1
|
Luo J, Zhou Y, Wang M, Zhang J, Jiang E. Inflammasomes: potential therapeutic targets in hematopoietic stem cell transplantation. Cell Commun Signal 2024; 22:596. [PMID: 39695742 DOI: 10.1186/s12964-024-01974-3] [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: 10/05/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024] Open
Abstract
The realm of hematopoietic stem cell transplantation (HSCT) has witnessed remarkable advancements in elevating the cure and survival rates for patients with both malignant and non-malignant hematologic diseases. Nevertheless, a considerable number of patients continue to face challenges, including transplant-related complications, infection, graft failure, and mortality. Inflammasomes, the multi-protein complexes of the innate immune system, respond to various danger signals by releasing inflammatory cytokines and even mediating cell death. While moderate activation of inflammasomes is essential for immune defense and homeostasis maintenance, excessive activation precipitates inflammatory damage. The intricate interplay between HSCT and inflammasomes arises from their pivotal roles in immune responses and inflammation. This review examines the molecular architecture and composition of various types of inflammasomes, highlighting their activation and effector mechanisms within the context of the HSCT process and its associated complications. Additionally, we summarize the therapeutic implications of targeting inflammasomes and related factors in HSCT.
Collapse
Affiliation(s)
- Jieya Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yunxia Zhou
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Haihe Laboratory of Cell Ecosystem, Tianjin Medical University, Tianjin, 300051, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Junan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
2
|
Zhang R, Hu M, Liu Y, Li W, Xu Z, He S, Lu Y, Gong Y, Wang X, Hai S, Li S, Qi S, Li Y, Shu Y, Du D, Zhang H, Xu H, Zhou Z, Lei P, Chen HN, Dai L. Integrative Omics Uncovers Low Tumorous Magnesium Content as A Driver Factor of Colorectal Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae053. [PMID: 39052867 PMCID: PMC11514849 DOI: 10.1093/gpbjnl/qzae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 03/04/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Magnesium (Mg) deficiency is associated with increased risk and malignancy in colorectal cancer (CRC), yet the underlying mechanisms remain elusive. Here, we used genomic, proteomic, and phosphoproteomic data to elucidate the impact of Mg deficiency on CRC. Genomic analysis identified 160 genes with higher mutation frequencies in Low-Mg tumors, including key driver genes such as KMT2C and ERBB3. Unexpectedly, initiation driver genes of CRC, such as TP53 and APC, displayed higher mutation frequencies in High-Mg tumors. Additionally, proteomic and phosphoproteomic data indicated that low Mg content in tumors may activate epithelial-mesenchymal transition (EMT) by modulating inflammation or remodeling the phosphoproteome of cancer cells. Notably, we observed a negative correlation between the phosphorylation of DBN1 at S142 (DBN1S142p) and Mg content. A mutation in S142 to D (DBN1S142D) mimicking DBN1S142p up-regulated MMP2 and enhanced cell migration, while treatment with MgCl2 reduced DBN1S142p, thereby reversing this phenotype. Mechanistically, Mg2+ attenuated the DBN1-ACTN4 interaction by decreasing DBN1S142p, which in turn enhanced the binding of ACTN4 to F-actin and promoted F-actin polymerization, ultimately reducing MMP2 expression. These findings shed new light on the crucial role of Mg deficiency in CRC progression and suggest that Mg supplementation may be a promising preventive and therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Rou Zhang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Hu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Liu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanmeng Li
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Xu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Siyu He
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Lu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiuxuan Wang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Hai
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuangqing Li
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiqian Qi
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Li
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Shu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Du
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huiyuan Zhang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Heng Xu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zongguang Zhou
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Lei
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Zhou Y, Yang M, Yan X, Zhang L, Lu N, Ma Y, Zhang Y, Cui M, Zhang M, Zhang M. Oral Nanotherapeutics of Andrographolide/Carbon Monoxide Donor for Synergistically Anti-inflammatory and Pro-resolving Treatment of Ulcerative Colitis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:36061-36075. [PMID: 37463480 DOI: 10.1021/acsami.3c09342] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease of unknown etiology affecting the colon and rectum. Current therapeutics are focused on suppressing inflammation but are ineffective. Combining anti-inflammatory therapeutic approaches with pro-resolution might be a superior strategy for UC treatment. Andrographolide (AG), an active compound from the plant Andrographis paniculata, presented anti-inflammatory effects in various inflammatory diseases. Gaseous mediators, such as carbon monoxide (CO), have a role in inflammatory resolution. Herein, we developed a dextran-functionalized PLGA nanocarrier for efficient delivery of AG and a carbon monoxide donor (CORM-2) for synergistically anti-inflammatory/pro-resolving treatment of UC (AG/CORM-2@NP-Dex) based on PLGA with good biocompatibility, slow drug release, efficient targeting, and biodegradability. The resulting nanocarrier had a nano-scaled diameter of ∼200 nm and a spherical shape. After being coated with dextran (Dex), the resulting AG/CORM-2@NP-Dex could be efficiently internalized by Colon-26 and Raw 264.7 cells in vitro and preferentially localized to the inflamed colon with chitosan/alginate hydrogel protection by gavage. AG/CORM-2@NP-Dex performed anti-inflammatory effects by eliminating the over-production of pro-inflammatory mediator, nitric oxide (NO), and down-regulating the expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), while it showed pro-resolving function by accelerating M1 to M2 macrophage conversion and up-regulating resolution-related genes (IL-10, TGF-β, and HO-1). In the colitis model, oral administration of AG/CORM-2@NP-Dex in a chitosan/alginate hydrogel also showed synergistically anti-inflammatory/pro-resolving effects, therefore relieving UC effectively. Without appreciable systemic toxicity, this bifunctional nanocarrier represents a novel therapeutic approach for UC and is expected to achieve long-term inflammatory remission.
Collapse
Affiliation(s)
- Ying Zhou
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an 710032, China
| | - Mei Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xian Jiaotong University, Xi'an 710061, China
| | - Xiangji Yan
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lingmin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ning Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Yana Ma
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Manli Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| |
Collapse
|
4
|
Diemer JL, Yu K, Kelly M, Zhen T, Anderson S, Lopez G, Liu P. Characterization of leukemia progression in the Cbfb-MYH11 knockin mice by single cell RNA sequencing. Leukemia 2023; 37:1549-1553. [PMID: 37225965 PMCID: PMC10437305 DOI: 10.1038/s41375-023-01926-9] [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: 12/19/2022] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/26/2023]
Abstract
A recurrent inversion of chromosome 16 fuses the genes CBFB and MYH11 and the resultant CBFβ-SMMHC fusion protein acts as a driver of the M4Eo subtype of acute myeloid leukemia (AML). This study utilized single cell RNA-sequencing (scRNA-seq) to assess gene expression changes during the disease progression from normal to pre-leukemic to overt leukemia in the conditional Cbfb-MYH11 knock-in mouse model. The study investigated dynamic shifts in cell types as disease progresses, and gene expression differences between normal and diseased cells. The study identified a novel cell population in the pre-leukemic state with expression of genes involved in immune activation. Overall, this study discovered hematopoietic cells first affected by the expression of CBFβ-SMMHC and identified unique signature genes for the pre-leukemic cells that separate them from the normal hematopoietic cellular milieu.
Collapse
Affiliation(s)
- Jamie L Diemer
- Oncogenesis and Development Section, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Kai Yu
- Oncogenesis and Development Section, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Michael Kelly
- Laboratory of Cochlear Development, Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
- Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Center for Cancer Research, Natioal Cancer Institute, NIH, Bethesda, MD, USA
| | - Tao Zhen
- Oncogenesis and Development Section, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Stacie Anderson
- Flow Cytometry Core, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Guadalupe Lopez
- Oncogenesis and Development Section, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Paul Liu
- Oncogenesis and Development Section, Division of Intramural Research, National Human Genome Research Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
5
|
Intratumoral microbiota: roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther 2023; 8:35. [PMID: 36646684 PMCID: PMC9842669 DOI: 10.1038/s41392-022-01304-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/31/2022] [Accepted: 12/26/2022] [Indexed: 01/18/2023] Open
Abstract
Microorganisms, including bacteria, viruses, fungi, and other eukaryotes, play critical roles in human health. An altered microbiome can be associated with complex diseases. Intratumoral microbial components are found in multiple tumor tissues and are closely correlated with cancer initiation and development and therapy efficacy. The intratumoral microbiota may contribute to promotion of the initiation and progression of cancers by DNA mutations, activating carcinogenic pathways, promoting chronic inflammation, complement system, and initiating metastasis. Moreover, the intratumoral microbiota may not only enhance antitumor immunity via mechanisms including STING signaling activation, T and NK cell activation, TLS production, and intratumoral microbiota-derived antigen presenting, but also decrease antitumor immune responses and promote cancer progression through pathways including upregulation of ROS, promoting an anti-inflammatory environment, T cell inactivation, and immunosuppression. The effect of intratumoral microbiota on antitumor immunity is dependent on microbiota composition, crosstalk between microbiota and the cancer, and status of cancers. The intratumoral microbiota may regulate cancer cell physiology and the immune response by different signaling pathways, including ROS, β-catenin, TLR, ERK, NF-κB, and STING, among others. These viewpoints may help identify the microbiota as diagnosis or prognosis evaluation of cancers, and as new therapeutic strategy and potential therapeutic targets for cancer therapy.
