharmaceutical Science, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Yoshiko Igarashi
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Advanced Biomedicine Genome Pharmaceutical Science, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, Advanced Biomedicine Genome Pharmaceutical Science, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
- Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
| | - Keiichi Koizumi
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, University of Toyama, Toyama, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | | | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kumiko Saeki
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takashi Nakagawa
- Frontier Research Core for Life Science, University of Toyama, Toyama, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
44
|
Szebeni GJ, Vizler C, Nagy LI, Kitajka K, Puskas LG. Pro-Tumoral Inflammatory Myeloid Cells as Emerging Therapeutic Targets. Int J Mol Sci 2016; 17:ijms17111958. [PMID: 27886105 PMCID: PMC5133952 DOI: 10.3390/ijms17111958] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/01/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
Since the observation of Virchow, it has long been known that the tumor microenvironment constitutes the soil for the infiltration of inflammatory cells and for the release of inflammatory mediators. Under certain circumstances, inflammation remains unresolved and promotes cancer development. Here, we review some of these indisputable experimental and clinical evidences of cancer related smouldering inflammation. The most common myeloid infiltrate in solid tumors is composed of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). These cells promote tumor growth by several mechanisms, including their inherent immunosuppressive activity, promotion of neoangiogenesis, mediation of epithelial-mesenchymal transition and alteration of cellular metabolism. The pro-tumoral functions of TAMs and MDSCs are further enhanced by their cross-talk offering a myriad of potential anti-cancer therapeutic targets. We highlight these main pro-tumoral mechanisms of myeloid cells and give a general overview of their phenotypical and functional diversity, offering examples of possible therapeutic targets. Pharmacological targeting of inflammatory cells and molecular mediators may result in therapies improving patient condition and prognosis. Here, we review experimental and clinical findings on cancer-related inflammation with a major focus on creating an inventory of current small molecule-based therapeutic interventions targeting cancer-related inflammatory cells: TAMs and MDSCs.
Collapse
Affiliation(s)
- Gabor J Szebeni
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
- Synaptogenex Ltd., Őzsuta utca 20995/1, H-1037 Budapest, Hungary.
| | - Csaba Vizler
- Department of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| | - Lajos I Nagy
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
| | - Klara Kitajka
- Department of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| | - Laszlo G Puskas
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
- Department of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| |
Collapse
|
45
|
Movahedi K, Van Ginderachter JA. The Ontogeny and Microenvironmental Regulation of Tumor-Associated Macrophages. Antioxid Redox Signal 2016; 25:775-791. [PMID: 27020982 DOI: 10.1089/ars.2016.6704] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
SIGNIFICANCE Tumor progression is supported by non-cancerous stromal cells, of which tumor-associated macrophages (TAMs) are prominent constituents. These cells could be considered promising therapeutic targets, but this requires a better understanding of their heterogeneity under the influence of tumor microenvironmental cues and/or ontogenic differences. Recent Advances: The availability of oxygen is an important regulator of the TAM phenotype, as well as of its access to myelopoietic growth factors. Very recent evidence also demonstrated that macrophages can be derived from embryonal precursors or from monocytes post-birth, introducing yet another level of heterogeneity among macrophages. CRITICAL ISSUES The relative contribution of ontogenically distinct macrophages to tumor characteristics is, to a large extent, still an open question. In addition, further knowledge on the role of tumor microenvirontal cues that shape TAMs is warranted. FUTURE DIRECTIONS More detailed insights into the TAM-regulating factors will provide new opportunities for therapeutic intervention. Interference with the phenotypes of TAM, which are known to be immunosuppressive and to contribute to dysfunctional tumor blood vessels, is anticipated to be beneficial in combination with chemotherapy and/or immunotherapy. Antioxid. Redox Signal. 25, 775-791.