Collapse
|
6
|
A Bioinformatics View on Acute Myeloid Leukemia Surface Molecules by Combined Bayesian and ABC Analysis. Bioengineering (Basel) 2022; 9:bioengineering9110642. [DOI: 10.3390/bioengineering9110642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
“Big omics data” provoke the challenge of extracting meaningful information with clinical benefit. Here, we propose a two-step approach, an initial unsupervised inspection of the structure of the high dimensional data followed by supervised analysis of gene expression levels, to reconstruct the surface patterns on different subtypes of acute myeloid leukemia (AML). First, Bayesian methodology was used, focusing on surface molecules encoded by cluster of differentiation (CD) genes to assess whether AML is a homogeneous group or segregates into clusters. Gene expressions of 390 patient samples measured using microarray technology and 150 samples measured via RNA-Seq were compared. Beyond acute promyelocytic leukemia (APL), a well-known AML subentity, the remaining AML samples were separated into two distinct subgroups. Next, we investigated which CD molecules would best distinguish each AML subgroup against APL, and validated discriminative molecules of both datasets by searching the scientific literature. Surprisingly, a comparison of both omics analyses revealed that CD339 was the only overlapping gene differentially regulated in APL and other AML subtypes. In summary, our two-step approach for gene expression analysis revealed two previously unknown subgroup distinctions in AML based on surface molecule expression, which may guide the differentiation of subentities in a given clinical–diagnostic context.
Collapse
|
7
|
Xu Y, Sun Y, Yin R, Dong T, Song K, Fang Y, Liu G, Shen B, Li H. Differential expression of plasma exosomal microRNA in severe acute pancreatitis. Front Pharmacol 2022; 13:980930. [PMID: 36249739 PMCID: PMC9554001 DOI: 10.3389/fphar.2022.980930] [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: 06/29/2022] [Accepted: 09/06/2022] [Indexed: 11/15/2022] Open
Abstract
The incidence rate of acute pancreatitis is increasing, and severe acute pancreatitis (SAP) is associated with a high mortality rate, which may be reduced by a deeper understanding of its pathogenesis. In addition, an early determination of the severity of acute pancreatitis remains challenging. The aim of this study was to match potential biomarkers for early identification and monitoring of acute pancreatitis and to shed light on the underlying pathogenic mechanisms of SAP. The expression levels of plasma exosomal microRNA (miRNA) in patients with pancreatitis have been associated with the disease. Thus, this study compared the expression levels of exosomal miRNA in plasma collected from four patients with SAP and from four healthy participants. Analyses of the miRNA expression profiles indicated that three previously unreported miRNAs were differentially expressed in the patient group: Novel1, which was downregulated, and Novel2 and Novel3, which were upregulated. The miRNA target genes for those novel miRNAs were predicted using Metascape. Of these miRNA target genes, those that were also differentially expressed at different time points after disease induction in a mouse model of acute pancreatitis were determined. The gene for complement component 3 (C3), a target gene of Novel3, was the only gene matched in both the patient group and the mouse model. C3 appeared at most of the time points assessed after induction of acute pancreatitis in mice. These findings are foundational evidence that C3 warrants further study as an early biomarker of SAP, for investigating underlying pathogenic mechanisms of SAP, and as a therapeutic target for ameliorating the occurrence or development of SAP.
Collapse
Affiliation(s)
- Yansong Xu
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuansong Sun
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ran Yin
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tao Dong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Kai Song
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Guodong Liu
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Chuzhou, Anhui, China
- *Correspondence: Guodong Liu, ; Bing Shen, ; He Li,
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Chuzhou, Anhui, China
- *Correspondence: Guodong Liu, ; Bing Shen, ; He Li,
| | - He Li
- Department of Emergency, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- *Correspondence: Guodong Liu, ; Bing Shen, ; He Li,
| |
Collapse
|
8
|
Fang PH, Lai YY, Chen CL, Wang HY, Chang YN, Lin YC, Yan YT, Lai CH, Cheng B. Cobalt protoporphyrin promotes human keratinocyte migration under hyperglycemic conditions. Mol Med 2022; 28:71. [PMID: 35739477 PMCID: PMC9219158 DOI: 10.1186/s10020-022-00499-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/14/2022] [Indexed: 11/24/2022] Open
Abstract
Background Complete healing of diabetic wounds continues to be a clinically unmet need. Although robust therapies such as stem cell therapy and growth factor treatment are clinically applied, these treatments are costly for most diabetic wound patients. Therefore, a cheaper alternative is needed. Cobalt protoporphyrin (CoPP) has recently been demonstrated to promote tissue regeneration. In this study, the therapeutic benefits of CoPP in diabetic wound healing were examined. Methods An in vitro wound healing model that mimics re-epithelialization was established to examine the effect of CoPP on the migratory capability of human keratinocytes (HaCaT) in either normal glucose (NG) or high glucose (HG) media, as well as in the presence of either H2O2 or lipopolysaccharide (LPS). At the end of the migration assays, cells were collected and subjected to Western blotting analysis and immunostaining. Results HaCaT were found to migrate significantly more slowly in the HG media compared to the NG media. CoPP treatment was found to enhance cell migration in HG media, but was found to decrease cell migration and proliferation when HaCaT were cultured in NG media. CoPP treatment induced high levels of expression of Nrf-2/HO-1 and FoxO1 in HaCaT cultured in either glucose concentration, although the FoxO1 expression was found to be significantly higher in HaCaT that underwent the migration assay in NG media compared to those in HG media. The higher level of FoxO1 expression seen in CoPP-treated HaCaT cultured in NG media resulted in upregulation of CCL20 and downregulation of TGFβ1. In contrast, HaCaT migrated in HG media were found to have high levels of expression of TGFβ1, and low levels of expression of CCL20. Interestingly, in the presence of H2O2, CoPP-pretreated HaCaT cultured in either NG or HG media had similar expression level of Nrf-2/HO-1 and FoxO1 to each other. Moreover, the anti-apoptotic effect of CoPP pretreatment was noticed in HaCaT cultured in either glucose concentration. Additionally, CoPP pretreatment was shown to promote tight junction formation in HaCaT suffering from LPS-induced damage. Conclusions CoPP enhances cell migratory capacity under hyperglycemic conditions, and protects cells from oxidative and LPS-induced cellular damage in HG media containing either H2O2 or LPS. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00499-0.
Collapse
Affiliation(s)
- Peng-Hsiang Fang
- Department of Veterinary Medicine, National Chung-Hsing University, No.145, Xing Da Road, 402, Taichung, Taiwan
| | - Ying-Ying Lai
- Bachelor Program of Biotechnology, National Chung-Hsing University, Taichung, Taiwan
| | - Chih-Ling Chen
- Bachelor Program of Biotechnology, National Chung-Hsing University, Taichung, Taiwan
| | - Hsin-Yu Wang
- Bachelor Program of Biotechnology, National Chung-Hsing University, Taichung, Taiwan
| | - Ya-Ning Chang
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, No.145, Xing Da Road, 402, Taichung, Taiwan
| | - Yung-Chang Lin
- Department of Veterinary Medicine, National Chung-Hsing University, No.145, Xing Da Road, 402, Taichung, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Cheng-Hung Lai
- Department of Veterinary Medicine, National Chung-Hsing University, No.145, Xing Da Road, 402, Taichung, Taiwan.
| | - Bill Cheng
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, No.145, Xing Da Road, 402, Taichung, Taiwan.
| |
Collapse
|
9
|
Dutta K, Friscic J, Hoffmann MH. Targeting the tissue-complosome for curbing inflammatory disease. Semin Immunol 2022; 60:101644. [PMID: 35902311 DOI: 10.1016/j.smim.2022.101644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 01/15/2023]
Abstract
Hyperactivated local tissue is a cardinal feature of immune-mediated inflammatory diseases of various organs such as the joints, the gut, the skin, or the lungs. Tissue-resident structural and stromal cells, which get primed during repeated or long-lasting bouts of inflammation form the basis of this sensitization of the tissue. During priming, cells change their metabolism to make them fit for the heightened energy demands that occur during persistent inflammation. Epigenetic changes and, curiously, an activation of intracellularly expressed parts of the complement system drive this metabolic invigoration and enable tissue-resident cells and infiltrating immune cells to employ an arsenal of inflammatory functions, including activation of inflammasomes. Here we provide a current overview on complement activation and inflammatory transformation in tissue-occupying cells, focusing on fibroblasts during arthritis, and illustrate ways how therapeutics directed at complement C3 could potentially target the complosome to unprime cells in the tissue and induce long-lasting abatement of inflammation.
Collapse
Affiliation(s)
- Kuheli Dutta
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany
| | - Jasna Friscic
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany
| | - Markus H Hoffmann
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
10
|
Karantanos T, Teodorescu P, Perkins B, Christodoulou I, Esteb C, Varadhan R, Helmenstine E, Rajkhowa T, Paun BC, Bonifant C, Dalton WB, Gondek LP, Moliterno AR, Levis MJ, Ghiaur G, Jones RJ. The role of the atypical chemokine receptor CCRL2 in myelodysplastic syndrome and secondary acute myeloid leukemia. SCIENCE ADVANCES 2022; 8:eabl8952. [PMID: 35179961 PMCID: PMC8856621 DOI: 10.1126/sciadv.abl8952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/23/2021] [Indexed: 06/06/2023]
Abstract
The identification of new pathways supporting the myelodysplastic syndrome (MDS) primitive cells growth is required to develop targeted therapies. Within myeloid malignancies, men have worse outcomes than women, suggesting male sex hormone-driven effects in malignant hematopoiesis. Androgen receptor promotes the expression of five granulocyte colony-stimulating factor receptor-regulated genes. Among them, CCRL2 encodes an atypical chemokine receptor regulating cytokine signaling in granulocytes, but its role in myeloid malignancies is unknown. Our study revealed that CCRL2 is up-regulated in primitive cells from patients with MDS and secondary acute myeloid leukemia (sAML). CCRL2 knockdown suppressed MDS92 and MDS-L cell growth and clonogenicity in vitro and in vivo and decreased JAK2/STAT3/STAT5 phosphorylation. CCRL2 coprecipitated with JAK2 and potentiated JAK2-STAT interaction. Erythroleukemia cells expressing JAK2V617F showed less effect of CCRL2 knockdown, whereas fedratinib potentiated the CCRL2 knockdown effect. Conclusively, our results implicate CCRL2 as an MDS/sAML cell growth mediator, partially through JAK2/STAT signaling.
Collapse
Affiliation(s)
- Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Patric Teodorescu
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Brandy Perkins
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Ilias Christodoulou
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Christopher Esteb
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Eric Helmenstine
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Trivikram Rajkhowa
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Bogdan C. Paun
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Challice Bonifant
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - W. Brian Dalton
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Lukasz P. Gondek
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Alison R. Moliterno
- Division of Adult Hematology, Department of Medicine, Johns Hopkins University, Baltimore MD, USA
| | - Mark J. Levis
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Gabriel Ghiaur
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Richard J. Jones
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| |
Collapse
|
11
|
Ou B, Liu Y, Yang X, Xu X, Yan Y, Zhang J. C5aR1-positive neutrophils promote breast cancer glycolysis through WTAP-dependent m6A methylation of ENO1. Cell Death Dis 2021; 12:737. [PMID: 34312368 PMCID: PMC8313695 DOI: 10.1038/s41419-021-04028-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/20/2022]
Abstract
Neutrophils are significant compositions of solid tumors and exert distinct functions in different types of tumors. However, the precise role of neutrophils in the progression of breast cancer (BC) is presently unclear. In this study, by investigating the single-cell RNA sequencing data, we identify a new neutrophil subset, C5aR1-positive neutrophils, that correlates with tumor progression and poor survival for BC patients. Furthermore, it is discovered that C5aR1-positive neutrophils enhance BC cell glycolysis via upregulating ENO1 expression. Mechanically, C5aR1-positive neutrophil-secreted IL1β and TNFα cooperatively activate ERK1/2 signaling, which phosphorylates WTAP at serine341 and thereby stabilizes WTAP protein. The stabilization of WTAP further promotes RNA m6A methylation of ENO1, impacting the glycolytic activity of BC cells. Importantly, C5aR1-positive neutrophils also promote breast cancer growth in vivo, and this effect is abolished by WTAP silencing. In clinical BC samples, increased C5aR1-positive neutrophils correlate with elevated IL1β, TNFα, and ENO1 expression. A high co-expression of C5aR1-positive neutrophil gene signature and ENO1 predicts worse prognosis of BC patients compared with a low co-expression. Collectively, our study reveals a novel subset of C5aR1-positive neutrophils that induces breast cancer glycolysis via increasing ERK1/2-WTAP-dependent m6A methylation of ENO1. These findings support the potential for exploration of C5aR1-positive neutrophils as a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Baochi Ou
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, 230022, Hefei, Anhui, China.
| | - Yuan Liu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, No. 85, Wujin Road, 200080, Shanghai, China
| | - Xiaowei Yang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, 230022, Hefei, Anhui, China
| | - Xiaojun Xu
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, 230022, Hefei, Anhui, China
| | - Yunwen Yan
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, 230022, Hefei, Anhui, China
| | - Jingjie Zhang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, 230022, Hefei, Anhui, China
| |
Collapse
|
12
|
Ratajczak MZ, Adamiak M, Ratajczak J, Kucia M. Heme Oxygenase 1 (HO-1) as an Inhibitor of Trafficking of Normal and Malignant Hematopoietic Stem Cells - Clinical and Translational Implications. Stem Cell Rev Rep 2021; 17:821-828. [PMID: 33196976 PMCID: PMC8166705 DOI: 10.1007/s12015-020-10083-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/12/2022]
Abstract
Evidence indicates that bone marrow (BM)-residing hematopoietic stem/progenitor cells (HSPCs) are released into peripheral blood (PB) after administration of pro-mobilizing drugs, which induce a state of sterile inflammation in the BM microenvironment. In the reverse process, as seen after hematopoietic transplantation, intravenously injected HSPCs home and engraft into BM niches. Here again, conditioning for transplantation by myeloablative chemo- or radiotherapy induces a state of sterile inflammation that promotes HSPC seeding to BM stem cell niches. Therefore, the trafficking of HSPCs and their progeny, including granulocytes and monocytes/macrophages, is regulated by a response to pro-inflammatory stimuli. This responsiveness to inflammatory cues is also preserved after malignant transformation of hematopoietic cells. Results from our laboratory indicate that the responsiveness of hematopoietic cells to pro-inflammatory stimuli is orchestrated by Nlrp3 inflammasome. As reported, HO-1 effectively attenuates intracellular activation of Nlrp3 inflammasome as well as the pro-inflammatory effects of several humoral mediators, including complement cascade (ComC) cleavage fragments that promote migration of hematopoietic cells. Based on this finding, inhibition of HO-1 activity may become a practical strategy to enhance the mobilization and homing of normal HSPCs, and, alternatively, its activation may prevent unwanted spread and in vivo expansion of leukemic cells. Graphical Abstract.
Collapse
Affiliation(s)
- Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
13
|
Dander E, Palmi C, D’Amico G, Cazzaniga G. The Bone Marrow Niche in B-Cell Acute Lymphoblastic Leukemia: The Role of Microenvironment from Pre-Leukemia to Overt Leukemia. Int J Mol Sci 2021; 22:ijms22094426. [PMID: 33922612 PMCID: PMC8122951 DOI: 10.3390/ijms22094426] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic lesions predisposing to pediatric B-cell acute lymphoblastic leukemia (B-ALL) arise in utero, generating a clinically silent pre-leukemic phase. We here reviewed the role of the surrounding bone marrow (BM) microenvironment in the persistence and transformation of pre-leukemic clones into fully leukemic cells. In this context, inflammation has been highlighted as a crucial microenvironmental stimulus able to promote genetic instability, leading to the disease manifestation. Moreover, we focused on the cross-talk between the bulk of leukemic cells with the surrounding microenvironment, which creates a “corrupted” BM malignant niche, unfavorable for healthy hematopoietic precursors. In detail, several cell subsets, including stromal, endothelial cells, osteoblasts and immune cells, composing the peculiar leukemic niche, can actively interact with B-ALL blasts. Through deregulated molecular pathways they are able to influence leukemia development, survival, chemoresistance, migratory and invasive properties. The concept that the pre-leukemic and leukemic cell survival and evolution are strictly dependent both on genetic lesions and on the external signals coming from the microenvironment paves the way to a new idea of dual targeting therapeutic strategy.
Collapse
Affiliation(s)
- Erica Dander
- Correspondence: (E.D.); (C.P.); Tel.: +39-(0)-39-2332229 (E.D. & C.P.); Fax: +39-(0)39-2332167 (E.D. & C.P.)
| | - Chiara Palmi
- Correspondence: (E.D.); (C.P.); Tel.: +39-(0)-39-2332229 (E.D. & C.P.); Fax: +39-(0)39-2332167 (E.D. & C.P.)
| | | | | |
Collapse
|
14
|
Lu P, Ma Y, Wei S, Liang X. The dual role of complement in cancers, from destroying tumors to promoting tumor development. Cytokine 2021; 143:155522. [PMID: 33849765 DOI: 10.1016/j.cyto.2021.155522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Complement is an important branch of innate immunity; however, its biological significance goes far beyond the scope of simple nonspecific defense and involves a variety of physiological functions, including the adaptive immune response. In this review, to unravel the complex relationship between complement and tumors, we reviewed the high diversity of complement components in cancer and the heterogeneity of their production and activation pathways. In the tumor microenvironment, complement plays a dual regulatory role in the occurrence and development of tumors, affecting the outcomes of the immune response. We explored the differential expression levels of various complement components in human cancers via the Oncomine database. The gene expression profiling interactive analysis (GEPIA) tool and Kaplan-Meier plotter (K-M plotter) confirmed the correlation between differentially expressed complement genes and tumor prognosis. The tumor immune estimation resource (TIMER) database was used to statistically analyze the effect of complement on tumor immune infiltration. Finally, with a view to the role of complement in regulating T cell metabolism, complement could be a potential target for immunotherapies. Targeting complement to regulate the antitumor immune response seems to have potential for future treatment strategies. However, there are still many complex problems, such as who will benefit from this therapy and how to select the right therapeutic target and determine the appropriate drug concentration. The solutions to these problems depend on a deeper understanding of complement generation, activation, and regulatory and control mechanisms.
Collapse
Affiliation(s)
- Ping Lu
- Department of Medical Oncology, Hubei Cancer Hospital, the Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China
| | - Yifei Ma
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, the Seventh Clinical School Affiliated with Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China
| | - Shaozhong Wei
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, the Seventh Clinical School Affiliated with Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China.
| | - Xinjun Liang
- Department of Medical Oncology, Hubei Cancer Hospital, the Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Colorectal Cancer Clinical Research Center of HuBei Province, Wuhan, China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, China.
| |
Collapse
|
15
|
Xiong J, Kuang X, Lu T, Yu K, Liu X, Zhang Z, Wang W, Zhao L, Fang Q, Wu D, Wang J. C3a and C5a facilitates the metastasis of myeloma cells by activating Nrf2. Cancer Gene Ther 2021; 28:265-278. [PMID: 32873871 DOI: 10.1038/s41417-020-00217-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 11/09/2022]
Abstract
Multiple myeloma (MM) is still an incurable hematological malignancy, with even poorer prognosis in MM patients with distant invasion. The present study was designed to explore the effects of C3a and C5a on the migration, invasion, and adhesion of MM tumor cells and to investigate the underlying mechanisms. As a result, the levels of C3a and C5a in plasma of MM patients were significantly higher than those of healthy donors. Consistently, the expression of C3a and C5a receptors on myeloma cells of MM patients was also significantly higher than that on sorted plasma cells of normal donors. C3a and C5a have been confirmed to increase the migration, invasion and adhesion of MM cell lines by activating the MEK/ERK pathway and increasing the nuclear transfer of Nrf2 in vitro. Moreover, the MM cell line U266 with Nrf2 downregulation was incubated with C3a and C5a, followed by injection into the tail vein of NOD-SCID mice. We found that Nrf2 downregulation attenuated the migration of anaphylatoxin C3a and C5a to MM tumor cells in bone marrow, liver and lung in vivo. In conclusion, our results indicate that activation of the complement cascade in MM patients may contribute to the migration, invasion and adhesion of MM cells, and this type of tumor cells dissemination in MM is, at least partially, regulated by Nrf2. Thereby, complement suppression or Nrf2 downregulation might offer a novel therapeutic opportunity for MM.