Collapse
Affiliation(s)
- Kiavash Movahedi
- 1 Myeloid Cell Immunology Lab, VIB Inflammation Research Center , Ghent, Belgium .,2 Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Jo A Van Ginderachter
- 1 Myeloid Cell Immunology Lab, VIB Inflammation Research Center , Ghent, Belgium .,2 Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels, Belgium
| |
Collapse
|
46
|
Whiteford JR, De Rossi G, Woodfin A. Mutually Supportive Mechanisms of Inflammation and Vascular Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:201-78. [PMID: 27572130 DOI: 10.1016/bs.ircmb.2016.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is often accompanied by angiogenesis, the development of new blood vessels from existing ones. This vascular response is a response to chronic hypoxia and/or ischemia, but is also contributory to the progression of disorders including atherosclerosis, arthritis, and tumor growth. Proinflammatory and proangiogenic mediators and signaling pathways form a complex and interrelated network in these conditions, and many factors exert multiple effects. Inflammation drives angiogenesis by direct and indirect mechanisms, promoting endothelial proliferation, migration, and vessel sprouting, but also by mediating extracellular matrix remodeling and release of sequestered growth factors, and recruitment of proangiogenic leukocyte subsets. The role of inflammation in promoting angiogenesis is well documented, but by facilitating greater infiltration of leukocytes and plasma proteins into inflamed tissues, angiogenesis can also propagate chronic inflammation. This review examines the mutually supportive relationship between angiogenesis and inflammation, and considers how these interactions might be exploited to promote resolution of chronic inflammatory or angiogenic disorders.
Collapse
Affiliation(s)
- J R Whiteford
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - G De Rossi
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - A Woodfin
- Cardiovascular Division, King's College, University of London, London, United Kingdom.
| |
Collapse
|
47
|
Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol 2016; 142:23-44. [PMID: 27166859 DOI: 10.1016/j.pneurobio.2016.05.001] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/20/2016] [Accepted: 05/01/2016] [Indexed: 02/08/2023]
Abstract
Microglia/macrophages are the major immune cells involved in the defence against brain damage. Their morphology and functional changes are correlated with the release of danger signals induced by stroke. These cells are normally responsible for clearing away dead neural cells and restoring neuronal functions. However, when excessively activated by the damage-associated molecular patterns following stroke, they can produce a large number of proinflammatory cytokines that can disrupt neural cells and the blood-brain barrier and influence neurogenesis. These effects indicate the important roles of microglia/macrophages in the pathophysiological processes of stroke. However, the modifiable and adaptable nature of microglia/macrophages may also be beneficial for brain repair and not just result in damage. These distinct roles may be attributed to the different microglia/macrophage phenotypes because the M1 population is mainly destructive, while the M2 population is neuroprotective. Additionally, different gene expression signature changes in microglia/macrophages have been found in diverse inflammatory milieus. These biofunctional features enable dual roles for microglia/macrophages in brain damage and repair. Currently, it is thought that the proper inflammatory milieu may provide a suitable microenvironment for neurogenesis; however, detailed mechanisms underlying the inflammatory responses that initiate or inhibit neurogenesis remain unknown. This review summarizes recent progress concerning the mechanisms involved in brain damage, repair and regeneration related to microglia/macrophage activation and phenotype transition after stroke. We also argue that future translational studies should be targeting multiple key regulating molecules to improve brain repair, which should be accompanied by the concept of a "therapeutic time window" for sequential therapies.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China.
| |
Collapse
|
48
|
Porta C, Riboldi E, Ippolito A, Sica A. Molecular and epigenetic basis of macrophage polarized activation. Semin Immunol 2016; 27:237-48. [PMID: 26561250 DOI: 10.1016/j.smim.2015.10.003] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022]
Abstract
Macrophages are unique cells for origin, heterogeneity and plasticity. At steady state most of macrophages are derived from fetal sources and maintained in adulthood through self-renewing. Despite sharing common progenitors, a remarkable heterogeneity characterized tissue-resident macrophages indicating that local signals educate them to express organ-specific functions. Macrophages are extremely plastic: chromatin landscape and transcriptional programs can be dynamically re-shaped in response to microenvironmental changes. Owing to their ductility, macrophages are crucial orchestrators of both initiation and resolution of immune responses and key supporters of tissue development and functions in homeostatic and pathological conditions. Herein, we describe current understanding of heterogeneity and plasticity of macrophages using the M1-M2 dichotomy as operationally useful simplification of polarized activation. We focused on the complex network of signaling cascades, metabolic pathways, transcription factors, and epigenetic changes that control macrophage activation. In particular, this network was addressed in sepsis, as a paradigm of a pathological condition determining dynamic macrophage reprogramming.