Collapse
Affiliation(s)
- Jie Xiong
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis under Ministry of Health, Collaborative Innovation Center of Hematology, Suzhou Institute of Blood and Marrow Transplantation, 188 Shizi Street, 215006, Suzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Xingyi Kuang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Tingting Lu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Kunlin Yu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Xu Liu
- Department of Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, China
| | - Zhaoyuan Zhang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Weili Wang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Lu Zhao
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, China
| | - Depei Wu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis under Ministry of Health, Collaborative Innovation Center of Hematology, Suzhou Institute of Blood and Marrow Transplantation, 188 Shizi Street, 215006, Suzhou, Jiangsu, China.
| | - Jishi Wang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University. Hematopoietic Stem Cell Transplantation Center of Guizhou Province, Key Laboratory of Hematological Disease Diagnostic & Treat Centre of Guizhou Province. Guizhou Medical University, 550001, Guiyang, China.
| |
Collapse
|
16
|
O’Brien RM, Cannon A, Reynolds JV, Lysaght J, Lynam-Lennon N. Complement in Tumourigenesis and the Response to Cancer Therapy. Cancers (Basel) 2021; 13:1209. [PMID: 33802004 PMCID: PMC7998562 DOI: 10.3390/cancers13061209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
In recent years, our knowledge of the complement system beyond innate immunity has progressed significantly. A modern understanding is that the complement system has a multifaceted role in malignancy, impacting carcinogenesis, the acquisition of a metastatic phenotype and response to therapies. The ability of local immune cells to produce and respond to complement components has provided valuable insights into their regulation, and the subsequent remodeling of the tumour microenvironment. These novel discoveries have advanced our understanding of the immunosuppressive mechanisms supporting tumour growth and uncovered potential therapeutic targets. This review discusses the current understanding of complement in cancer, outlining both direct and immune cell-mediated roles. The role of complement in response to therapies such as chemotherapy, radiation and immunotherapy is also presented. While complement activities are largely context and cancer type-dependent, it is evident that promising therapeutic avenues have been identified, in particular in combination therapies.
Collapse
Affiliation(s)
- Rebecca M. O’Brien
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Aoife Cannon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - John V. Reynolds
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - Joanne Lysaght
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| |
Collapse
|
17
|
Jiang Y, Xu Z, Ma N, Yin L, Hao C, Li J. Effects of signaling pathway inhibitors on hematopoietic stem cells. Mol Med Rep 2020; 23:9. [PMID: 33179097 PMCID: PMC7687261 DOI: 10.3892/mmr.2020.11647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
While there are numerous small molecule inhibitory drugs available for a wide range of signalling pathways, at present, they are generally not used in combination in clinical settings. Previous reports have reported that the effects of glycogen synthase kinase (GSK)3β, p38MAPK, mTOR and histone deacetylase signaling combined together to suppress the stem-like nature of hematopoietic stem cells (HSCs), driving these cells to differentiate, cease proliferating and thereby impairing normal hematopoietic functionality. The present study aimed to determine the effect of HDACs, mTOR, GSK-3β and p38MAPK inhibitor combinations on the efficient expansion of HSCs using flow cytometry. Moreover, it specifically aimed to determine how inhibitors of the GSK3β signaling pathway, in combination with inhibitors of P38MAPK and mTOR signaling or histone deacetylase (HDAC) inhibitors, could affect HSC expansion, with the goal of identifying novel combination strategies useful for the expansion of HSCs. The results indicated that p38MAPK and/or GSK3β inhibitors increased Lin− cell and Lin−Sca-1+c-kit+ (LSK) cell numbers in vitro. Taken together, these results suggested that a combination of p38MAPK and GSK3β signaling may regulate HSC differentiation in vitro. These findings further indicated that the suppression of p38MAPK and/or GSK3β signalling may modulate HSC differentiation and self-renewal to enhance HSC expansion.
Collapse
Affiliation(s)
- Yuyu Jiang
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Zhaofeng Xu
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Na Ma
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Lizhi Yin
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Caiqin Hao
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Jing Li
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| |
Collapse
|
18
|
Bujko K, Cymer M, Adamiak M, Ratajczak MZ. An Overview of Novel Unconventional Mechanisms of Hematopoietic Development and Regulators of Hematopoiesis - a Roadmap for Future Investigations. Stem Cell Rev Rep 2020; 15:785-794. [PMID: 31642043 PMCID: PMC6925068 DOI: 10.1007/s12015-019-09920-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem cells (HSCs) are the best-characterized stem cells in adult tissues. Nevertheless, as of today, many open questions remain. First, what is the phenotype of the most primitive "pre-HSC" able to undergo asymmetric divisions during ex vivo expansion that gives rise to HSC for all hemato-lymphopoietic lineages. Next, most routine in vitro assays designed to study HSC specification into hematopoietic progenitor cells (HPCs) for major hematopoietic lineages are based on a limited number of peptide-based growth factors and cytokines, neglecting the involvement of several other regulators that are endowed with hematopoietic activity. Examples include many hormones, such as pituitary gonadotropins, gonadal sex hormones, IGF-1, and thyroid hormones, as well as bioactive phosphosphingolipids and extracellular nucleotides (EXNs). Moreover, in addition to regulation by stromal-derived factor 1 (SDF-1), trafficking of these cells during mobilization or homing after transplantation is also regulated by bioactive phosphosphingolipids, EXNs, and three ancient proteolytic cascades, the complement cascade (ComC), the coagulation cascade (CoA), and the fibrinolytic cascade (FibC). Finally, it has emerged that bone marrow responds by "sterile inflammation" to signals sent from damaged organs and tissues, systemic stress, strenuous exercise, gut microbiota, and the administration of certain drugs. This review will address the involvement of these unconventional regulators and present a broader picture of hematopoiesis.
Collapse
Affiliation(s)
- Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Monika Cymer
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Adamiak
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA. .,Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
19
|
Abdelbaset-Ismail A, Cymer M, Borkowska-Rzeszotek S, Brzeźniakiewicz-Janus K, Rameshwar P, Kakar SS, Ratajczak J, Ratajczak MZ. Bioactive Phospholipids Enhance Migration and Adhesion of Human Leukemic Cells by Inhibiting Heme Oxygenase 1 (HO-1) and Inducible Nitric Oxygenase Synthase (iNOS) in a p38 MAPK-Dependent Manner. Stem Cell Rev Rep 2020; 15:139-154. [PMID: 30302660 PMCID: PMC6366663 DOI: 10.1007/s12015-018-9853-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioactive phospholipids, including sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), lysophosphatidylcholine (LPC), and its derivative lysophosphatidic acid (LPA), have emerged as important mediators regulating the trafficking of normal and cancer cells. While the role of S1P in regulating migration of hematopoietic cells is well established, in this work we compared its biological effects to the effects of C1P, LPC, and LPA. We employed 10 human myeloid and lymphoid cell lines as well as blasts from AML patients. We observed that human leukemic cells express functional receptors for phospholipids and respond to stimulation by phosphorylation of p42/44 MAPK and AKT. We also found that bioactive phospholipids enhanced cell migration and adhesion of leukemic cells by downregulating expression of HO-1 and iNOS in a p38 MAPK-dependent manner but did not affect cell proliferation. By contrast, downregulation of p38 MAPK by SB203580 enhanced expression of HO-1 and iNOS and decreased migration of leukemic cells in vitro and their seeding efficiency to vital organs in vivo after injection into immunodeficient mice. Based on these findings, we demonstrate that, besides S1P, human leukemic cells also respond to C1P, LPC, and LPA. Since the prometastatic effects of bioactive phospholipids in vivo were mediated, at least in part, by downregulating HO-1 and iNOS expression in a p38 MAPK-dependent manner, we propose that inhibitors of p38 MAPK or stimulators of HO-1 activity will find application in inhibiting the spread of leukemic cells in response to bioactive phospholipids.
Collapse
Affiliation(s)
- Ahmed Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA.,Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Monika Cymer
- Center for Preclinical Research and Technology, Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | | | | | | | - Sham S Kakar
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA. .,Center for Preclinical Research and Technology, Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
20
|
Innate immunity orchestrates the mobilization and homing of hematopoietic stem/progenitor cells by engaging purinergic signaling-an update. Purinergic Signal 2020; 16:153-166. [PMID: 32415576 DOI: 10.1007/s11302-020-09698-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Bone marrow (BM) as an active hematopoietic organ is highly sensitive to changes in body microenvironments and responds to external physical stimuli from the surrounding environment. In particular, BM tissue responds to several cues related to infections, strenuous exercise, tissue/organ damage, circadian rhythms, and physical challenges such as irradiation. These multiple stimuli affect BM cells to a large degree through a coordinated response of the innate immunity network as an important guardian for maintaining homeostasis of the body. In this review, we will foc++us on the role of purinergic signaling and innate immunity in the trafficking of hematopoietic stem/progenitor cells (HSPCs) during their egression from the BM into peripheral blood (PB), as seen along pharmacological mobilization, and in the process of homing and subsequent engraftment into BM after hematopoietic transplantation. Innate immunity mediates these processes by engaging, in addition to certain peptide-based factors, other important non-peptide mediators, including bioactive phosphosphingolipids and extracellular nucleotides, as the main topic of this review. Elucidation of these mechanisms will allow development of more efficient stem cell mobilization protocols to harvest the required number of HSPCs for transplantation and to accelerate hematopoietic reconstitution in transplanted patients.
Collapse
|
21
|
He Z, Zhang S, Ma D, Fang Q, Yang L, Shen S, Chen Y, Ren L, Wang J. HO-1 promotes resistance to an EZH2 inhibitor through the pRB-E2F pathway: correlation with the progression of myelodysplastic syndrome into acute myeloid leukemia. J Transl Med 2019; 17:366. [PMID: 31711520 PMCID: PMC6849246 DOI: 10.1186/s12967-019-2115-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Myelodysplastic syndrome (MDS) can progress to acute myeloid leukemia (AML), and conventional chemotherapy (decitabine) does not effectively inhibit tumor cells. Enhancer of zeste homologue 2 (EZH2) and Heme oxygenase-1 (HO-1) are two key factors in patients resistance and deterioration. Methods In total, 58 MDS patients were divided into four groups. We analyzed the difference in HO-1 and EZH2 expression among the groups by real-time PCR. After treatment with Hemin or Znpp IX, flow cytometry was used to detect apoptosis and assess the cell cycle distribution of tumor cells. Following injection of mice with very high-risk MDS cells, spleen and bone marrow samples were studied by immunohistochemistry (IHC) and hematoxylin and eosin (H&E) staining. MDS cells overexpressing EZH2 and HO-1 were analyzed by high-throughput sequencing. The effect of HO-1 on the pRB-E2F pathway was analyzed by Western blotting. The effects of decitabine on P15INK4B and TP53 in MDS cells after inhibiting HO-1 were detected by Western blotting. Results Real-time PCR results showed that EZH2 and HO-1 expression levels were higher in MDS patients than in normal donors. The levels of HO-1 and EZH2 were simultaneously increased in the high-risk and very high-risk groups. Linear correlation analysis and laser scanning confocal microscopy results indicated that EZH2 was related to HO-1. MDS cells that highly expressed EZH2 and HO-1 infiltrated the tissues of experimental mice. IHC results indicated that these phenomena were related to the pRB-E2F pathway. High-throughput sequencing indicated that the progression of MDS to AML was related to EZH2. Using the E2F inhibitor HLM006474 and the EZH2 inhibitor JQEZ5, we showed that HO-1 could regulate EZH2 expression. HO-1 could stimulate the transcription and activation of EZH2 through the pRB-E2F pathway in MDS patients during chemotherapy, which reduced TP53 and P15INK4B expression. Conclusions EZH2 was associated with HO-1 in high-risk and very high-risk MDS patients. HO-1 could influence MDS resistance and progression to AML.
Collapse
Affiliation(s)
- Zhengchang He
- Department of Hematology, Hematological Institute of Guizhou Province, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Siyu Zhang
- Department of Hematology, Hematological Institute of Guizhou Province, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Dan Ma
- Department of Hematology, Hematological Institute of Guizhou Province, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Qin Fang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Liping Yang
- Clinical Medicine Research Center of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Shaoxian Shen
- Intensive Care Unit, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Ying Chen
- Department of Hematology, Hematological Institute of Guizhou Province, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Lingli Ren
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Jishi Wang
- Department of Hematology, Hematological Institute of Guizhou Province, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China.
| |
Collapse
|
22
|
Chen Z, Zhang P, Xu Y, Yan J, Liu Z, Lau WB, Lau B, Li Y, Zhao X, Wei Y, Zhou S. Surgical stress and cancer progression: the twisted tango. Mol Cancer 2019; 18:132. [PMID: 31477121 PMCID: PMC6717988 DOI: 10.1186/s12943-019-1058-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Surgical resection is an important avenue for cancer treatment, which, in most cases, can effectively alleviate the patient symptoms. However, accumulating evidence has documented that surgical resection potentially enhances metastatic seeding of tumor cells. In this review, we revisit the literature on surgical stress, and outline the mechanisms by which surgical stress, including ischemia/reperfusion injury, activation of sympathetic nervous system, inflammation, systemically hypercoagulable state, immune suppression and effects of anesthetic agents, promotes tumor metastasis. We also propose preventive strategies or resolution of tumor metastasis caused by surgical stress.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Peidong Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Ya Xu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China.,Deyang People's Hospital, Deyang, Sichuan, People's Republic of China
| | - Jiahui Yan
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Zixuan Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University Hospital, Philadelphia, USA
| | - Bonnie Lau
- Department of Surgery, Emergency Medicine, Kaiser Santa Clara Medical Center, Affiliate of Stanford University, Stanford, USA
| | - Ying Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, People's Republic of China
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Yuquan Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
23
|
Zhou Z, Ma D, Liu P, Wang P, Wei D, Yu K, Li P, Fang Q, Wang J. Deletion of HO-1 blocks development of B lymphocytes in mice. Cell Signal 2019; 63:109378. [PMID: 31369826 DOI: 10.1016/j.cellsig.2019.109378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/29/2022]
Abstract
B lymphocytes, a key cluster of cells composing the immune system, can protect against abnormal biological factors. Heme oxygenase-1 (HO-1) plays important roles in cell proliferation and immune regulation, but its effects on the development and growth of B lymphocytes are still unknown. Herein, the count of B lymphocytes in HO-1 gene knockout (HO-1+/-) mice was significantly lower than that of the HO-1 gene wild-type (HO-1WT) mice. Meanwhile, the cell count of HO-1+/- mice did not recover after irradiation for one week, due to the G0/G1 phase arrest of Pro-B cells and the augmented apoptosis of Pre-B cells. Up-regulation of HO-1 by lentivirus attenuated the Pro-B cell cycle arrest and Pre-B cell apoptosis. To understand the molecular mechanism by which HO-1 knockout blocked B lymphocyte development, protein-to-protein interaction network and Western blot were used. The PI3K/AKT signaling pathway mediated the regulatory effects of HO-1 on B lymphocytes. In conclusion, HO-1 is a crucial transcriptional repressor for B cell development.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Danna Wei
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Kunling Yu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Peifan Li
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China.
| |
Collapse
|
24
|
Lenkiewicz A, Bujko K, Brzezniakiewicz-Janus K, Xu B, Ratajczak MZ. The Complement Cascade as a Mediator of Human Malignant Hematopoietic Cell Trafficking. Front Immunol 2019; 10:1292. [PMID: 31231394 PMCID: PMC6567995 DOI: 10.3389/fimmu.2019.01292] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/21/2019] [Indexed: 12/13/2022] Open
Abstract
The complement cascade (ComC) cleavage fragments C3a and C5a regulate the trafficking of normal, differentiated hematopoietic cells, although they do not chemoattract more primitive hematopoietic stem/progenitor cells (HSPCs). By contrast, human myeloid and lymphoid leukemia cell lines and clonogenic blasts from chronic myelogenous leukemia (CML) and acute myelogenous leukemia (AML) patients respond to C3 and C5 cleavage fragments by chemotaxis and increased adhesion. Consistent with this finding, C3a and C5a receptors are expressed by leukemic cells at the mRNA (RT-PCR) and protein (FACS) levels, and these cells respond to C3a and C5a stimulation by phosphorylation of p44/42 MAPK and AKT. However, neither of these ComC cleavage fragments have an effect on cell proliferation or survival. In parallel, we found that inducible heme oxygenase 1 (HO-1)-an anti-inflammatory enzyme, is a negative regulator of ComC-mediated trafficking of malignant cells and that stimulation of these cells by C3 or C5 cleavage fragments downregulates HO-1 expression in a p38 MAPK-dependent manner, rendering cells exposed to C3a or C5a more mobile. We propose that, while the ComC is not directly involved in the proliferation of malignant hematopoietic cells, its activation in leukemia/lymphoma patients (e.g., as a result of accompanying infections or sterile inflammation after radio-chemotherapy) enhances the motility of malignant cells and contributes to their dissemination in a p38 MAPK-HO-1 axis-dependent manner. Based on this idea, we propose that inhibition of p38 MAPK or upregulation of HO-1 by available small-molecule modulators would have a beneficial effect on ameliorating expansion and dissemination of leukemia/lymphoma cells in clinical situations in which the ComC becomes activated. Finally, since we detected expression of C3 and C5 mRNA in human leukemic cell lines, further study of the potential role of the complosome in regulating the behavior of these cells is needed.
Collapse
Affiliation(s)
- Anna Lenkiewicz
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | | | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology of Xiamen University, Xiamen, China
| | - Mariusz Z Ratajczak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.,Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
25
|
Pio R, Ajona D, Ortiz-Espinosa S, Mantovani A, Lambris JD. Complementing the Cancer-Immunity Cycle. Front Immunol 2019; 10:774. [PMID: 31031765 PMCID: PMC6473060 DOI: 10.3389/fimmu.2019.00774] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Reactivation of cytotoxic CD8+ T-cell responses has set a new direction for cancer immunotherapy. Neutralizing antibodies targeting immune checkpoint programmed cell death protein 1 (PD-1) or its ligand (PD-L1) have been particularly successful for tumor types with limited therapeutic options such as melanoma and lung cancer. However, reactivation of T cells is only one step toward tumor elimination, and a substantial fraction of patients fails to respond to these therapies. In this context, combination therapies targeting more than one of the steps of the cancer-immune cycle may provide significant benefits. To find the best combinations, it is of upmost importance to understand the interplay between cancer cells and all the components of the immune response. This review focuses on the elements of the complement system that come into play in the cancer-immunity cycle. The complement system, an essential part of innate immunity, has emerged as a major regulator of cancer immunity. Complement effectors such as C1q, anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1, have been associated with tolerogenic cell death and inhibition of antitumor T-cell responses through the recruitment and/or activation of immunosuppressive cell subpopulations such as myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), or M2 tumor-associated macrophages (TAMs). Evidence is provided to support the idea that complement blocks many of the effector routes associated with the cancer-immunity cycle, providing the rationale for new therapeutic combinations aimed to enhance the antitumor efficacy of anti-PD-1/PD-L1 checkpoint inhibitors.