Collapse
Affiliation(s)
- Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Elena Riboldi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Alessandro Ippolito
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy.
| |
Collapse
|
49
|
Gregory CD, Ford CA, Voss JJLP. Microenvironmental Effects of Cell Death in Malignant Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 930:51-88. [PMID: 27558817 DOI: 10.1007/978-3-319-39406-0_3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although apoptosis is well recognized as a cell death program with clear anticancer roles, accumulating evidence linking apoptosis with tissue repair and regeneration indicates that its relationship with malignant disease is more complex than previously thought. Here we review how the responses of neighboring cells in the microenvironment of apoptotic tumor cells may contribute to the cell birth/cell death disequilibrium that provides the basis for cancerous tissue emergence and growth. We describe the bioactive properties of apoptotic cells and consider, in particular, how apoptosis of tumor cells can engender a range of responses including pro-oncogenic signals having proliferative, angiogenic, reparatory, and immunosuppressive features. Drawing on the parallels between wound healing, tissue regeneration and cancer, we propose the concept of the "onco-regenerative niche," a cell death-driven generic network of tissue repair and regenerative mechanisms that are hijacked in cancer. Finally, we consider how the responses to cell death in tumors can be targeted to provide more effective and long-lasting therapies.
Collapse
Affiliation(s)
- Christopher D Gregory
- MRC Centre for Inflammation Research, University of Edinburgh Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK.
| | - Catriona A Ford
- MRC Centre for Inflammation Research, University of Edinburgh Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Jorine J L P Voss
- MRC Centre for Inflammation Research, University of Edinburgh Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
50
|
Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H, Hoehn M, Fink GR, Schroeter M, Graf R. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. Neuroimage 2015; 128:54-62. [PMID: 26747749 PMCID: PMC4767221 DOI: 10.1016/j.neuroimage.2015.12.044] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/16/2015] [Accepted: 12/23/2015] [Indexed: 01/06/2023] Open
Abstract
Inflammatory cells such as microglia need energy to exert their functions and to maintain their cellular integrity and membrane potential. Subsequent to cerebral ischemia, inflammatory cells infiltrate tissue with limited blood flow where neurons and astrocytes died due to insufficient supply with oxygen and glucose. Using dual tracer positron emission tomography (PET), we found that concomitant with the presence of inflammatory cells, transport and consumption of glucose increased up to normal levels but returned to pathological levels as soon as inflammatory cells disappeared. Thus, inflammatory cells established sufficient glucose supply to satisfy their energy demands even in regions with insufficient supply for neurons and astrocytes to survive. Our data suggest that neurons and astrocytes died from oxygen deficiency and inflammatory cells metabolized glucose non-oxidatively in regions with residual availability. As a consequence, glucose metabolism of inflammatory cells can mask metabolic deficits in neurodegenerative diseases. We further found that the PET tracer did not bind to inflammatory cells in severely hypoperfused regions and thus only a part of the inflammation was detected. We conclude that glucose consumption of inflammatory cells should be taken into account when analyzing disease-related alterations of local cerebral metabolism.
Inflammatory cells consume high amounts of glucose in supply-limited brain regions. Glucose metabolism of inflammatory cells masks metabolic deficits in the brain. In vivo markers only reach inflammatory cells in regions with residual blood supply. Measuring inflammation and metabolism provide complementary information.
Collapse
Affiliation(s)
- Heiko Backes
- Max Planck Institute for Metabolism Research, Cologne, Germany.
| | - Maureen Walberer
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Neurology, University Hospital, Cologne, Germany
| | - Anne Ladwig
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Neurology, University Hospital, Cologne, Germany
| | - Maria A Rueger
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Neurology, University Hospital, Cologne, Germany
| | - Bernd Neumaier
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Germany
| | - Heike Endepols
- Department of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Germany
| | - Mathias Hoehn
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital, Cologne, Germany; Institute of Neuroscience and Medicine (INM-3), Cognitive Neurology Section, Research Centre Juelich, Germany
| | - Michael Schroeter
- Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Neurology, University Hospital, Cologne, Germany
| | - Rudolf Graf
- Max Planck Institute for Metabolism Research, Cologne, Germany
| |
Collapse
|