Collapse
Affiliation(s)
- Ruben Pio
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Daniel Ajona
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sergio Ortiz-Espinosa
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Ajona D, Ortiz-Espinosa S, Pio R, Lecanda F. Complement in Metastasis: A Comp in the Camp. Front Immunol 2019; 10:669. [PMID: 31001273 PMCID: PMC6457318 DOI: 10.3389/fimmu.2019.00669] [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: 01/10/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
The complement system represents a pillar of the innate immune response. This system, critical for host defense against pathogens, encompasses more than 50 soluble, and membrane-bound proteins. Emerging evidence underscores its clinical relevance in tumor progression and its role in metastasis, one of the hallmarks of cancer. The multistep process of metastasis entails the acquisition of advantageous functions required for the formation of secondary tumors. Thus, targeting components of the complement system could impact not only on tumor initiation but also on several crucial steps along tumor dissemination. This novel vulnerability could be concomitantly exploited with current strategies overcoming tumor-mediated immunosuppression to provide a substantial clinical benefit in the treatment of metastatic disease. In this review, we offer a tour d'horizon on recent advances in this area and their prospective potential for cancer treatment.
Collapse
Affiliation(s)
- Daniel Ajona
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Fernando Lecanda
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
27
|
Karasu E, Nilsson B, Köhl J, Lambris JD, Huber-Lang M. Targeting Complement Pathways in Polytrauma- and Sepsis-Induced Multiple-Organ Dysfunction. Front Immunol 2019; 10:543. [PMID: 30949180 PMCID: PMC6437067 DOI: 10.3389/fimmu.2019.00543] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022] Open
Abstract
Exposure to traumatic or infectious insults results in a rapid activation of the complement cascade as major fluid defense system of innate immunity. The complement system acts as a master alarm system during the molecular danger response after trauma and significantly contributes to the clearance of DAMPs and PAMPs. However, depending on the origin and extent of the damaged macro- and micro -milieu, the complement system can also be either excessively activated or inhibited. In both cases, this can lead to a maladaptive immune response and subsequent multiple cellular and organ dysfunction. The arsenal of complement-specific drugs offers promising strategies for various critical conditions after trauma, hemorrhagic shock, sepsis, and multiple organ failure. The imbalanced immune response needs to be detected in a rational and real-time manner before the translational therapeutic potential of these drugs can be fully utilized. Overall, the temporal-spatial complement response after tissue trauma and during sepsis remains somewhat enigmatic and demands a clinical triad: reliable tissue damage assessment, complement activation monitoring, and potent complement targeting to highly specific rebalance the fluid phase innate immune response.
Collapse
Affiliation(s)
- Ebru Karasu
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Laboratory C5:3, Uppsala University, Uppsala, Sweden
| | - Jörg Köhl
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - John D Lambris
- Department of Pathology & Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
28
|
NLRP3 inflammasome couples purinergic signaling with activation of the complement cascade for the optimal release of cells from bone marrow. Leukemia 2019; 33:815-825. [PMID: 30846866 PMCID: PMC6477784 DOI: 10.1038/s41375-019-0436-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
The mechanisms that regulate egress of hematopoietic stem/progenitor cells (HSPCs) into peripheral blood (PB) in response to stress, inflammation, tissue/organ injury, or administration of mobilization-inducing drugs are still not well understood, and because of the importance of stem cell trafficking in maintaining organism homeostasis, several complementary pathways are believed to be involved. Our group proposes that mobilization of HSPCs is mainly a result of sterile inflammation in the bone marrow (BM) microenvironment in response to pro-mobilizing stimuli and that during the initiation phase of the mobilization process BM-residing cells belonging to the innate immunity system, including granulocytes and monocytes, release danger-associated molecular pattern molecules (DAMPs, also known as alarmins), reactive oxygen species (ROS), as well as proteolytic and lipolytic enzymes. These factors together orchestrate the release of HSPCs into PB. One of the most important DAMPs released in the initiation phase of mobilization is extracellular adenosine triphosphate, a potent activator of the inflammasome. As a result of its activation, IL-1β and IL-18 as well as other pro-mobilizing mediators, including DAMPs such as high molecular group box 1 (Hmgb1) and S100 calcium-binding protein A9 (S100a9), are released. These DAMPs are important activators of the complement cascade (ComC) in the mannan-binding lectin (MBL)-dependent pathway. Specifically, Hmgb1 and S100a9 bind to MBL, which leads to activation of MBL-associated proteases, which activate the ComC and in parallel also trigger activation of the coagulation cascade (CoaC). In this review, we will highlight the novel role of the innate immunity cell-expressed NLRP3 inflammasome, which, during the initiation phase of HSPC mobilization, couples purinergic signaling with the MBL-dependent pathway of the ComC and, in parallel, the CoaC for optimal release of HSPCs. These data are important to optimize the pharmacological mobilization of HSPCs.
Collapse
|
29
|
Gruszka AM, Valli D, Restelli C, Alcalay M. Adhesion Deregulation in Acute Myeloid Leukaemia. Cells 2019; 8:E66. [PMID: 30658474 PMCID: PMC6356639 DOI: 10.3390/cells8010066] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion is a process through which cells interact with and attach to neighboring cells or matrix using specialized surface cell adhesion molecules (AMs). Adhesion plays an important role in normal haematopoiesis and in acute myeloid leukaemia (AML). AML blasts express many of the AMs identified on normal haematopoietic precursors. Differential expression of AMs between normal haematopoietic cells and leukaemic blasts has been documented to a variable extent, likely reflecting the heterogeneity of the disease. AMs govern a variety of processes within the bone marrow (BM), such as migration, homing, and quiescence. AML blasts home to the BM, as the AM-mediated interaction with the niche protects them from chemotherapeutic agents. On the contrary, they detach from the niches and move from the BM into the peripheral blood to colonize other sites, i.e., the spleen and liver, possibly in a process that is reminiscent of epithelial-to-mesenchymal-transition in metastatic solid cancers. The expression of AMs has a prognostic impact and there are ongoing efforts to therapeutically target adhesion in the fight against leukaemia.
Collapse
Affiliation(s)
- Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Cecilia Restelli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy.
| |
Collapse
|
30
|
Zhang R, Liu Q, Li T, Liao Q, Zhao Y. Role of the complement system in the tumor microenvironment. Cancer Cell Int 2019; 19:300. [PMID: 31787848 PMCID: PMC6858723 DOI: 10.1186/s12935-019-1027-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
The complement system has traditionally been considered a component of innate immunity against invading pathogens and "nonself" cells. Recent studies have demonstrated the immunoregulatory functions of complement activation in the tumor microenvironment (TME). The TME plays crucial roles in tumorigenesis, progression, metastasis and recurrence. Imbalanced complement activation and the deposition of complement proteins have been demonstrated in many types of tumors. Plasma proteins, receptors, and regulators of complement activation regulate several biological functions of stromal cells in the TME and promote the malignant biological properties of tumors. Interactions between the complement system and cancer cells contribute to the proliferation, epithelial-mesenchymal transition, migration and invasion of tumor cells. In this review, we summarize recent advances related to the function of the complement system in the TME and discuss the therapeutic potential of targeting complement-mediated immunoregulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Qiaofei Liu
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Tong Li
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Quan Liao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Yupei Zhao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| |
Collapse
|
31
|
May O, Merle NS, Grunenwald A, Gnemmi V, Leon J, Payet C, Robe-Rybkine T, Paule R, Delguste F, Satchell SC, Mathieson PW, Hazzan M, Boulanger E, Dimitrov JD, Fremeaux-Bacchi V, Frimat M, Roumenina LT. Heme Drives Susceptibility of Glomerular Endothelium to Complement Overactivation Due to Inefficient Upregulation of Heme Oxygenase-1. Front Immunol 2018; 9:3008. [PMID: 30619356 PMCID: PMC6306430 DOI: 10.3389/fimmu.2018.03008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/05/2018] [Indexed: 11/27/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a severe disease characterized by microvascular endothelial cell (EC) lesions leading to thrombi formation, mechanical hemolysis and organ failure, predominantly renal. Complement system overactivation is a hallmark of aHUS. To investigate this selective susceptibility of the microvascular renal endothelium to complement attack and thrombotic microangiopathic lesions, we compared complement and cyto-protection markers on EC, from different vascular beds, in in vitro and in vivo models as well as in patients. No difference was observed for complement deposits or expression of complement and coagulation regulators between macrovascular and microvascular EC, either at resting state or after inflammatory challenge. After prolonged exposure to hemolysis-derived heme, higher C3 deposits were found on glomerular EC, in vitro and in vivo, compared with other EC in culture and in mice organs (liver, skin, brain, lungs and heart). This could be explained by a reduced complement regulation capacity due to weaker binding of Factor H and inefficient upregulation of thrombomodulin (TM). Microvascular EC also failed to upregulate the cytoprotective heme-degrading enzyme heme-oxygenase 1 (HO-1), normally induced by hemolysis products. Only HUVEC (Human Umbilical Vein EC) developed adaptation to heme, which was lost after inhibition of HO-1 activity. Interestingly, the expression of KLF2 and KLF4—known transcription factors of TM, also described as possible transcription modulators of HO-1- was weaker in micro than macrovascular EC under hemolytic conditions. Our results show that the microvascular EC, and especially glomerular EC, fail to adapt to the stress imposed by hemolysis and acquire a pro-coagulant and complement-activating phenotype. Together, these findings indicate that the vulnerability of glomerular EC to hemolysis is a key factor in aHUS, amplifying complement overactivation and thrombotic microangiopathic lesions.
Collapse
Affiliation(s)
- Olivia May
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | - Nicolas S Merle
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Grunenwald
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France.,University of Lille, INSERM, CHU Lille, Department of Pathology, UMR-S 1172 - Jean-Pierre Aubert Research Center, Lille, France
| | - Viviane Gnemmi
- University of Lille, INSERM, CHU Lille, Department of Pathology, UMR-S 1172 - Jean-Pierre Aubert Research Center, Lille, France
| | - Juliette Leon
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cloé Payet
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Tania Robe-Rybkine
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Romain Paule
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | - Marc Hazzan
- INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | | | - Jordan D Dimitrov
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Veronique Fremeaux-Bacchi
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Marie Frimat
- INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | - Lubka T Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
32
|
Ratajczak MZ, Bujko K, Mack A, Kucia M, Ratajczak J. Cancer from the perspective of stem cells and misappropriated tissue regeneration mechanisms. Leukemia 2018; 32:2519-2526. [PMID: 30375490 PMCID: PMC6286324 DOI: 10.1038/s41375-018-0294-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022]
Abstract
Tumorigenesis can be considered as pathologically misappropriated tissue regeneration. In this review we will address some unresolved issues that support this concept. First, we will address the issue of the identity of cancer-initiating cells and the presence of cancer stem cells in growing tumors. We will also ask are there rare and distinct populations of cancer stem cells in established tumor cell lines, or are all of the cells cancer stem cells? Second, the most important clinical problem with cancer is its metastasis, and here a challenging question arises: by employing radio-chemotherapy for tumor treatment, do we unintentionally create a prometastatic microenvironment in collateral organs? Specifically, many factors upregulated in response to radio-chemotherapy-induced injury may attract highly migratory cancer cells that survived initial treatment. Third, what is the contribution of normal circulating stem cells to the growing malignancy? Do circulating normal stem cells recognize a tumor as a hypoxia-damaged tissue that needs vascular and stromal support and thereby contribute to tumor expansion? Fourth, is it reasonable to inhibit only one prometastatic ligand-receptor axis when cancer stem cells express several receptors for several chemotactic factors that may compensate for inhibition of the targeted receptor? Fifth, since most aggressive cancer cells mimic early-development stem cells, which properties of embryonic stem cells are retained in cancer cells? Would it be reasonable to inhibit cancer cell signaling pathways involved in the migration and proliferation of embryonic stem cells? We will also briefly address some new players in cancerogenesis, including extracellular microvesicles, bioactive phospholipids, and extracellular nucleotides.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute, Division of Hematology and Oncology, James Graham Brown Cancer Center, University Louisville, 500 South Floyd Street, Louisville, 40202, Kentucky, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| | - Kamila Bujko
- Stem Cell Institute, Division of Hematology and Oncology, James Graham Brown Cancer Center, University Louisville, 500 South Floyd Street, Louisville, 40202, Kentucky, USA
| | - Aaron Mack
- Stem Cell Institute, Division of Hematology and Oncology, James Graham Brown Cancer Center, University Louisville, 500 South Floyd Street, Louisville, 40202, Kentucky, USA
| | - Magda Kucia
- Stem Cell Institute, Division of Hematology and Oncology, James Graham Brown Cancer Center, University Louisville, 500 South Floyd Street, Louisville, 40202, Kentucky, USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute, Division of Hematology and Oncology, James Graham Brown Cancer Center, University Louisville, 500 South Floyd Street, Louisville, 40202, Kentucky, USA
| |
Collapse
|
33
|
Kochanek DM, Ghouse SM, Karbowniczek MM, Markiewski MM. Complementing Cancer Metastasis. Front Immunol 2018; 9:1629. [PMID: 30061895 PMCID: PMC6054933 DOI: 10.3389/fimmu.2018.01629] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Complement is an effector of innate immunity and a bridge connecting innate immunity and subsequent adaptive immune responses. It is essential for protection against infections and for orchestrating inflammatory responses. Recent studies have also demonstrated contribution of the complement system to several homeostatic processes that are traditionally not considered to be involved in immunity. Thus, complement regulates homeostasis and immunity. However, dysregulation of this system contributes to several pathologies including inflammatory and autoimmune diseases. Unexpectedly, studies of the last decade have also revealed that complement promotes cancer progression. Since the initial discovery of tumor promoting role of complement, numerous preclinical and clinical studies demonstrated contribution of several complement components to regulation of tumor growth through their direct interactions with the corresponding receptors on tumor cells or through suppression of antitumor immunity. Most of this work, however, focused on a role of complement in regulating growth of primary tumors. Only recently, a few studies showed that complement promotes cancer metastasis through its contribution to epithelial-to-mesenchymal transition and the premetastatic niche. This latter work has shown that complement activation and generation of complement effectors including C5a occur in organs that are target for metastasis prior to arrival of the very first tumor cells. C5a through its interactions with C5a receptor 1 inhibits antitumor immunity by activating and recruiting immunosuppressive cells from the bone marrow to the premetastatic niche and by regulating function and self-renewal of pulmonary tissue-resident alveolar macrophages. These new advancements provide additional evidence for multifaceted functions of complement in cancer.
Collapse
Affiliation(s)
- Dawn M Kochanek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Shanawaz M Ghouse
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Magdalena M Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| |
Collapse
|
34
|
Up-regulation of HO-1 promotes resistance of B-cell acute lymphocytic leukemia cells to HDAC4/5 inhibitor LMK-235 via the Smad7 pathway. Life Sci 2018; 207:386-394. [PMID: 29886060 DOI: 10.1016/j.lfs.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/22/2018] [Accepted: 06/04/2018] [Indexed: 02/03/2023]
Abstract
PURPOSE HDAC4/5 and Smad7 are potential therapeutic targets for the onset and progression of B-cell acute lymphocytic leukemia (B-ALL) and indices for clinical prognosis. In contrast, HO-1 (heat shock protein 32) plays a key role in protecting tumor cells from apoptosis. METHODS HDAC4/5, HO-1 and Smad7 expressions in 34 newly diagnosed B-ALL cases were detected by real-time PCR and Western blot. Lentivirus and small interference RNA were used to transfect B-ALL cells. The expression of Smad7 was detected after treatment with LMK-235 or Hemin and ZnPP. Apoptosis and proliferation were evaluated by flow cytometry, CCK-8 assay and Western blot. RESULTS HDAC4/5 was overexpressed in B-ALL patients with high HO-1 levels. Increasing the concentration of HDAC4/5 inhibitor LMK-235 induced the decrease of Smad7 and HO-1 expressions and the apoptosis of B-ALL cells by suppressing the phosphorylation of AKT (Protein kinase B). Up-regulating HO-1 alleviated the decrease of Smad7 expression and enhanced B-ALL resistance to LMK-235 by activating p-AKT which reduced the apoptosis of B-ALL cells and influenced the survival of leukemia patients. Silencing Smad7 also augmented the apoptosis rate of B-ALL cells by suppressing p-AKT. CONCLUSION HO-1 played a key role in protecting tumor cells from apoptosis, and HDAC4/5 were related with the apoptosis of B-ALL cells. LMK-235 may be able to improve the poor survival of leukemia patients.
Collapse
|
35
|
Ratajczak MZ, Adamiak M, Kucia M, Tse W, Ratajczak J, Wiktor-Jedrzejczak W. The Emerging Link Between the Complement Cascade and Purinergic Signaling in Stress Hematopoiesis. Front Immunol 2018; 9:1295. [PMID: 29922299 PMCID: PMC5996046 DOI: 10.3389/fimmu.2018.01295] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/24/2018] [Indexed: 01/08/2023] Open
Abstract
Innate immunity plays an important role in orchestrating the immune response, and the complement cascade (ComC) is a major component of this ancient defense system, which is activated by the classical-, alternative-, or mannan-binding lectin (MBL) pathways. However, the MBL-dependent ComC-activation pathway has been somewhat underappreciated for many years; recent evidence indicates that it plays a crucial role in regulating the trafficking of hematopoietic stem/progenitor cells (HSPCs) by promoting their egress from bone marrow (BM) into peripheral blood (PB). This process is initiated by the release of danger-associated molecular patterns (DAMPs) from BM cells, including the most abundant member of this family, adenosine triphosphate (ATP). This nucleotide is well known as a ubiquitous intracellular molecular energy source, but when secreted becomes an important extracellular nucleotide signaling molecule and mediator of purinergic signaling. What is important for the topic of this review, ATP released from BM cells is recognized as a DAMP by MBL, and the MBL-dependent pathway of ComC activation induces a state of "sterile inflammation" in the BM microenvironment. This activation of the ComC by MBL leads to the release of several potent mediators, including the anaphylatoxins C5a and desArgC5a, which are crucial for egress of HSPCs into the circulation. In parallel, as a ligand for purinergic receptors, ATP affects mobilization of HSPCs by activating other pro-mobilizing pathways. This emerging link between the release of ATP, which on the one hand is an activator of the MBL pathway of the ComC and on the other hand is a purinergic signaling molecule, will be discussed in this review. This mechanism plays an important role in triggering defense mechanisms in response to tissue/organ injury but may also have a negative impact by triggering autoimmune disorders, aging of HSPCs, induction of myelodysplasia, and graft-versus-host disease after transplantation of histoincompatible hematopoietic cells.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - William Tse
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | | |
Collapse
|
36
|
Bartaula-Brevik S, Lindstad Brattås MK, Tvedt THA, Reikvam H, Bruserud Ø. Splenic tyrosine kinase (SYK) inhibitors and their possible use in acute myeloid leukemia. Expert Opin Investig Drugs 2018; 27:377-387. [DOI: 10.1080/13543784.2018.1459562] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Sushma Bartaula-Brevik
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Tor Henrik Anderson Tvedt
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Håkon Reikvam
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
37
|
Ratajczak MZ, Adamiak M, Plonka M, Abdel-Latif A, Ratajczak J. Mobilization of hematopoietic stem cells as a result of innate immunity-mediated sterile inflammation in the bone marrow microenvironment-the involvement of extracellular nucleotides and purinergic signaling. Leukemia 2018; 32:1116-1123. [PMID: 29556022 PMCID: PMC5940655 DOI: 10.1038/s41375-018-0087-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/01/2018] [Accepted: 02/07/2018] [Indexed: 12/22/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) circulate in peripheral blood (PB) under normal conditions and their number increases in response to stress, inflammation, tissue/organ injury, and may increase up to 100-fold after administration of mobilization-inducing drugs. Mounting evidence suggests that mobilizing agent-induced mobilization of HSPCs from bone marrow into PB is a result of innate immunity-mediated sterile inflammation in the bone marrow (BM) microenvironment. A critical initiating role in this process is played by tissue/organ injury-mediated or pharmacologically induced release from bone marrow-residing granulocytes and monocytes of (i) danger-associated molecular patterns (DAMPs), (ii) reactive oxygen species (ROS), and (iii) proteolytic and lipolytic enzymes. All these factors together trigger activation of the complement and coagulation cascades, both of which orchestrate egress of HSPCs into BM sinusoids and lymphatics. Recent evidence also indicates that, in addition to attenuation of the SDF-1–CXCR4 and VLA-4–VCAM-1 retention axes in the BM microenvironment and the presence of a mobilization-directing phosphosphingolipid gradient in PB, an important role in the mobilization process is played by extracellular nucleotides and purinergic signaling. In particular, a new finding by our laboratory is that, while extracellular ATP promotes mobilization of HSPCs, its derivative, adenosine, has the opposite (inhibitory) effect.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA. .,Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| | - Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland
| | - Monika Plonka
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
38
|
Reis ES, Mastellos DC, Ricklin D, Mantovani A, Lambris JD. Complement in cancer: untangling an intricate relationship. Nat Rev Immunol 2018; 18:5-18. [PMID: 28920587 PMCID: PMC5816344 DOI: 10.1038/nri.2017.97] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In tumour immunology, complement has traditionally been considered as an adjunctive component that enhances the cytolytic effects of antibody-based immunotherapies, such as rituximab. Remarkably, research in the past decade has uncovered novel molecular mechanisms linking imbalanced complement activation in the tumour microenvironment with inflammation and suppression of antitumour immune responses. These findings have prompted new interest in manipulating the complement system for cancer therapy. This Review summarizes our current understanding of complement-mediated effector functions in the tumour microenvironment, focusing on how complement activation can act as a negative or positive regulator of tumorigenesis. It also offers insight into clinical aspects, including the feasibility of using complement biomarkers for cancer diagnosis and the use of complement inhibitors during cancer treatment.
Collapse
Affiliation(s)
- Edimara S Reis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| | | | - Daniel Ricklin
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Alberto Mantovani
- Humanitas Clinical and Research Center and Humanitas University, Rozzano-Milan 20089, Italy
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania 19104, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Ratajczak MZ, Pedziwiatr D, Cymer M, Kucia M, Kucharska-Mazur J, Samochowiec J. Sterile Inflammation of Brain, due to Activation of Innate Immunity, as a Culprit in Psychiatric Disorders. Front Psychiatry 2018; 9:60. [PMID: 29541038 PMCID: PMC5835766 DOI: 10.3389/fpsyt.2018.00060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Evidence has accumulated that the occurrence of psychiatric disorders is related to chronic inflammation. In support of this linkage, changes in the levels of circulating pro-inflammatory cytokines and chemokines in the peripheral blood (PB) of psychiatric patients as well as correlations between chronic inflammatory processes and psychiatric disorders have been described. Furthermore, an inflammatory process known as "sterile inflammation" when initiated directly in brain tissue may trigger the onset of psychoses. In this review, we will present the hypothesis that prolonged or chronic activation of the complement cascade (ComC) directly triggers inflammation in the brain and affects the proper function of this organ. Based on the current literature and our own work on mechanisms activating the ComC we hypothesize that inflammation in the brain is initiated by the mannan-binding lectin pathway of ComC activation. This activation is triggered by an increase in brain tissue of danger-associated molecular pattern (DAMP) mediators, including extracellular ATP and high-mobility group box 1 (HMGB1) protein, which are recognized by circulating pattern-recognition receptors, including mannan-binding lectin (MBL), that activate the ComC. On the other hand, this process is controlled by the anti-inflammatory action of heme oxygenase 1 (HO-1). In this review, we will try to connect changes in the release of DAMPs in the brain with inflammatory processes triggered by the innate immunity involving activation of the ComC as well as the inflammation-limiting effects of the anti-inflammatory HO-1 pathway. We will also discuss parallel observations that during ComC activation subsets of stem cells are mobilized into PB from bone marrow that are potentially involved in repair mechanisms.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Daniel Pedziwiatr
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Monika Cymer
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
40
|
Ajona D, Ortiz-Espinosa S, Pio R. Complement anaphylatoxins C3a and C5a: Emerging roles in cancer progression and treatment. Semin Cell Dev Biol 2017; 85:153-163. [PMID: 29155219 DOI: 10.1016/j.semcdb.2017.11.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/07/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Recent insights into the role of complement anaphylatoxins C3a and C5a in cancer provide new opportunities for the development of innovative biomarkers and therapeutic strategies. These two complement activation products can maintain chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and increase the motility and metastatic potential of cancer cells. Still, the diverse heterogeneity of responses mediated by these peptides poses a challenge both to our understanding of the role played by these molecules in cancer progression and to the development of effective treatments. This review attempts to summarize the evidence surrounding the involvement of anaphylatoxins in the biological contexts associated with tumor progression. We also describe the recent developments that support the inhibition of anaphylatoxins, or their cognate receptors C3aR and C5aR1, as a treatment option for maximizing the clinical efficacy of current immunotherapies that target the PD-1/PD-L1 immune checkpoint.
Collapse
Affiliation(s)
- Daniel Ajona
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Ruben Pio
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain.
| |
Collapse
|
41
|
Hajishengallis G, Reis ES, Mastellos DC, Ricklin D, Lambris JD. Novel mechanisms and functions of complement. Nat Immunol 2017; 18:1288-1298. [PMID: 29144501 PMCID: PMC5706779 DOI: 10.1038/ni.3858] [Citation(s) in RCA: 340] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/24/2017] [Indexed: 12/13/2022]
Abstract
Progress at the beginning of the 21st century transformed the perception of complement from that of a blood-based antimicrobial system to that of a global regulator of immunity and tissue homeostasis. More recent years have witnessed remarkable advances in structure-function insights and understanding of the mechanisms and locations of complement activation, which have added new layers of complexity to the biology of complement. This complexity is readily reflected by the multifaceted and contextual involvement of complement-driven networks in a wide range of inflammatory and neurodegenerative disorders and cancer. This Review provides an updated view of new and previously unanticipated functions of complement and how these affect immunity and disease pathogenesis.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dimitrios C Mastellos
- Division of Biodiagnostic Sciences and Technologies, INRASTES, National Center for Scientific Research 'Demokritos', Aghia Paraskevi, Athens, Greece
| | - Daniel Ricklin
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Adamiak M, Abdelbaset-Ismail A, Moore JB, Zhao J, Abdel-Latif A, Wysoczynski M, Ratajczak MZ. Inducible Nitric Oxide Synthase (iNOS) Is a Novel Negative Regulator of Hematopoietic Stem/Progenitor Cell Trafficking. Stem Cell Rev Rep 2017; 13:92-103. [PMID: 27752990 PMCID: PMC5346113 DOI: 10.1007/s12015-016-9693-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nitric oxide (NO) is a gaseous free radical molecule involved in several biological processes related to inflammation, tissue damage, and infections. Based on reports that NO inhibits migration of granulocytes and monocytes, we became interested in the role of inducible NO synthetase (iNOS) in pharmacological mobilization of hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood (PB). To address the role of NO in HSPC trafficking, we upregulated or downregulated iNOS expression in hematopoietic cell lines. Next, we performed mobilization studies in iNOS-/- mice and evaluated engraftment of iNOS-/- HSPCs in wild type (control) animals. Our results indicate that iNOS is a novel negative regulator of hematopoietic cell migration and prevents egress of HSPCs into PB during mobilization. At the molecular level, downregulation of iNOS resulted in downregulation of heme oxygenase 1 (HO-1), and, conversely, upregulation of iNOS enhanced HO-1 activity. Since HO-1 is a negative regulator of cell migration, the inhibitory effects of iNOS identified by us can be at least partially explained by its enhancing the HO-1 level in BM cells.
Collapse
Affiliation(s)
- Mateusz Adamiak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street Rm. 107, Louisville, KY, 40202, USA.,Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| | - Ahmed Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street Rm. 107, Louisville, KY, 40202, USA
| | - Joseph B Moore
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - J Zhao
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street Rm. 107, Louisville, KY, 40202, USA. .,Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
43
|
Ratajczak MZ. HO-1 inhibits migration of leukemic cells. Oncotarget 2017; 8:89429-89430. [PMID: 29163759 PMCID: PMC5685680 DOI: 10.18632/oncotarget.21044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/15/2017] [Indexed: 01/25/2023] Open
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, KY, USA and Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
44
|
Abstract
In addition to being a component of innate immunity and an ancient defense mechanism against invading pathogens, complement activation also participates in the adaptive immune response, inflammation, hemostasis, embryogenesis, and organ repair and development. Activation of the complement system via classical, lectin, or alternative pathways generates anaphylatoxins (C3a and C5a) and membrane attack complex (C5b-9) and opsonizes targeted cells. Complement activation end products and their receptors mediate cell-cell interactions that regulate several biological functions in the extravascular tissue. Signaling of anaphylatoxin receptors or assembly of membrane attack complex promotes cell dedifferentiation, proliferation, and migration in addition to reducing apoptosis. As a result, complement activation in the tumor microenvironment enhances tumor growth and increases metastasis. In this Review, I discuss immune and nonimmune functions of complement proteins and the tumor-promoting effect of complement activation.
Collapse
|